101
|
Personalized Dietary Advice to Increase Protein Intake in Older Adults Does Not Affect the Gut Microbiota, Appetite or Central Processing of Food Stimuli in Community-Dwelling Older Adults: A Six-Month Randomized Controlled Trial. Nutrients 2023; 15:nu15020332. [PMID: 36678203 PMCID: PMC9862486 DOI: 10.3390/nu15020332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Expert groups argue to raise the recommended daily allowance for protein in older adults from 0.8 to 1.2 g/kg/day to prevent undernutrition. However, protein is thought to increase satiety, possibly through effects on gut microbiota and central appetite regulation. If true, raising daily protein intake may work counterproductively. In a randomized controlled trial, we evaluated the effects of dietary advice aimed at increasing protein intake to 1.2 g/kg adjusted body weight/day (g/kg aBW/day) on appetite and gut microbiota in 90 community-dwelling older adults with habitual protein intake <1.0 g/kg aBW/day (Nintervention = 47, Ncontrol = 43). Food intake was determined by 24-h dietary recalls and gut microbiota by 16S rRNA sequencing. Functional magnetic resonance imaging (fMRI) scans were performed in a subgroup of 48 participants to evaluate central nervous system responses to food-related stimuli. Both groups had mean baseline protein intake of 0.8 ± 0.2 g/kg aBW/day. At 6 months’ follow-up this increased to 1.2 ± 0.2 g/kg aBW/day for the intervention group and 0.9 ± 0.2 g/kg aBW/day for the control group. Microbiota composition was not affected, nor were appetite or brain activity in response to food-related stimuli. Increasing protein intake in older adults to 1.2 g/kg aBW/day does not negatively impact the gut microbiota or suppress appetite.
Collapse
|
102
|
Ramamurthy E, Welch G, Cheng J, Yuan Y, Gunsalus L, Bennett DA, Tsai LH, Pfenning AR. Cell type-specific histone acetylation profiling of Alzheimer's disease subjects and integration with genetics. Front Mol Neurosci 2023; 15:948456. [PMID: 36683855 PMCID: PMC9853565 DOI: 10.3389/fnmol.2022.948456] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/11/2022] [Indexed: 01/07/2023] Open
Abstract
We profile genome-wide histone 3 lysine 27 acetylation (H3K27ac) of 3 major brain cell types from hippocampus and dorsolateral prefrontal cortex (dlPFC) of subjects with and without Alzheimer's Disease (AD). We confirm that single nucleotide polymorphisms (SNPs) associated with late onset AD (LOAD) show a strong tendency to reside in microglia-specific gene regulatory elements. Despite this significant colocalization, we find that microglia harbor more acetylation changes associated with age than with amyloid-β (Aβ) load. In contrast, we detect that an oligodendrocyte-enriched glial (OEG) population contains the majority of differentially acetylated peaks associated with Aβ load. These differential peaks reside near both early onset risk genes (APP, PSEN1, PSEN2) and late onset AD risk loci (including BIN1, PICALM, CLU, ADAM10, ADAMTS4, SORL1, FERMT2), Aβ processing genes (BACE1), as well as genes involved in myelinating and oligodendrocyte development processes. Interestingly, a number of LOAD risk loci associated with differentially acetylated risk genes contain H3K27ac peaks that are specifically enriched in OEG. These findings implicate oligodendrocyte gene regulation as a potential mechanism by which early onset and late onset risk genes mediate their effects, and highlight the deregulation of myelinating processes in AD. More broadly, our dataset serves as a resource for the study of functional effects of genetic variants and cell type specific gene regulation in AD.
Collapse
Affiliation(s)
- Easwaran Ramamurthy
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Gwyneth Welch
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Jemmie Cheng
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Yixin Yuan
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Laura Gunsalus
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, United States
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, United States
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Andreas R. Pfenning
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
103
|
Thompson RC, Simons NW, Wilkins L, Cheng E, Del Valle DM, Hoffman GE, Cervia C, Fennessy B, Mouskas K, Francoeur NJ, Johnson JS, Lepow L, Le Berichel J, Chang C, Beckmann AG, Wang YC, Nie K, Zaki N, Tuballes K, Barcessat V, Cedillo MA, Yuan D, Huckins L, Roussos P, Marron TU, Glicksberg BS, Nadkarni G, Heath JR, Gonzalez-Kozlova E, Boyman O, Kim-Schulze S, Sebra R, Merad M, Gnjatic S, Schadt EE, Charney AW, Beckmann ND. Molecular states during acute COVID-19 reveal distinct etiologies of long-term sequelae. Nat Med 2023; 29:236-246. [PMID: 36482101 PMCID: PMC9873574 DOI: 10.1038/s41591-022-02107-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/25/2022] [Indexed: 12/13/2022]
Abstract
Post-acute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are debilitating, clinically heterogeneous and of unknown molecular etiology. A transcriptome-wide investigation was performed in 165 acutely infected hospitalized individuals who were followed clinically into the post-acute period. Distinct gene expression signatures of post-acute sequelae were already present in whole blood during acute infection, with innate and adaptive immune cells implicated in different symptoms. Two clusters of sequelae exhibited divergent plasma-cell-associated gene expression patterns. In one cluster, sequelae associated with higher expression of immunoglobulin-related genes in an anti-spike antibody titer-dependent manner. In the other, sequelae associated independently of these titers with lower expression of immunoglobulin-related genes, indicating lower non-specific antibody production in individuals with these sequelae. This relationship between lower total immunoglobulins and sequelae was validated in an external cohort. Altogether, multiple etiologies of post-acute sequelae were already detectable during SARS-CoV-2 infection, directly linking these sequelae with the acute host response to the virus and providing early insights into their development.
Collapse
Affiliation(s)
- Ryan C Thompson
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Esther Cheng
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Marie Del Valle
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Department of Genetics and Genomic Sciences, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlo Cervia
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Brian Fennessy
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Konstantinos Mouskas
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Susan and Leonard Feinstein Inflammatory Bowel Disease Clinical Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nancy J Francoeur
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Lauren Lepow
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Le Berichel
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christie Chang
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ying-Chih Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicholas Zaki
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kevin Tuballes
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vanessa Barcessat
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mario A Cedillo
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan Yuan
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Laura Huckins
- Department of Genetics and Genomic Sciences, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Thomas U Marron
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin S Glicksberg
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish Nadkarni
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James R Heath
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Edgar Gonzalez-Kozlova
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Seunghee Kim-Schulze
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Sema4, a Mount Sinai venture, Stamford, CT, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Sema4, a Mount Sinai venture, Stamford, CT, USA
| | - Alexander W Charney
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Noam D Beckmann
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Medicine, Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
104
|
Gisby JS, Buang NB, Papadaki A, Clarke CL, Malik TH, Medjeral-Thomas N, Pinheiro D, Mortimer PM, Lewis S, Sandhu E, McAdoo SP, Prendecki MF, Willicombe M, Pickering MC, Botto M, Thomas DC, Peters JE. Multi-omics identify falling LRRC15 as a COVID-19 severity marker and persistent pro-thrombotic signals in convalescence. Nat Commun 2022; 13:7775. [PMID: 36522333 PMCID: PMC9753891 DOI: 10.1038/s41467-022-35454-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Patients with end-stage kidney disease (ESKD) are at high risk of severe COVID-19. Here, we perform longitudinal blood sampling of ESKD haemodialysis patients with COVID-19, collecting samples pre-infection, serially during infection, and after clinical recovery. Using plasma proteomics, and RNA-sequencing and flow cytometry of immune cells, we identify transcriptomic and proteomic signatures of COVID-19 severity, and find distinct temporal molecular profiles in patients with severe disease. Supervised learning reveals that the plasma proteome is a superior indicator of clinical severity than the PBMC transcriptome. We show that a decreasing trajectory of plasma LRRC15, a proposed co-receptor for SARS-CoV-2, is associated with a more severe clinical course. We observe that two months after the acute infection, patients still display dysregulated gene expression related to vascular, platelet and coagulation pathways, including PF4 (platelet factor 4), which may explain the prolonged thrombotic risk following COVID-19.
Collapse
Affiliation(s)
- Jack S Gisby
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - Norzawani B Buang
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - Artemis Papadaki
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - Candice L Clarke
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Talat H Malik
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - Nicholas Medjeral-Thomas
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Damiola Pinheiro
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - Paige M Mortimer
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - Shanice Lewis
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - Eleanor Sandhu
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Stephen P McAdoo
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Maria F Prendecki
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Michelle Willicombe
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Matthew C Pickering
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - Marina Botto
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK
| | - David C Thomas
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK.
- Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK.
| | - James E Peters
- Centre for Inflammatory Disease, Dept of Immunology and Inflammation, Imperial College London, London, UK.
| |
Collapse
|
105
|
Duffy FJ, Hertoghs N, Du Y, Neal ML, Oyong D, McDermott S, Minkah N, Carnes J, Schwedhelm KV, McElrath MJ, De Rosa SC, Newell E, Aitchison JD, Stuart K. Longitudinal immune profiling after radiation-attenuated sporozoite vaccination reveals coordinated immune processes correlated with malaria protection. Front Immunol 2022; 13:1042741. [PMID: 36591224 PMCID: PMC9798120 DOI: 10.3389/fimmu.2022.1042741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Background Identifying immune processes required for liver-stage sterilizing immunity to malaria remains an open problem. The IMRAS trial comprised 5x immunizations with radiation-attenuated sporozoites resulting in 55% protection from subsequent challenge. Methods To identify correlates of vaccination and protection, we performed detailed systems immunology longitudinal profiling of the entire trial time course including whole blood transcriptomics, detailed PBMC cell phenotyping and serum antigen array profiling of 11 IMRAS radiation-attenuated sporozoite (RAS) vaccinees at up to 21 timepoints each. Results RAS vaccination induced serum antibody responses to CSP, TRAP, and AMA1 in all vaccinees. We observed large numbers of differentially expressed genes associated with vaccination response and protection, with distinctly differing transcriptome responses elicited after each immunization. These included inflammatory and proliferative responses, as well as increased abundance of monocyte and DC subsets after each immunization. Increases in Vδ2 γδ; T cells and MAIT cells were observed in response to immunization over the course of study, and CD1c+ CD40+ DC abundance was significantly associated with protection. Interferon responses strongly differed between protected and non-protected individuals with high interferon responses after the 1st immunization, but not the 2nd-5th. Blood transcriptional interferon responses were correlated with abundances of different circulating classical and non-classical monocyte populations. Conclusions This study has revealed multiple coordinated immunological processes induced by vaccination and associated with protection. Our work represents the most detailed immunological profiling of a RAS vaccine trial performed to date and will guide the design and interpretation of future malaria vaccine trials.
Collapse
Affiliation(s)
- Fergal J. Duffy
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States,*Correspondence: Fergal J. Duffy, ; Ken Stuart,
| | - Nina Hertoghs
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Ying Du
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Maxwell L. Neal
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Damian Oyong
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Suzanne McDermott
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Nana Minkah
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Jason Carnes
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Katharine V. Schwedhelm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Evan Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - John D. Aitchison
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States
| | - Ken Stuart
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States,*Correspondence: Fergal J. Duffy, ; Ken Stuart,
| |
Collapse
|
106
|
Groth T, Diehl AD, Gunawan R, Neelamegham S. GlycoEnzOnto: a GlycoEnzyme pathway and molecular function ontology. Bioinformatics 2022; 38:5413-5420. [PMID: 36282863 PMCID: PMC9750110 DOI: 10.1093/bioinformatics/btac704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/22/2022] [Accepted: 10/24/2022] [Indexed: 12/25/2022] Open
Abstract
MOTIVATION The 'glycoEnzymes' include a set of proteins having related enzymatic, metabolic, transport, structural and cofactor functions. Currently, there is no established ontology to describe glycoEnzyme properties and to relate them to glycan biosynthesis pathways. RESULTS We present GlycoEnzOnto, an ontology describing 403 human glycoEnzymes curated along 139 glycosylation pathways, 134 molecular functions and 22 cellular compartments. The pathways described regulate nucleotide-sugar metabolism, glycosyl-substrate/donor transport, glycan biosynthesis and degradation. The role of each enzyme in the glycosylation initiation, elongation/branching and capping/termination phases is described. IUPAC linear strings present systematic human/machine-readable descriptions of individual reaction steps and enable automated knowledge-based curation of biochemical networks. All GlycoEnzOnto knowledge is integrated with the Gene Ontology biological processes. GlycoEnzOnto enables improved transcript overrepresentation analyses and glycosylation pathway identification compared to other available schema, e.g. KEGG and Reactome. Overall, GlycoEnzOnto represents a holistic glycoinformatics resource for systems-level analyses. AVAILABILITY AND IMPLEMENTATION https://github.com/neel-lab/GlycoEnzOnto. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Theodore Groth
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Alexander D Diehl
- Department of Biomedical Informatics, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Rudiyanto Gunawan
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Sriram Neelamegham
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
- Department of Medicine, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| |
Collapse
|
107
|
Park HJ, Kelly JM, Hoffman JR, Takaesu F, Schwartzman W, Ulziibayar A, Kitsuka T, Heuer E, Yimit A, Malbrue R, Anderson C, Morrison A, Naguib A, Mckee C, Harrison A, Boe B, Armstrong A, Salavitabar A, Yates A, Shinoka T, Carrillo S, Breuer CK, Davis ME. Computational analysis of serum-derived extracellular vesicle miRNAs in juvenile sheep model of single stage Fontan procedure. EXTRACELLULAR VESICLE 2022; 1:100013. [PMID: 36330420 PMCID: PMC9623551 DOI: 10.1016/j.vesic.2022.100013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Patients with single ventricle heart defects requires a series of staged open-heart procedures, termed Fontan palliation. However, while lifesaving, these operations are associated with significant morbidity and early mortality. The attendant complications are thought to arise in response to the abnormal hemodynamics induced by Fontan palliation, although the pathophysiology underlying these physicochemical changes in cardiovascular and other organs remain unknown. Here, we investigated the microRNA (miRNA) content in serum and serum-derived extracellular vesicles (EVs) by sequencing small RNAs from a physiologically relevant sheep model of the Fontan operation. The differential expression analysis identified the enriched miRNA clusters in (1) serum vs. serum-derived EVs and (2) pre-Fontan EVs vs. post-Fontan EVs. Metascape analysis showed that the overexpressed subset of EV miRNAs by Fontan procedure target liver-specific cells, underscoring a potentially important pathway involved in the liver dysfunction that occurs as a consequence of Fontan palliation. We also found that post-Fontan EV miRNAs were associated with senescence and cell death, whereas pre-Fontan EV miRNAs were associated with stem cell maintenance and epithelial-to-mesenchymal transition. This study shows great potential to identify novel circulating EV biomarkers from Fontan sheep serum that may be used for the diagnosis, prognosis, and therapeutics for patients that have undergone Fontan palliation.
Collapse
Affiliation(s)
- Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, USA
- Department of Molecular Science and Technology, Ajou University, Republic of Korea
| | - John M. Kelly
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jessica R. Hoffman
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, USA
- Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Felipe Takaesu
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Anudari Ulziibayar
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Takahiro Kitsuka
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Eric Heuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Asigul Yimit
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Raphael Malbrue
- The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | - Cole Anderson
- Biomedical Engineering Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Adrienne Morrison
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Aymen Naguib
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Christopher Mckee
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Andrew Harrison
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Brian Boe
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Aimee Armstrong
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Arash Salavitabar
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Andrew Yates
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Toshiharu Shinoka
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Sergio Carrillo
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Christopher K. Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, USA
- Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
- Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| |
Collapse
|
108
|
Vestal BE, Wynn E, Moore CM. lmerSeq: an R package for analyzing transformed RNA-Seq data with linear mixed effects models. BMC Bioinformatics 2022; 23:489. [DOI: 10.1186/s12859-022-05019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022] Open
Abstract
Abstract
Background
Studies that utilize RNA Sequencing (RNA-Seq) in conjunction with designs that introduce dependence between observations (e.g. longitudinal sampling) require specialized analysis tools to accommodate this additional complexity. This R package contains a set of utilities to fit linear mixed effects models to transformed RNA-Seq counts that properly account for this dependence when performing statistical analyses.
Results
In a simulation study comparing lmerSeq and two existing methodologies that also work with transformed RNA-Seq counts, we found that lmerSeq was comprehensively better in terms of nominal error rate control and statistical power.
Conclusions
Existing R packages for analyzing transformed RNA-Seq data with linear mixed models are limited in the variance structures they allow and/or the transformation methods they support. The lmerSeq package offers more flexibility in both of these areas and gave substantially better results in our simulations.
Collapse
|
109
|
Lamon S, Le Carré J, Petito G, Duong HP, Luthi F, Hiam D, Léger B. The effect of the menstrual cycle on the circulating microRNA pool in human plasma: a pilot study. Hum Reprod 2022; 38:46-56. [PMID: 36350564 PMCID: PMC9825267 DOI: 10.1093/humrep/deac243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/10/2022] [Indexed: 11/10/2022] Open
Abstract
STUDY QUESTION Do ovarian hormone changes influence the levels of cell-free or circulating microRNA (cf-miRNA) across the menstrual cycle? SUMMARY ANSWER This exploratory study suggests that fluctuations in hormonal levels throughout the menstrual cycle may alter cf-miRNAs levels. WHAT IS KNOWN ALREADY cf-miRNA levels vary with numerous pathological and physiological conditions in both males and females and are regulated by exogenous and endogenous factors, including hormones. STUDY DESIGN, SIZE, DURATION A prospective, monocentric study was conducted between March and November 2021. Since this was a pilot study, the sample size was based on feasibility as well as previous similar human studies conducted in different tissues. A total of 20 participants were recruited for the study. PARTICIPANTS/MATERIALS, SETTING, METHODS We conducted an exploratory study where blood samples were collected from 16 eumenorrheic females in the early follicular phase, the ovulation phase and the mid-luteal phase of the menstrual cycle. The levels of oestrogen, progesterone, LH and FSH were measured in serum by electrochemiluminescence. The levels of 174 plasma-enriched miRNAs were profiled using a PCR-based panel, including stringent internal and external controls to account for the potential differences in RNA extraction and reverse-transcription stemming from low-RNA input samples. MAIN RESULTS AND THE ROLE OF CHANCE This exploratory study suggests that cf-miRNAs may play an active role in the regulation of the female cycle by mediating the expression of genes during fluctuating hormonal changes. Linear mixed-models, adjusted for the relevant variables, showed associations between phases of the menstrual cycle, ovarian hormones and plasma cf-miRNA levels. Validated gene targets of the cf-miRNAs varying with the menstrual cycle were enriched within female reproductive tissues and are primarily involved in cell proliferation and apoptosis. LARGE SCALE DATA All relevant data are available from the Mendeley database: LEGER, Bertrand (2022), 'MiRNA and menstrual cycle', Mendeley Data, V1, doi: 10.17632/2br3zp79m3.1. LIMITATIONS, REASONS FOR CAUTION Our study was conducted on a small participant cohort. However, it was tightly controlled for endogenous and exogenous confounders, which is critical to ensure robust and reproducible cf-miRNA research. Both adjusted and non-adjusted P-values are presented throughout the article. WIDER IMPLICATIONS OF THE FINDINGS Measures of ovarian hormones should be rigorously included in future studies assessing cf-miRNA levels in females and used as time-varying confounders. Our results reinforce the importance of accounting for female-specific biological processes in physiology research by implementing practical or statistical mitigation strategies during data collection and analysis. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the Clinique romande de réadaptation, Sion, Switzerland. S.L. was supported by an Australian Research Council (ARC) Future Fellowship (FT10100278). D.H. was supported by an Executive Dean's Postdoctoral Research Fellowship from Deakin University. The authors declare no competing interests.
Collapse
Affiliation(s)
- Séverine Lamon
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Joane Le Carré
- Department of Medical Research, Clinique romande de réadaptation Suva, Sion, Switzerland
| | - Giuseppe Petito
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “L. Vanvitelli”, Caserta, Italy
| | - Hong Phuoc Duong
- Department of Medical Research, Clinique romande de réadaptation Suva, Sion, Switzerland
| | - François Luthi
- Department of Medical Research, Clinique romande de réadaptation Suva, Sion, Switzerland
| | - Danielle Hiam
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Bertrand Léger
- Correspondence address. Department of Medical Research, Clinique romande de réadaptation, Avenue GrandChampsec 90, CH-1951 Sion, Switzerland. E-mail:
| |
Collapse
|
110
|
Dong P, Hoffman GE, Apontes P, Bendl J, Rahman S, Fernando MB, Zeng B, Vicari JM, Zhang W, Girdhar K, Townsley KG, Misir R, Brennand KJ, Haroutunian V, Voloudakis G, Fullard JF, Roussos P. Population-level variation in enhancer expression identifies disease mechanisms in the human brain. Nat Genet 2022; 54:1493-1503. [PMID: 36163279 PMCID: PMC9547946 DOI: 10.1038/s41588-022-01170-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
Abstract
Identification of risk variants for neuropsychiatric diseases within enhancers underscores the importance of understanding population-level variation in enhancer function in the human brain. Besides regulating tissue-specific and cell-type-specific transcription of target genes, enhancers themselves can be transcribed. By jointly analyzing large-scale cell-type-specific transcriptome and regulome data, we cataloged 30,795 neuronal and 23,265 non-neuronal candidate transcribed enhancers. Examination of the transcriptome in 1,382 brain samples identified robust expression of transcribed enhancers. We explored gene-enhancer coordination and found that enhancer-linked genes are strongly implicated in neuropsychiatric disease. We identified expression quantitative trait loci (eQTLs) for both genes and enhancers and found that enhancer eQTLs mediate a substantial fraction of neuropsychiatric trait heritability. Inclusion of enhancer eQTLs in transcriptome-wide association studies enhanced functional interpretation of disease loci. Overall, our study characterizes the gene-enhancer regulome and genetic mechanisms in the human cortex in both healthy and diseased states.
Collapse
Affiliation(s)
- Pengfei Dong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pasha Apontes
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, New York, NY, USA
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Rahman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael B Fernando
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Biao Zeng
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James M Vicari
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wen Zhang
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kayla G Townsley
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruth Misir
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen J Brennand
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Vahram Haroutunian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, New York, NY, USA.
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| |
Collapse
|
111
|
Bendl J, Hauberg ME, Girdhar K, Im E, Vicari JM, Rahman S, Fernando MB, Townsley KG, Dong P, Misir R, Kleopoulos SP, Reach SM, Apontes P, Zeng B, Zhang W, Voloudakis G, Brennand KJ, Nixon RA, Haroutunian V, Hoffman GE, Fullard JF, Roussos P. The three-dimensional landscape of cortical chromatin accessibility in Alzheimer's disease. Nat Neurosci 2022; 25:1366-1378. [PMID: 36171428 PMCID: PMC9581463 DOI: 10.1038/s41593-022-01166-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/16/2022] [Indexed: 02/06/2023]
Abstract
To characterize the dysregulation of chromatin accessibility in Alzheimer's disease (AD), we generated 636 ATAC-seq libraries from neuronal and nonneuronal nuclei isolated from the superior temporal gyrus and entorhinal cortex of 153 AD cases and 56 controls. By analyzing a total of ~20 billion read pairs, we expanded the repertoire of known open chromatin regions (OCRs) in the human brain and identified cell-type-specific enhancer-promoter interactions. We show that interindividual variability in OCRs can be leveraged to identify cis-regulatory domains (CRDs) that capture the three-dimensional structure of the genome (3D genome). We identified AD-associated effects on chromatin accessibility, the 3D genome and transcription factor (TF) regulatory networks. For one of the most AD-perturbed TFs, USF2, we validated its regulatory effect on lysosomal genes. Overall, we applied a systematic approach to understanding the role of the 3D genome in AD. We provide all data as an online resource for widespread community-based analysis.
Collapse
Affiliation(s)
- Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mads E Hauberg
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative of Integrative Psychiatric Research (iPSYCH), Aarhus University, Aarhus, Denmark
- Centre for Integrative Sequencing (iSEQ), Aarhus University, Aarhus, Denmark
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eunju Im
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Psychiatry, New York University Langone Health, New York, NY, USA
| | - James M Vicari
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Rahman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael B Fernando
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kayla G Townsley
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pengfei Dong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruth Misir
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven P Kleopoulos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah M Reach
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pasha Apontes
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Biao Zeng
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wen Zhang
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen J Brennand
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Psychiatry, New York University Langone Health, New York, NY, USA
- Department of Cell Biology, New York University Langone Health, New York, NY, USA
- New York University Neuroscience Institute, New York, NY, USA
| | - Vahram Haroutunian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
112
|
Dinnon KH, Leist SR, Okuda K, Dang H, Fritch EJ, Gully KL, De la Cruz G, Evangelista MD, Asakura T, Gilmore RC, Hawkins P, Nakano S, West A, Schäfer A, Gralinski LE, Everman JL, Sajuthi SP, Zweigart MR, Dong S, McBride J, Cooley MR, Hines JB, Love MK, Groshong SD, VanSchoiack A, Phelan SJ, Liang Y, Hether T, Leon M, Zumwalt RE, Barton LM, Duval EJ, Mukhopadhyay S, Stroberg E, Borczuk A, Thorne LB, Sakthivel MK, Lee YZ, Hagood JS, Mock JR, Seibold MA, O’Neal WK, Montgomery SA, Boucher RC, Baric RS. SARS-CoV-2 infection produces chronic pulmonary epithelial and immune cell dysfunction with fibrosis in mice. Sci Transl Med 2022; 14:eabo5070. [PMID: 35857635 PMCID: PMC9273046 DOI: 10.1126/scitranslmed.abo5070] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/17/2022] [Indexed: 01/27/2023]
Abstract
A subset of individuals who recover from coronavirus disease 2019 (COVID-19) develop post-acute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (PASC), but the mechanistic basis of PASC-associated lung abnormalities suffers from a lack of longitudinal tissue samples. The mouse-adapted SARS-CoV-2 strain MA10 produces an acute respiratory distress syndrome in mice similar to humans. To investigate PASC pathogenesis, studies of MA10-infected mice were extended from acute to clinical recovery phases. At 15 to 120 days after virus clearance, pulmonary histologic findings included subpleural lesions composed of collagen, proliferative fibroblasts, and chronic inflammation, including tertiary lymphoid structures. Longitudinal spatial transcriptional profiling identified global reparative and fibrotic pathways dysregulated in diseased regions, similar to human COVID-19. Populations of alveolar intermediate cells, coupled with focal up-regulation of profibrotic markers, were identified in persistently diseased regions. Early intervention with antiviral EIDD-2801 reduced chronic disease, and early antifibrotic agent (nintedanib) intervention modified early disease severity. This murine model provides opportunities to identify pathways associated with persistent SARS-CoV-2 pulmonary disease and test countermeasures to ameliorate PASC.
Collapse
Affiliation(s)
- Kenneth H. Dinnon
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Sarah R. Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ethan J. Fritch
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kendra L. Gully
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Mia D. Evangelista
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Takanori Asakura
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Rodney C. Gilmore
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Padraig Hawkins
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Satoko Nakano
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ande West
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Lisa E. Gralinski
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jamie L. Everman
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado 80206, USA
| | - Satria P. Sajuthi
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado 80206, USA
| | - Mark R. Zweigart
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Stephanie Dong
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jennifer McBride
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Michelle R. Cooley
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jesse B. Hines
- Golden Point Scientific Laboratories, Hoover, Alabama 35216, USA
| | - Miriya K. Love
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Steve D. Groshong
- Division of Pathology, Department of Medicine, National Jewish Health, Denver, Colorado 80206, USA
| | | | | | - Yan Liang
- NanoString Technologies, Seattle, Washington 98109, USA
| | - Tyler Hether
- NanoString Technologies, Seattle, Washington 98109, USA
| | - Michael Leon
- NanoString Technologies, Seattle, Washington 98109, USA
| | - Ross E. Zumwalt
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Lisa M. Barton
- Office of the Chief Medical Examiner, Oklahoma City, Oklahoma 73105, USA
| | - Eric J. Duval
- Office of the Chief Medical Examiner, Oklahoma City, Oklahoma 73105, USA
| | | | - Edana Stroberg
- Office of the Chief Medical Examiner, Oklahoma City, Oklahoma 73105, USA
| | - Alain Borczuk
- Weill Cornell Medicine, New York, New York 10065, USA
| | - Leigh B. Thorne
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Muthu K. Sakthivel
- Department of Radiology, University of North Carolina at Chapel Hill, North Carolina 27599, USA
| | - Yueh Z. Lee
- Department of Radiology, University of North Carolina at Chapel Hill, North Carolina 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - James S. Hagood
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pediatrics, Pulmonology Division and Program for Rare and Interstitial Lung Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jason R. Mock
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Max A. Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado 80206, USA
- Department of Pediatrics, National Jewish Health, Denver, Colorado 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Stephanie A. Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Richard C. Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ralph S. Baric
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Rapidly Emerging Antiviral Drug Discovery Initiative, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
113
|
Voloudakis G, Vicari JM, Venkatesh S, Hoffman GE, Dobrindt K, Zhang W, Beckmann ND, Higgins CA, Argyriou S, Jiang S, Hoagland D, Gao L, Corvelo A, Cho K, Lee KM, Bian J, Lee JS, Iyengar SK, Luoh SW, Akbarian S, Striker R, Assimes TL, Schadt EE, Lynch JA, Merad M, tenOever BR, Charney AW, Brennand KJ, Fullard JF, Roussos P. A translational genomics approach identifies IL10RB as the top candidate gene target for COVID-19 susceptibility. NPJ Genom Med 2022; 7:52. [PMID: 36064543 PMCID: PMC9441828 DOI: 10.1038/s41525-022-00324-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 08/19/2022] [Indexed: 02/06/2023] Open
Abstract
Recent efforts have identified genetic loci that are associated with coronavirus disease 2019 (COVID-19) infection rates and disease outcome severity. Translating these genetic findings into druggable genes that reduce COVID-19 host susceptibility is a critical next step. Using a translational genomics approach that integrates COVID-19 genetic susceptibility variants, multi-tissue genetically regulated gene expression (GReX), and perturbagen signatures, we identified IL10RB as the top candidate gene target for COVID-19 host susceptibility. In a series of validation steps, we show that predicted GReX upregulation of IL10RB and higher IL10RB expression in COVID-19 patient blood is associated with worse COVID-19 outcomes and that in vitro IL10RB overexpression is associated with increased viral load and activation of disease-relevant molecular pathways.
Collapse
Affiliation(s)
- Georgios Voloudakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA.
| | - James M Vicari
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanan Venkatesh
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
| | - Gabriel E Hoffman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristina Dobrindt
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Wen Zhang
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noam D Beckmann
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christina A Higgins
- Department of Microbiology, Grossman School of Medicine, New York University, New York, NY, USA
- Department of Medicine, Grossman School of Medicine, New York University, New York, NY, USA
- Vilcek Institute of Graduate Biomedical Sciences, New York University, New York, NY, USA
| | - Stathis Argyriou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shan Jiang
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daisy Hoagland
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Virus Engineering Center for Therapeutics and Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lina Gao
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- VA Portland Health Care System, Portland, OR, USA
| | | | - Kelly Cho
- VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kyung Min Lee
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Jiantao Bian
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
- Division of Epidemiology, University of Utah, Salt Lake City, UT, USA
| | - Jennifer S Lee
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Sudha K Iyengar
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- VA Northeast Ohio Healthcare System, Cleveland VA Medical Center, Cleveland, OH, USA
| | - Shiuh-Wen Luoh
- VA Portland Health Care System, Portland, OR, USA
- Department of Medicine, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Striker
- Division of Infectious Diseases, Department of Medicine, University of Wisconsin, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Themistocles L Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Sema4, Stamford, CT, USA
| | - Julie A Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
- Division of Epidemiology, University of Utah, Salt Lake City, UT, USA
| | - Miriam Merad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin R tenOever
- Department of Microbiology, Grossman School of Medicine, New York University, New York, NY, USA
- Department of Medicine, Grossman School of Medicine, New York University, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Virus Engineering Center for Therapeutics and Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander W Charney
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen J Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - John F Fullard
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
114
|
Sparks R, Lau WW, Liu C, Han KL, Vrindten KL, Sun G, Cox M, Andrews SF, Bansal N, Failla LE, Manischewitz J, Grubbs G, King LR, Koroleva G, Leimenstoll S, Snow L, Chen J, Tang J, Mukherjee A, Sellers BA, Apps R, McDermott AB, Martins AJ, Bloch EM, Golding H, Khurana S, Tsang JS. Influenza vaccination and single cell multiomics reveal sex dimorphic immune imprints of prior mild COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.02.17.22271138. [PMID: 35233581 PMCID: PMC8887138 DOI: 10.1101/2022.02.17.22271138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Viral infections can have profound and durable functional impacts on the immune system. There is an urgent need to characterize the long-term immune effects of SARS-CoV-2 infection given the persistence of symptoms in some individuals and the continued threat of novel variants. Here we use systems immunology, including longitudinal multimodal single cell analysis (surface proteins, transcriptome, and V(D)J sequences) from 33 previously healthy individuals after recovery from mild, non-hospitalized COVID-19 and 40 age- and sex-matched healthy controls with no history of COVID-19 to comparatively assess the post-infection immune status (mean: 151 days after diagnosis) and subsequent innate and adaptive responses to seasonal influenza vaccination. Identification of both sex-specific and -independent temporally stable changes, including signatures of T-cell activation and repression of innate defense/immune receptor genes (e.g., Toll-like receptors) in monocytes, suggest that mild COVID-19 can establish new post-recovery immunological set-points. COVID-19-recovered males had higher innate, influenza-specific plasmablast, and antibody responses after vaccination compared to healthy males and COVID-19-recovered females, partly attributable to elevated pre-vaccination frequencies of a GPR56 expressing CD8+ T-cell subset in male recoverees that are "poised" to produce higher levels of IFNγ upon inflammatory stimulation. Intriguingly, by day 1 post-vaccination in COVID-19-recovered subjects, the expression of the repressed genes in monocytes increased and moved towards the pre-vaccination baseline of healthy controls, suggesting that the acute inflammation induced by vaccination could partly reset the immune states established by mild COVID-19. Our study reveals sex-dimorphic immune imprints and in vivo functional impacts of mild COVID-19 in humans, suggesting that prior COVID-19, and possibly respiratory viral infections in general, could change future responses to vaccination and in turn, vaccines could help reset the immune system after COVID-19, both in an antigen-agnostic manner.
Collapse
Affiliation(s)
- Rachel Sparks
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,These authors contributed equally
| | - William W. Lau
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,These authors contributed equally
| | - Can Liu
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,Graduate Program in Biological Sciences, University of Maryland, College Park, MD, USA,These authors contributed equally
| | - Kyu Lee Han
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Kiera L. Vrindten
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Guangping Sun
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Milann Cox
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | | | - Neha Bansal
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Laura E. Failla
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Jody Manischewitz
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Lisa R. King
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Galina Koroleva
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | | | - LaQuita Snow
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | | | - Jinguo Chen
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | | | | | - Richard Apps
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | | | - Andrew J. Martins
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Evan M. Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - John S. Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA,Correspondence:
| |
Collapse
|
115
|
Velsko IM, Semerau L, Inskip SA, García-Collado MI, Ziesemer K, Ruber MS, Benítez de Lugo Enrich L, Molero García JM, Valle DG, Peña Ruiz AC, Salazar-García DC, Hoogland MLP, Warinner C. Ancient dental calculus preserves signatures of biofilm succession and interindividual variation independent of dental pathology. PNAS NEXUS 2022; 1:pgac148. [PMID: 36714834 PMCID: PMC9802386 DOI: 10.1093/pnasnexus/pgac148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023]
Abstract
Dental calculus preserves oral microbes, enabling comparative studies of the oral microbiome and health through time. However, small sample sizes and limited dental health metadata have hindered health-focused investigations to date. Here, we investigate the relationship between tobacco pipe smoking and dental calculus microbiomes. Dental calculus from 75 individuals from the 19th century Middenbeemster skeletal collection (Netherlands) were analyzed by metagenomics. Demographic and dental health parameters were systematically recorded, including the presence/number of pipe notches. Comparative data sets from European populations before and after the introduction of tobacco were also analyzed. Calculus species profiles were compared with oral pathology to examine associations between microbiome community, smoking behavior, and oral health status. The Middenbeemster individuals exhibited relatively poor oral health, with a high prevalence of periodontal disease, caries, heavy calculus deposits, and antemortem tooth loss. No associations between pipe notches and dental pathologies, or microbial species composition, were found. Calculus samples before and after the introduction of tobacco showed highly similar species profiles. Observed interindividual microbiome differences were consistent with previously described variation in human populations from the Upper Paleolithic to the present. Dental calculus may not preserve microbial indicators of health and disease status as distinctly as dental plaque.
Collapse
Affiliation(s)
- Irina M Velsko
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig 04103, Germany
| | - Lena Semerau
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig 04103, Germany
- Faculty of Biological Sciences, Friedrich Schiller University, Jena 07743, Germany
| | - Sarah A Inskip
- School of Archaeology and Ancient History, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Maite I García-Collado
- GIPYPAC, Department of Geography, Prehistory and Archaeology, University of the Basque Country, Leioa 48940, Spain
- BioArCh, Department of Archaeology, University of York, York YO10 5NG, UK
| | - Kirsten Ziesemer
- University Library, Vrije Universiteit, Einsteinweg 2, Amsterdam 1081 HV, The Netherlands
| | - Maria Serrano Ruber
- School of Archaeology and Ancient History, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Luis Benítez de Lugo Enrich
- Departmento de Prehistoria, Historia Antigua y Arqueología, Universidad Complutense de Madrid, Madrid 28040, Spain
| | | | - David Gallego Valle
- Facultad de Letras, Universidad de Castilla-La Mancha, Ciudad Real 13004, Spain
| | | | - Domingo C Salazar-García
- Departament de Prehistòria, Historia i Arqueología, Universitat de València, València 46010, Spain
- Department of Geological Sciences, University of Cape Town, Rondebosch 7701, South Africa
| | - Menno L P Hoogland
- Faculty of Archaeology, Leiden University, Einsteinweg, Leiden 2333 CC, The Netherlands
| | - Christina Warinner
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig 04103, Germany
- Faculty of Biological Sciences, Friedrich Schiller University, Jena 07743, Germany
- Department of Anthropology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
116
|
Tcw J, Qian L, Pipalia NH, Chao MJ, Liang SA, Shi Y, Jain BR, Bertelsen SE, Kapoor M, Marcora E, Sikora E, Andrews EJ, Martini AC, Karch CM, Head E, Holtzman DM, Zhang B, Wang M, Maxfield FR, Poon WW, Goate AM. Cholesterol and matrisome pathways dysregulated in astrocytes and microglia. Cell 2022; 185:2213-2233.e25. [PMID: 35750033 PMCID: PMC9340815 DOI: 10.1016/j.cell.2022.05.017] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/07/2020] [Accepted: 05/16/2022] [Indexed: 12/12/2022]
Abstract
The impact of apolipoprotein E ε4 (APOE4), the strongest genetic risk factor for Alzheimer's disease (AD), on human brain cellular function remains unclear. Here, we investigated the effects of APOE4 on brain cell types derived from population and isogenic human induced pluripotent stem cells, post-mortem brain, and APOE targeted replacement mice. Population and isogenic models demonstrate that APOE4 local haplotype, rather than a single risk allele, contributes to risk. Global transcriptomic analyses reveal human-specific, APOE4-driven lipid metabolic dysregulation in astrocytes and microglia. APOE4 enhances de novo cholesterol synthesis despite elevated intracellular cholesterol due to lysosomal cholesterol sequestration in astrocytes. Further, matrisome dysregulation is associated with upregulated chemotaxis, glial activation, and lipid biosynthesis in astrocytes co-cultured with neurons, which recapitulates altered astrocyte matrisome signaling in human brain. Thus, APOE4 initiates glia-specific cell and non-cell autonomous dysregulation that may contribute to increased AD risk.
Collapse
Affiliation(s)
- Julia Tcw
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Lu Qian
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nina H Pipalia
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michael J Chao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Sanofi US, Cambridge, MA 02141, USA
| | - Shuang A Liang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Yang Shi
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63108, USA; Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Bharat R Jain
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Sarah E Bertelsen
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Manav Kapoor
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Edoardo Marcora
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elizabeth Sikora
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elizabeth J Andrews
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
| | - Alessandra C Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
| | - Celeste M Karch
- Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Elizabeth Head
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63108, USA; Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Frederick R Maxfield
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Wayne W Poon
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; NeuCyte, Inc., Mountain View, CA 94043, USA
| | - Alison M Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
117
|
Exposure to the Amino Acids Histidine, Lysine, and Threonine Reduces mTOR Activity and Affects Neurodevelopment in a Human Cerebral Organoid Model. Nutrients 2022; 14:nu14102175. [PMID: 35631316 PMCID: PMC9145399 DOI: 10.3390/nu14102175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/08/2022] [Accepted: 05/19/2022] [Indexed: 02/05/2023] Open
Abstract
Evidence of the impact of nutrition on human brain development is compelling. Previous in vitro and in vivo results show that three specific amino acids, histidine, lysine, and threonine, synergistically inhibit mTOR activity and behavior. Therefore, the prenatal availability of these amino acids could be important for human neurodevelopment. However, methods to study the underlying mechanisms in a human model of neurodevelopment are limited. Here, we pioneer the use of human cerebral organoids to investigate the impact of amino acid supplementation on neurodevelopment. In this study, cerebral organoids were exposed to 10 mM and 50 mM of the amino acids threonine, histidine, and lysine. The impact was determined by measuring mTOR activity using Western blots, general cerebral organoid size, and gene expression by RNA sequencing. Exposure to threonine, histidine, and lysine led to decreased mTOR activity and markedly reduced organoid size, supporting findings in rodent studies. RNA sequencing identified comprehensive changes in gene expression, with enrichment in genes related to specific biological processes (among which are mTOR signaling and immune function) and to specific cell types, including proliferative precursor cells, microglia, and astrocytes. Altogether, cerebral organoids are responsive to nutritional exposure by increasing specific amino acid concentrations and reflect findings from previous rodent studies. Threonine, histidine, and lysine exposure impacts the early development of human cerebral organoids, illustrated by the inhibition of mTOR activity, reduced size, and altered gene expression.
Collapse
|
118
|
Hoffman JR, Park HJ, Bheri S, Jayaraman AR, Davis ME. Comparative computational RNA analysis of cardiac-derived progenitor cells and their extracellular vesicles. Genomics 2022; 114:110349. [PMID: 35346780 PMCID: PMC9510608 DOI: 10.1016/j.ygeno.2022.110349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/18/2022] [Accepted: 03/23/2022] [Indexed: 01/14/2023]
Abstract
Stem/progenitor cells, including cardiac-derived c-kit+ progenitor cells (CPCs), are under clinical evaluation for treatment of cardiac disease. Therapeutic efficacy of cardiac cell therapy can be attributed to paracrine signaling and the release of extracellular vesicles (EVs) carrying diverse cargo molecules. Despite some successes and demonstrated safety, large variation in cell populations and preclinical/clinical outcomes remains a problem. Here, we investigated this variability by sequencing coding and non-coding RNAs of CPCs and CPC-EVs from 30 congenital heart disease patients and used machine learning methods to determine potential mechanistic insights. CPCs retained RNAs related to extracellular matrix organization and exported RNAs related to various signaling pathways to CPC-EVs. CPC-EVs are enriched in miRNA clusters related to cell proliferation and angiogenesis. With network analyses, we identified differences in non-coding RNAs which give insight into age-dependent functionality of CPCs. By taking a quantitative computational approach, we aimed to uncover sources of CPC cell therapy variability.
Collapse
Affiliation(s)
- Jessica R. Hoffman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA.,Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arun R. Jayaraman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA.,Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, USA.,Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia, USA.,Corresponding author at: Professor of Biomedical Engineering, 2015 Uppergate Drive, 310, Atlanta, GA 30322, USA,
| |
Collapse
|
119
|
The transcriptome from asexual to sexual in vitro development of Cystoisospora suis (Apicomplexa: Coccidia). Sci Rep 2022; 12:5972. [PMID: 35396557 PMCID: PMC8993856 DOI: 10.1038/s41598-022-09714-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/15/2022] [Indexed: 11/08/2022] Open
Abstract
The apicomplexan parasite Cystoisospora suis is an enteropathogen of suckling piglets with woldwide distribution. As with all coccidian parasites, its lifecycle is characterized by asexual multiplication followed by sexual development with two morphologically distinct cell types that presumably fuse to form a zygote from which the oocyst arises. However, knowledge of the sexual development of C. suis is still limited. To complement previous in vitro studies, we analysed transcriptional profiles at three different time points of development (corresponding to asexual, immature and mature sexual stages) in vitro via RNASeq. Overall, transcription of genes encoding proteins with important roles in gametes biology, oocyst wall biosynthesis, DNA replication and axonema formation as well as proteins with important roles in merozoite biology was identified. A homologue of an oocyst wall tyrosine rich protein of Toxoplasma gondii was expressed in macrogametes and oocysts of C. suis. We evaluated inhibition of sexual development in a host-free culture for C. suis by antiserum specific to this protein to evaluate whether it could be exploited as a candidate for control strategies against C. suis. Based on these data, targets can be defined for future strategies to interrupt parasite transmission during sexual development.
Collapse
|
120
|
de Witte LD, Wang Z, Snijders GLJL, Mendelev N, Liu Q, Sneeboer MAM, Boks MPM, Ge Y, Haghighi F. Contribution of Age, Brain Region, Mood Disorder Pathology, and Interindividual Factors on the Methylome of Human Microglia. Biol Psychiatry 2022; 91:572-581. [PMID: 35027166 PMCID: PMC11181298 DOI: 10.1016/j.biopsych.2021.10.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Transcriptome studies have revealed age-, disease-, and region-associated microglial phenotypes reflecting changes in microglial function during development, aging, central nervous system homeostasis, and pathology. The molecular mechanisms that contribute to these transcriptomic changes are largely unknown. The aim of this study was to characterize the DNA methylation landscape of human microglia and the factors that contribute to variations in the microglia methylome. We hypothesized that both age and brain region would have a large impact on DNA methylation in microglia. METHODS Microglia from postmortem brain tissue of four different brain regions of 22 donors, encompassing 1 patient with schizophrenia, 13 patients with mood disorder pathology, and 8 control subjects, were isolated and assayed using a genome-wide methylation array. RESULTS We found that human microglial cells have a methylation profile distinct from bulk brain tissue and neurons, and age explained a considerable part of the variation. Additionally, we showed that interindividual factors had a much larger effect on the methylation landscape of microglia than brain region, which was also seen at the transcriptome level. In our exploratory analysis, we found various differentially methylated regions that were related to disease status (mood disorder vs. control). This included differentially methylated regions that are linked to gene expression in microglia, as well as to myeloid cell function or neuropsychiatric disorders. CONCLUSIONS Although based on relatively small samples, these findings suggest that the methylation profile of microglia is responsive to interindividual variations and thereby plays an important role in the heterogeneity of microglia observed at the transcriptome level.
Collapse
Affiliation(s)
- Lot D de Witte
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zhaoyu Wang
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gijsje L J L Snijders
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Natalia Mendelev
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Qingkun Liu
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marjolein A M Sneeboer
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands; Department of Translational Neuroscience, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Marco P M Boks
- Department of Psychiatry, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fatemeh Haghighi
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
121
|
Lirakis M, Nolte V, Schlötterer C. Pool-GWAS on reproductive dormancy in Drosophila simulans suggests a polygenic architecture. G3 GENES|GENOMES|GENETICS 2022; 12:6523974. [PMID: 35137042 PMCID: PMC8895979 DOI: 10.1093/g3journal/jkac027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/29/2021] [Indexed: 11/29/2022]
Abstract
The genetic basis of adaptation to different environments has been of long-standing interest to evolutionary biologists. Dormancy is a well-studied adaptation to facilitate overwintering. In Drosophila melanogaster, a moderate number of genes with large effects have been described, which suggests a simple genetic basis of dormancy. On the other hand, genome-wide scans for dormancy suggest a polygenic architecture in insects. In D. melanogaster, the analysis of the genetic architecture of dormancy is complicated by the presence of cosmopolitan inversions. Here, we performed a genome-wide scan to characterize the genetic basis of this ecologically extremely important trait in the sibling species of D. melanogaster, D. simulans that lacks cosmopolitan inversions. We performed Pool-GWAS in a South African D. simulans population for dormancy incidence at 2 temperature regimes (10 and 12°C, LD 10:14). We identified several genes with SNPs that showed a significant association with dormancy (P-value < 1e-13), but the overall modest response suggests that dormancy is a polygenic trait with many loci of small effect. Our results shed light on controversies on reproductive dormancy in Drosophila and have important implications for the characterization of the genetic basis of this trait.
Collapse
Affiliation(s)
- Manolis Lirakis
- Institut für Populationsgenetik, Vetmeduni Vienna, 1210 Wien, Austria
- Vienna Graduate School of Population Genetics, Vetmeduni Vienna, 1210 Wien, Austria
| | - Viola Nolte
- Institut für Populationsgenetik, Vetmeduni Vienna, 1210 Wien, Austria
| | | |
Collapse
|
122
|
Housman G, Briscoe E, Gilad Y. Evolutionary insights into primate skeletal gene regulation using a comparative cell culture model. PLoS Genet 2022; 18:e1010073. [PMID: 35263340 PMCID: PMC8936463 DOI: 10.1371/journal.pgen.1010073] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 03/21/2022] [Accepted: 02/02/2022] [Indexed: 01/10/2023] Open
Abstract
The evolution of complex skeletal traits in primates was likely influenced by both genetic and environmental factors. Because skeletal tissues are notoriously challenging to study using functional genomic approaches, they remain poorly characterized even in humans, let alone across multiple species. The challenges involved in obtaining functional genomic data from the skeleton, combined with the difficulty of obtaining such tissues from nonhuman apes, motivated us to consider an alternative in vitro system with which to comparatively study gene regulation in skeletal cell types. Specifically, we differentiated six human (Homo sapiens) and six chimpanzee (Pan troglodytes) induced pluripotent stem cell lines (iPSCs) into mesenchymal stem cells (MSCs) and subsequently into osteogenic cells (bone cells). We validated differentiation using standard methods and collected single-cell RNA sequencing data from over 100,000 cells across multiple samples and replicates at each stage of differentiation. While most genes that we examined display conserved patterns of expression across species, hundreds of genes are differentially expressed (DE) between humans and chimpanzees within and across stages of osteogenic differentiation. Some of these interspecific DE genes show functional enrichments relevant in skeletal tissue trait development. Moreover, topic modeling indicates that interspecific gene programs become more pronounced as cells mature. Overall, we propose that this in vitro model can be used to identify interspecific regulatory differences that may have contributed to skeletal trait differences between species. Primates display a range of skeletal morphologies and susceptibilities to skeletal diseases, but the molecular basis of these phenotypic differences is unclear. Studies of gene expression variation in primate skeletal tissues are extremely restricted due to the ethical and practical challenges associated with collecting samples. Nevertheless, the ability to study gene regulation in primate skeletal tissues is crucial for understanding how the primate skeleton has evolved. We therefore developed a comparative primate skeletal cell culture model that allows us to access a spectrum of human and chimpanzee cell types as they differentiate from stem cells into bone cells. While most gene expression patterns are conserved across species, we also identified hundreds of differentially expressed genes between humans and chimpanzees within and across stages of differentiation. We also classified cells by osteogenic stage and identified additional interspecific differentially expressed genes which may contribute to skeletal trait differences. We anticipate that this model will be extremely useful for exploring questions related to gene regulation variation in primate bone biology and development.
Collapse
Affiliation(s)
- Genevieve Housman
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Emilie Briscoe
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Yoav Gilad
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
123
|
Laufer BI, Neier K, Valenzuela AE, Yasui DH, Schmidt RJ, Lein PJ, LaSalle JM. Placenta and fetal brain share a neurodevelopmental disorder DNA methylation profile in a mouse model of prenatal PCB exposure. Cell Rep 2022; 38:110442. [PMID: 35235788 PMCID: PMC8941983 DOI: 10.1016/j.celrep.2022.110442] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/19/2021] [Accepted: 02/03/2022] [Indexed: 12/27/2022] Open
Abstract
Polychlorinated biphenyls (PCBs) are developmental neurotoxicants implicated as environmental risk factors for neurodevelopmental disorders (NDDs). Here, we report the effects of prenatal exposure to a human-relevant mixture of PCBs on the DNA methylation profiles of mouse placenta and fetal brain. Thousands of differentially methylated regions (DMRs) distinguish placenta and fetal brain from PCB-exposed mice from sex-matched vehicle controls. In both placenta and fetal brain, PCB-associated DMRs are enriched for functions related to neurodevelopment and cellular signaling and enriched within regions of bivalent chromatin. The placenta and brain PCB DMRs overlap significantly and map to a shared subset of genes enriched for Wnt signaling, Slit/Robo signaling, and genes differentially expressed in NDD models. The consensus PCB DMRs also significantly overlap with DMRs from human NDD brain and placenta. These results demonstrate that PCB-exposed placenta contains a subset of DMRs that overlap fetal brain DMRs relevant to an NDD.
Collapse
Affiliation(s)
- Benjamin I Laufer
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA; UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA; MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Kari Neier
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA; UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA; MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA; Perinatal Origins of Disparities Center, University of California, Davis, Davis, CA 95616, USA
| | - Anthony E Valenzuela
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Dag H Yasui
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA; UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA; MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Rebecca J Schmidt
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA; Perinatal Origins of Disparities Center, University of California, Davis, Davis, CA 95616, USA; Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Pamela J Lein
- MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA; Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA; UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA; MIND Institute, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA; Perinatal Origins of Disparities Center, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
124
|
Oh VKS, Li RW. Large-Scale Meta-Longitudinal Microbiome Data with a Known Batch Factor. Genes (Basel) 2022; 13:392. [PMID: 35327945 PMCID: PMC8953633 DOI: 10.3390/genes13030392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/05/2022] [Accepted: 02/18/2022] [Indexed: 12/04/2022] Open
Abstract
Data contamination in meta-approaches where multiple biological samples are combined considerably affects the results of subsequent downstream analyses, such as differential abundance tests comparing multiple groups at a fixed time point. Little has been thoroughly investigated regarding the impact of the lurking variable of various batch sources, such as different days or different laboratories, in more complicated time series experimental designs, for instance, repeatedly measured longitudinal data and metadata. We highlight that the influence of batch factors is significant on subsequent downstream analyses, including longitudinal differential abundance tests, by performing a case study of microbiome time course data with two treatment groups and a simulation study of mimic microbiome longitudinal counts.
Collapse
Affiliation(s)
- Vera-Khlara S. Oh
- United States Department of Agriculture, Agricultural Research Service, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
- Department of Data Science, College of Natural Sciences, Jeju National University, Jeju City 690-756, Korea
| | - Robert W. Li
- United States Department of Agriculture, Agricultural Research Service, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
| |
Collapse
|
125
|
Dinnon KH, Leist SR, Okuda K, Dang H, Fritch EJ, Gully KL, De la Cruz G, Evangelista MD, Asakura T, Gilmore RC, Hawkins P, Nakano S, West A, Schäfer A, Gralinski LE, Everman JL, Sajuthi SP, Zweigart MR, Dong S, McBride J, Cooley MR, Hines JB, Love MK, Groshong SD, VanSchoiack A, Phelan SJ, Liang Y, Hether T, Leon M, Zumwalt RE, Barton LM, Duval EJ, Mukhopadhyay S, Stroberg E, Borczuk A, Thorne LB, Sakthivel MK, Lee YZ, Hagood JS, Mock JR, Seibold MA, O’Neal WK, Montgomery SA, Boucher RC, Baric RS. A model of persistent post SARS-CoV-2 induced lung disease for target identification and testing of therapeutic strategies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.02.15.480515. [PMID: 35194605 PMCID: PMC8863140 DOI: 10.1101/2022.02.15.480515] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
COVID-19 survivors develop post-acute sequelae of SARS-CoV-2 (PASC), but the mechanistic basis of PASC-associated lung abnormalities suffers from a lack of longitudinal samples. Mouse-adapted SARS-CoV-2 MA10 produces an acute respiratory distress syndrome (ARDS) in mice similar to humans. To investigate PASC pathogenesis, studies of MA10-infected mice were extended from acute disease through clinical recovery. At 15-120 days post-virus clearance, histologic evaluation identified subpleural lesions containing collagen, proliferative fibroblasts, and chronic inflammation with tertiary lymphoid structures. Longitudinal spatial transcriptional profiling identified global reparative and fibrotic pathways dysregulated in diseased regions, similar to human COVID-19. Populations of alveolar intermediate cells, coupled with focal upregulation of pro-fibrotic markers, were identified in persistently diseased regions. Early intervention with antiviral EIDD-2801 reduced chronic disease, and early anti-fibrotic agent (nintedanib) intervention modified early disease severity. This murine model provides opportunities to identify pathways associated with persistent SARS-CoV-2 pulmonary disease and test countermeasures to ameliorate PASC.
Collapse
Affiliation(s)
- Kenneth H. Dinnon
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah R. Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ethan J. Fritch
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kendra L. Gully
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mia D. Evangelista
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Takanori Asakura
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rodney C. Gilmore
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Padraig Hawkins
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Satoko Nakano
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ande West
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lisa E. Gralinski
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jamie L. Everman
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| | - Satria P. Sajuthi
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| | - Mark R. Zweigart
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephanie Dong
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer McBride
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michelle R. Cooley
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jesse B. Hines
- Golden Point Scientific Laboratories, Hoover, Alabama, USA
| | - Miriya K. Love
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Steve D. Groshong
- Division of Pathology, Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | | | | | - Yan Liang
- NanoString Technologies, Seattle, Washington, USA
| | - Tyler Hether
- NanoString Technologies, Seattle, Washington, USA
| | - Michael Leon
- NanoString Technologies, Seattle, Washington, USA
| | - Ross E. Zumwalt
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Lisa M. Barton
- Office of the Chief Medical Examiner, Oklahoma City, Oklahoma, USA
| | - Eric J. Duval
- Office of the Chief Medical Examiner, Oklahoma City, Oklahoma, USA
| | | | - Edana Stroberg
- Office of the Chief Medical Examiner, Oklahoma City, Oklahoma, USA
| | | | - Leigh B. Thorne
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Muthu K. Sakthivel
- Department of Radiology, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Yueh Z. Lee
- Department of Radiology, University of North Carolina at Chapel Hill, North Carolina, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - James S. Hagood
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pediatrics, Pulmonology Division and Program for Rare and Interstitial Lung Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jason R. Mock
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Max A. Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado-Denver, Denver, Colorado, USA
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephanie A. Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Richard C. Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ralph S. Baric
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Rapidly Emerging Antiviral Drug Discovery Initiative, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
126
|
Shohat S, Vol E, Shifman S. Gene essentiality in cancer cell lines is modified by the sex chromosomes. Genome Res 2022; 32:1993-2002. [PMID: 36418059 PMCID: PMC9808629 DOI: 10.1101/gr.276488.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
Human sex differences arise from gonadal hormones and sex chromosomes. Studying the direct effects of sex chromosomes in humans is still challenging. Here we studied how the sex chromosomes can modulate gene expression and the outcome of mutations across the genome by exploiting the tendency of cancer cell lines to lose or gain sex chromosomes. We inferred the dosage of the sex chromosomes in 355 female and 408 male cancer cell lines and used it to dissect the contributions of the Y and X Chromosomes to sex-biased gene expression. Furthermore, based on genome-wide CRISPR screens, we identified genes whose essentiality is different between male and female cells depending on the sex chromosomes. The most significant genes were X-linked genes compensated by Y-linked paralogs. Our sex-based analysis identifies genes that, when mutated, can affect male and female cells differently and reinforces the roles of the X and Y Chromosomes in sex-specific cell function.
Collapse
|
127
|
Lopes KDP, Snijders GJL, Humphrey J, Allan A, Sneeboer MAM, Navarro E, Schilder BM, Vialle RA, Parks M, Missall R, van Zuiden W, Gigase FAJ, Kübler R, van Berlekom AB, Hicks EM, Bӧttcher C, Priller J, Kahn RS, de Witte LD, Raj T. Genetic analysis of the human microglial transcriptome across brain regions, aging and disease pathologies. Nat Genet 2022; 54:4-17. [PMID: 34992268 PMCID: PMC9245609 DOI: 10.1038/s41588-021-00976-y] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
Microglia have emerged as important players in brain aging and pathology. To understand how genetic risk for neurological and psychiatric disorders is related to microglial function, large transcriptome studies are essential. Here we describe the transcriptome analysis of 255 primary human microglial samples isolated at autopsy from multiple brain regions of 100 individuals. We performed systematic analyses to investigate various aspects of microglial heterogeneities, including brain region and aging. We mapped expression and splicing quantitative trait loci and showed that many neurological disease susceptibility loci are mediated through gene expression or splicing in microglia. Fine-mapping of these loci nominated candidate causal variants that are within microglia-specific enhancers, finding associations with microglial expression of USP6NL for Alzheimer's disease and P2RY12 for Parkinson's disease. We have built the most comprehensive catalog to date of genetic effects on the microglial transcriptome and propose candidate functional variants in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Katia de Paiva Lopes
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gijsje J L Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Jack Humphrey
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda Allan
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marjolein A M Sneeboer
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Elisa Navarro
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian M Schilder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ricardo A Vialle
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Parks
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roy Missall
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Welmoed van Zuiden
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frederieke A J Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Raphael Kübler
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amber Berdenis van Berlekom
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Emily M Hicks
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chotima Bӧttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA.
| | - Towfique Raj
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
128
|
Hoffman GE, Ma Y, Montgomery KS, Bendl J, Jaiswal MK, Kozlenkov A, Peters MA, Dracheva S, Fullard JF, Chess A, Devlin B, Sieberts SK, Roussos P. Sex Differences in the Human Brain Transcriptome of Cases With Schizophrenia. Biol Psychiatry 2022; 91:92-101. [PMID: 34154796 PMCID: PMC8463632 DOI: 10.1016/j.biopsych.2021.03.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND While schizophrenia differs between males and females in the age of onset, symptomatology, and disease course, the molecular mechanisms underlying these differences remain uncharacterized. METHODS To address questions about the sex-specific effects of schizophrenia, we performed a large-scale transcriptome analysis of RNA sequencing data from 437 controls and 341 cases from two distinct cohorts from the CommonMind Consortium. RESULTS Analysis across the cohorts identified a reproducible gene expression signature of schizophrenia that was highly concordant with previous work. Differential expression across sex was reproducible across cohorts and identified X- and Y-linked genes, as well as those involved in dosage compensation. Intriguingly, the sex expression signature was also enriched for genes involved in neurexin family protein binding and synaptic organization. Differential expression analysis testing a sex-by-diagnosis interaction effect did not identify any genome-wide signature after multiple testing corrections. Gene coexpression network analysis was performed to reduce dimensionality from thousands of genes to dozens of modules and elucidate interactions among genes. We found enrichment of coexpression modules for sex-by-diagnosis differential expression signatures, which were highly reproducible across the two cohorts and involved a number of diverse pathways, including neural nucleus development, neuron projection morphogenesis, and regulation of neural precursor cell proliferation. CONCLUSIONS Overall, our results indicate that the effect size of sex differences in schizophrenia gene expression signatures is small and underscore the challenge of identifying robust sex-by-diagnosis signatures, which will require future analyses in larger cohorts.
Collapse
Affiliation(s)
- Gabriel E Hoffman
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Yixuan Ma
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Jaroslav Bendl
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Manoj Kumar Jaiswal
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - Alex Kozlenkov
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | | | - Stella Dracheva
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - John F Fullard
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Andrew Chess
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Panos Roussos
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York.
| |
Collapse
|
129
|
Ku AT, Wilkinson S, Sowalsky AG. Comparison of approaches to transcriptomic analysis in multi-sampled tumors. Brief Bioinform 2021; 22:6355417. [PMID: 34415294 DOI: 10.1093/bib/bbab337] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/14/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022] Open
Abstract
Intratumoral heterogeneity is a well-documented feature of human cancers and is associated with outcome and treatment resistance. However, a heterogeneous tumor transcriptome contributes an unknown level of variability to analyses of differentially expressed genes (DEGs) that may contribute to phenotypes of interest, including treatment response. Although current clinical practice and the vast majority of research studies use a single sample from each patient, decreasing costs of sequencing technologies and computing power have made repeated-measures analyses increasingly economical. Repeatedly sampling the same tumor increases the statistical power of DEG analysis, which is indispensable toward downstream analysis and also increases one's understanding of within-tumor variance, which may affect conclusions. Here, we compared five different methods for analyzing gene expression profiles derived from repeated sampling of human prostate tumors in two separate cohorts of patients. We also benchmarked the sensitivity of generalized linear models to linear mixed models for identifying DEGs contributing to relevant prostate cancer pathways based on a ground-truth model.
Collapse
Affiliation(s)
- Anson T Ku
- Laboratory of Genitourinary Cancer Pathogenesis (LGCP) at the National Cancer Institute (NCI), NIH, 37 Convent Drive, Building 37, Room 1062B, Bethesda, MD 20892, USA
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis (LGCP) at the National Cancer Institute (NCI), NIH, 37 Convent Drive, Building 37, Room 1062B, Bethesda, MD 20892, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis (LGCP) at the National Cancer Institute (NCI), NIH, 37 Convent Drive, Building 37, Room 1062B, Bethesda, MD 20892, USA
| |
Collapse
|
130
|
Li H, Zhu B, Xu Z, Adams T, Kaminski N, Zhao H. A Markov random field model for network-based differential expression analysis of single-cell RNA-seq data. BMC Bioinformatics 2021; 22:524. [PMID: 34702190 PMCID: PMC8549347 DOI: 10.1186/s12859-021-04412-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent development of single cell sequencing technologies has made it possible to identify genes with different expression (DE) levels at the cell type level between different groups of samples. In this article, we propose to borrow information through known biological networks to increase statistical power to identify differentially expressed genes (DEGs). RESULTS We develop MRFscRNAseq, which is based on a Markov random field (MRF) model to appropriately accommodate gene network information as well as dependencies among cell types to identify cell-type specific DEGs. We implement an Expectation-Maximization (EM) algorithm with mean field-like approximation to estimate model parameters and a Gibbs sampler to infer DE status. Simulation study shows that our method has better power to detect cell-type specific DEGs than conventional methods while appropriately controlling type I error rate. The usefulness of our method is demonstrated through its application to study the pathogenesis and biological processes of idiopathic pulmonary fibrosis (IPF) using a single-cell RNA-sequencing (scRNA-seq) data set, which contains 18,150 protein-coding genes across 38 cell types on lung tissues from 32 IPF patients and 28 normal controls. CONCLUSIONS The proposed MRF model is implemented in the R package MRFscRNAseq available on GitHub. By utilizing gene-gene and cell-cell networks, our method increases statistical power to detect differentially expressed genes from scRNA-seq data.
Collapse
Affiliation(s)
- Hongyu Li
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT 06511 USA
| | - Biqing Zhu
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511 USA
| | - Zhichao Xu
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT 06511 USA
| | - Taylor Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520 USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520 USA
| | - Hongyu Zhao
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT 06511 USA
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511 USA
| |
Collapse
|
131
|
Thompson RC, Simons NW, Wilkins L, Cheng E, Del Valle DM, Hoffman GE, Fennessy B, Mouskas K, Francoeur NJ, Johnson JS, Lepow L, Le Berichel J, Chang C, Beckmann AG, Wang YC, Nie K, Zaki N, Tuballes K, Barcessat V, Cedillo MA, Huckins L, Roussos P, Marron TU, Glicksberg BS, Nadkarni G, Gonzalez-Kozlova E, Kim-Schulze S, Sebra R, Merad M, Gnjatic S, Schadt EE, Charney AW, Beckmann ND. Acute COVID-19 gene-expression profiles show multiple etiologies of long-term sequelae. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.10.04.21264434. [PMID: 34642700 PMCID: PMC8509101 DOI: 10.1101/2021.10.04.21264434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Two years into the SARS-CoV-2 pandemic, the post-acute sequelae of infection are compounding the global health crisis. Often debilitating, these sequelae are clinically heterogeneous and of unknown molecular etiology. Here, a transcriptome-wide investigation of this new condition was performed in a large cohort of acutely infected patients followed clinically into the post-acute period. Gene expression signatures of post-acute sequelae were already present in whole blood during the acute phase of infection, with both innate and adaptive immune cells involved. Plasma cells stood out as driving at least two distinct clusters of sequelae, one largely dependent on circulating antibodies against the SARS-CoV-2 spike protein and the other antibody-independent. Altogether, multiple etiologies of post-acute sequelae were found concomitant with SARS-CoV-2 infection, directly linking the emergence of these sequelae with the host response to the virus.
Collapse
Affiliation(s)
- Ryan C. Thompson
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicole W. Simons
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lillian Wilkins
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Esther Cheng
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Diane Marie Del Valle
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Mount Sinai, New York, NY 10029, USA
| | - Gabriel E. Hoffman
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Brian Fennessy
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Konstantinos Mouskas
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Susan and Leonard Feinstein Inflammatory Bowel Disease Clinical Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nancy J. Francoeur
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Lauren Lepow
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Christie Chang
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Mount Sinai, New York, NY 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Ying-chih Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicholas Zaki
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kevin Tuballes
- Precision Immunology Institute, Mount Sinai, New York, NY 10029, USA
| | - Vanessa Barcessat
- Precision Immunology Institute, Mount Sinai, New York, NY 10029, USA
| | - Mario A. Cedillo
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Huckins
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Panagiotis Roussos
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Thomas U. Marron
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Benjamin S. Glicksberg
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
| | - Girish Nadkarni
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Division of Data Driven and Digital Medicine (D3M), Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
| | - Edgar Gonzalez-Kozlova
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Seunghee Kim-Schulze
- Precision Immunology Institute, Mount Sinai, New York, NY 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Sema4, a Mount Sinai venture, Stamford CT, 06902, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Merad
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Mount Sinai, New York, NY 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Oncological Sciences, Mount Sinai, New York, NY 10029, USA
| | - Eric E. Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Sema4, a Mount Sinai venture, Stamford CT, 06902, USA
| | - Alexander W. Charney
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Noam D. Beckmann
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
132
|
Novel insights into the molecular mechanisms underlying risk of colorectal cancer from smoking and red/processed meat carcinogens by modeling exposure in normal colon organoids. Oncotarget 2021; 12:1863-1877. [PMID: 34548904 PMCID: PMC8448508 DOI: 10.18632/oncotarget.28058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/13/2021] [Indexed: 12/23/2022] Open
Abstract
Tobacco smoke and red/processed meats are well-known risk factors for colorectal cancer (CRC). Most research has focused on studies of normal colon biopsies in epidemiologic studies or treatment of CRC cell lines in vitro. These studies are often constrained by challenges with accuracy of self-report data or, in the case of CRC cell lines, small sample sizes and lack of relationship to normal tissue at risk. In an attempt to address some of these limitations, we performed a 24-hour treatment of a representative carcinogens cocktail in 37 independent organoid lines derived from normal colon biopsies. Machine learning algorithms were applied to bulk RNA-sequencing and revealed cellular composition changes in colon organoids. We identified 738 differentially expressed genes in response to carcinogens exposure. Network analysis identified significantly different modules of co-expression, that included genes related to MSI-H tumor biology, and genes previously implicated in CRC through genome-wide association studies. Our study helps to better define the molecular effects of representative carcinogens from smoking and red/processed meat in normal colon epithelial cells and in the etiology of the MSI-H subtype of CRC, and suggests an overlap between molecular mechanisms involved in inherited and environmental CRC risk.
Collapse
|
133
|
Navarro E, Udine E, Lopes KDP, Parks M, Riboldi G, Schilder BM, Humphrey J, Snijders GJL, Vialle RA, Zhuang M, Sikder T, Argyrou C, Allan A, Chao MJ, Farrell K, Henderson B, Simon S, Raymond D, Elango S, Ortega RA, Shanker V, Swan M, Zhu CW, Ramdhani R, Walker RH, Tse W, Sano M, Pereira AC, Ahfeldt T, Goate AM, Bressman S, Crary JF, de Witte L, Frucht S, Saunders-Pullman R, Raj T. Dysregulation of mitochondrial and proteolysosomal genes in Parkinson's disease myeloid cells. NATURE AGING 2021; 1:850-863. [PMID: 35005630 PMCID: PMC8728893 DOI: 10.1038/s43587-021-00110-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022]
Abstract
An increasing number of identified Parkinson's disease (PD) risk loci contain genes highly expressed in innate immune cells, yet their role in pathology is not understood. We hypothesize that PD susceptibility genes modulate disease risk by influencing gene expression within immune cells. To address this, we have generated transcriptomic profiles of monocytes from 230 individuals with sporadic PD and healthy subjects. We observed a dysregulation of mitochondrial and proteasomal pathways. We also generated transcriptomic profiles of primary microglia from brains of 55 subjects and observed discordant transcriptomic signatures of mitochondrial genes in PD monocytes and microglia. We further identified 17 PD susceptibility genes whose expression, relative to each risk allele, is altered in monocytes. These findings reveal widespread transcriptomic alterations in PD monocytes, with some being distinct from microglia, and facilitate efforts to understand the roles of myeloid cells in PD as well as the development of biomarkers.
Collapse
Affiliation(s)
- Elisa Navarro
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evan Udine
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katia de Paiva Lopes
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Parks
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulietta Riboldi
- The Marlene and Paolo Fresco Institute for Parkinson's Disease and Movement Disorders, New York University Langone Health, New York, NY, USA
- Universita degli Studi di Milano, Molecular and Translational Medicine, Milan, Italy
| | - Brian M Schilder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jack Humphrey
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gijsje J L Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, NY, Bronx, USA
| | - Ricardo A Vialle
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maojuan Zhuang
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tamjeed Sikder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charalambos Argyrou
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda Allan
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael J Chao
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kurt Farrell
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brooklyn Henderson
- The Marlene and Paolo Fresco Institute for Parkinson's Disease and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Sarah Simon
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deborah Raymond
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sonya Elango
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roberto A Ortega
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vicki Shanker
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Swan
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolyn W Zhu
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Centers, James J. Peters VA Medical Center, New York, NY, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ritesh Ramdhani
- Department of Neurology, Zucker School of Medicine at Hofstra Northwell, New York, NY, USA
| | - Ruth H Walker
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Winona Tse
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Sano
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Centers, James J. Peters VA Medical Center, New York, NY, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana C Pereira
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tim Ahfeldt
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Bressman
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Crary
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lotje de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, NY, Bronx, USA
| | - Steven Frucht
- The Marlene and Paolo Fresco Institute for Parkinson's Disease and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Rachel Saunders-Pullman
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Towfique Raj
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
134
|
Bowles KR, Silva MC, Whitney K, Bertucci T, Berlind JE, Lai JD, Garza JC, Boles NC, Mahali S, Strang KH, Marsh JA, Chen C, Pugh DA, Liu Y, Gordon RE, Goderie SK, Chowdhury R, Lotz S, Lane K, Crary JF, Haggarty SJ, Karch CM, Ichida JK, Goate AM, Temple S. ELAVL4, splicing, and glutamatergic dysfunction precede neuron loss in MAPT mutation cerebral organoids. Cell 2021; 184:4547-4563.e17. [PMID: 34314701 PMCID: PMC8635409 DOI: 10.1016/j.cell.2021.07.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/06/2021] [Accepted: 06/30/2021] [Indexed: 12/21/2022]
Abstract
Frontotemporal dementia (FTD) because of MAPT mutation causes pathological accumulation of tau and glutamatergic cortical neuronal death by unknown mechanisms. We used human induced pluripotent stem cell (iPSC)-derived cerebral organoids expressing tau-V337M and isogenic corrected controls to discover early alterations because of the mutation that precede neurodegeneration. At 2 months, mutant organoids show upregulated expression of MAPT, glutamatergic signaling pathways, and regulators, including the RNA-binding protein ELAVL4, and increased stress granules. Over the following 4 months, mutant organoids accumulate splicing changes, disruption of autophagy function, and build-up of tau and P-tau-S396. By 6 months, tau-V337M organoids show specific loss of glutamatergic neurons as seen in individuals with FTD. Mutant neurons are susceptible to glutamate toxicity, which can be rescued pharmacologically by the PIKFYVE kinase inhibitor apilimod. Our results demonstrate a sequence of events that precede neurodegeneration, revealing molecular pathways associated with glutamate signaling as potential targets for therapeutic intervention in FTD.
Collapse
Affiliation(s)
- Kathryn R Bowles
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - M Catarina Silva
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kristen Whitney
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | | | - Joshua E Berlind
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jesse D Lai
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Amgen Research, One Amgen Center Dr., Thousand Oaks, CA 91320, USA
| | - Jacob C Garza
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Sidhartha Mahali
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kevin H Strang
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | - Jacob A Marsh
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Cynthia Chen
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Derian A Pugh
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - Yiyuan Liu
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - Ronald E Gordon
- Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | | | | | - Steven Lotz
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Keith Lane
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - John F Crary
- Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA.
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
135
|
Subramaniyan B, Larabee JL, Bodas M, Moore AR, Burgett AWG, Myers DA, Georgescu C, Wren JD, Papin JF, Walters MS. Characterization of the SARS-CoV-2 Host Response in Primary Human Airway Epithelial Cells from Aged Individuals. Viruses 2021; 13:v13081603. [PMID: 34452468 PMCID: PMC8402710 DOI: 10.3390/v13081603] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/29/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19), a global pandemic characterized by an exaggerated immune response and respiratory illness. Age (>60 years) is a significant risk factor for developing severe COVID-19. To better understand the host response of the aged airway epithelium to SARS-CoV-2 infection, we performed an in vitro study using primary human bronchial epithelial cells from donors >67 years of age differentiated on an air–liquid interface culture. We demonstrate that SARS-CoV-2 infection leads to early induction of a proinflammatory response and a delayed interferon response. In addition, we observed changes in the genes and pathways associated with cell death and senescence throughout infection. In summary, our study provides new and important insights into the temporal kinetics of the airway epithelial innate immune response to SARS-CoV-2 in older individuals.
Collapse
Affiliation(s)
- Bharathiraja Subramaniyan
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (B.S.); (M.B.); (A.R.M.)
| | - Jason L. Larabee
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Manish Bodas
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (B.S.); (M.B.); (A.R.M.)
| | - Andrew R. Moore
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (B.S.); (M.B.); (A.R.M.)
| | - Anthony W. G. Burgett
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Dean A. Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Constantin Georgescu
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (C.G.); (J.D.W.)
| | - Jonathan D. Wren
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (C.G.); (J.D.W.)
| | - James F. Papin
- Department of Pathology, Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Matthew S. Walters
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (B.S.); (M.B.); (A.R.M.)
- Correspondence:
| |
Collapse
|
136
|
Beckmann ND, Comella PH, Cheng E, Lepow L, Beckmann AG, Tyler SR, Mouskas K, Simons NW, Hoffman GE, Francoeur NJ, Del Valle DM, Kang G, Do A, Moya E, Wilkins L, Le Berichel J, Chang C, Marvin R, Calorossi S, Lansky A, Walker L, Yi N, Yu A, Chung J, Hartnett M, Eaton M, Hatem S, Jamal H, Akyatan A, Tabachnikova A, Liharska LE, Cotter L, Fennessy B, Vaid A, Barturen G, Shah H, Wang YC, Sridhar SH, Soto J, Bose S, Madrid K, Ellis E, Merzier E, Vlachos K, Fishman N, Tin M, Smith M, Xie H, Patel M, Nie K, Argueta K, Harris J, Karekar N, Batchelor C, Lacunza J, Yishak M, Tuballes K, Scott I, Kumar A, Jaladanki S, Agashe C, Thompson R, Clark E, Losic B, Peters L, Roussos P, Zhu J, Wang W, Kasarskis A, Glicksberg BS, Nadkarni G, Bogunovic D, Elaiho C, Gangadharan S, Ofori-Amanfo G, Alesso-Carra K, Onel K, Wilson KM, Argmann C, Bunyavanich S, Alarcón-Riquelme ME, Marron TU, Rahman A, Kim-Schulze S, Gnjatic S, Gelb BD, Merad M, Sebra R, Schadt EE, Charney AW. Downregulation of exhausted cytotoxic T cells in gene expression networks of multisystem inflammatory syndrome in children. Nat Commun 2021; 12:4854. [PMID: 34381049 PMCID: PMC8357784 DOI: 10.1038/s41467-021-24981-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) presents with fever, inflammation and pathology of multiple organs in individuals under 21 years of age in the weeks following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although an autoimmune pathogenesis has been proposed, the genes, pathways and cell types causal to this new disease remain unknown. Here we perform RNA sequencing of blood from patients with MIS-C and controls to find disease-associated genes clustered in a co-expression module annotated to CD56dimCD57+ natural killer (NK) cells and exhausted CD8+ T cells. A similar transcriptome signature is replicated in an independent cohort of Kawasaki disease (KD), the related condition after which MIS-C was initially named. Probing a probabilistic causal network previously constructed from over 1,000 blood transcriptomes both validates the structure of this module and reveals nine key regulators, including TBX21, a central coordinator of exhausted CD8+ T cell differentiation. Together, this unbiased, transcriptome-wide survey implicates downregulation of NK cells and cytotoxic T cell exhaustion in the pathogenesis of MIS-C.
Collapse
Affiliation(s)
- Noam D Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA.
| | - Phillip H Comella
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Lepow
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aviva G Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott R Tyler
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Konstantinos Mouskas
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicole W Simons
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nancy J Francoeur
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | | | - Gurpawan Kang
- Department of Medicine, Division of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anh Do
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Emily Moya
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lillian Wilkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Le Berichel
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christie Chang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Marvin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sharlene Calorossi
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alona Lansky
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Walker
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nancy Yi
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alex Yu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Chung
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Melody Eaton
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sandra Hatem
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hajra Jamal
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alara Akyatan
- Department of of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra Tabachnikova
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lora E Liharska
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Liam Cotter
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Fennessy
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Akhil Vaid
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guillermo Barturen
- Department of Medical Genomics, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government (GENYO), Granada, Spain
| | - Hardik Shah
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ying-Chih Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shwetha Hara Sridhar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juan Soto
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Swaroop Bose
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Kent Madrid
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Ethan Ellis
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Elyze Merzier
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Konstantinos Vlachos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Nataly Fishman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Manying Tin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Melissa Smith
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Hui Xie
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manishkumar Patel
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kimberly Argueta
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jocelyn Harris
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neha Karekar
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Craig Batchelor
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jose Lacunza
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mahlet Yishak
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kevin Tuballes
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ieisha Scott
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arvind Kumar
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Suraj Jaladanki
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charuta Agashe
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ryan Thompson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
| | - Evan Clark
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bojan Losic
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Peters
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panagiotis Roussos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jun Zhu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenhui Wang
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Benjamin S Glicksberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish Nadkarni
- Mount Sinai COVID Informatics Center, New York, NY, USA
- Department of Medicine, Mount Sinai, New York, NY, USA
- Hasso Plattner Institute for Digital Health at Mount Sinai, New York, NY, USA
- Charles Bronfman Institute for Personalized Medicine, New York, NY, USA
| | - Dusan Bogunovic
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cordelia Elaiho
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sandeep Gangadharan
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Ofori-Amanfo
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kasey Alesso-Carra
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenan Onel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karen M Wilson
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marta E Alarcón-Riquelme
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas U Marron
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adeeb Rahman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bruce D Gelb
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA
- Black Family Stem Cell Institute, New York, NY, USA
- Sema4, a Mount Sinai Venture, Stamford, CT, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA.
- Sema4, a Mount Sinai Venture, Stamford, CT, USA.
| | - Alexander W Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute of Data Science and Genomics Technology, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mount Sinai COVID Informatics Center, New York, NY, USA.
| |
Collapse
|
137
|
Fullard JF, Lee HC, Voloudakis G, Suo S, Javidfar B, Shao Z, Peter C, Zhang W, Jiang S, Corvelo A, Wargnier H, Woodoff-Leith E, Purohit DP, Ahuja S, Tsankova NM, Jette N, Hoffman GE, Akbarian S, Fowkes M, Crary JF, Yuan GC, Roussos P. Single-nucleus transcriptome analysis of human brain immune response in patients with severe COVID-19. Genome Med 2021; 13:118. [PMID: 34281603 PMCID: PMC8287557 DOI: 10.1186/s13073-021-00933-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/06/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, has been associated with neurological and neuropsychiatric illness in many individuals. We sought to further our understanding of the relationship between brain tropism, neuro-inflammation, and host immune response in acute COVID-19 cases. METHODS Three brain regions (dorsolateral prefrontal cortex, medulla oblongata, and choroid plexus) from 5 patients with severe COVID-19 and 4 controls were examined. The presence of the virus was assessed by western blot against viral spike protein, as well as viral transcriptome analysis covering > 99% of SARS-CoV-2 genome and all potential serotypes. Droplet-based single-nucleus RNA sequencing (snRNA-seq) was performed in the same samples to examine the impact of COVID-19 on transcription in individual cells of the brain. RESULTS Quantification of viral spike S1 protein and viral transcripts did not detect SARS-CoV-2 in the postmortem brain tissue. However, analysis of 68,557 single-nucleus transcriptomes from three distinct regions of the brain identified an increased proportion of stromal cells, monocytes, and macrophages in the choroid plexus of COVID-19 patients. Furthermore, differential gene expression, pseudo-temporal trajectory, and gene regulatory network analyses revealed transcriptional changes in the cortical microglia associated with a range of biological processes, including cellular activation, mobility, and phagocytosis. CONCLUSIONS Despite the absence of detectable SARS-CoV-2 in the brain at the time of death, the findings suggest significant and persistent neuroinflammation in patients with acute COVID-19.
Collapse
Affiliation(s)
- John F Fullard
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Hao-Chih Lee
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Georgios Voloudakis
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Shengbao Suo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Behnam Javidfar
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Zhiping Shao
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Cyril Peter
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Wen Zhang
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Shan Jiang
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | | | - Heather Wargnier
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Emma Woodoff-Leith
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Dushyant P Purohit
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Sadhna Ahuja
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Nadejda M Tsankova
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Nathalie Jette
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Gabriel E Hoffman
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Mary Fowkes
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - John F Crary
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Panos Roussos
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
- Mental Illness Research Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, 10468, NY, USA.
| |
Collapse
|
138
|
Panici B, Nakajima H, Carlston CM, Ozadam H, Cenik C, Cenik ES. Loss of coordinated expression between ribosomal and mitochondrial genes revealed by comprehensive characterization of a large family with a rare Mendelian disorder. Genomics 2021; 113:1895-1905. [PMID: 33862179 PMCID: PMC8266734 DOI: 10.1016/j.ygeno.2021.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/30/2021] [Accepted: 04/11/2021] [Indexed: 10/21/2022]
Abstract
Non-canonical intronic variants are a poorly characterized yet highly prevalent class of alterations associated with Mendelian disorders. Here, we report the first RNA expression and splicing analysis from a family whose members carry a non-canonical splice variant in an intron of RPL11 (c.396 +3A>G). This mutation is causative for Diamond Blackfan Anemia (DBA) in this family despite incomplete penetrance and variable expressivity. Our analyses revealed a complex pattern of disruptions with many novel junctions of RPL11. These include an RPL11 transcript that is translated with a late stop codon in the 3' untranslated region (3'UTR) of the main isoform. We observed that RPL11 transcript abundance is comparable among carriers regardless of symptom severity. Interestingly, both the small and large ribosomal subunit transcripts were significantly overexpressed in individuals with a history of anemia in addition to congenital abnormalities. Finally, we discovered that coordinated expression between mitochondrial components and RPL11 was lost in all carriers, which may lead to variable expressivity. Overall, this study highlights the importance of RNA splicing and expression analyses in families for molecular characterization of Mendelian diseases.
Collapse
Affiliation(s)
- Brendan Panici
- Department of Molecular Biosciences, University of Texas at Austin, Austin, USA.
| | - Hosei Nakajima
- Department of Molecular Biosciences, University of Texas at Austin, Austin, USA
| | | | - Hakan Ozadam
- Department of Molecular Biosciences, University of Texas at Austin, Austin, USA.
| | - Can Cenik
- Department of Molecular Biosciences, University of Texas at Austin, Austin, USA.
| | - Elif Sarinay Cenik
- Department of Molecular Biosciences, University of Texas at Austin, Austin, USA.
| |
Collapse
|
139
|
Voloudakis G, Hoffman G, Venkatesh S, Lee KM, Dobrindt K, Vicari JM, Zhang W, Beckmann ND, Jiang S, Hoagland D, Bian J, Gao L, Corvelo A, Cho K, Lee JS, Iyengar SK, Luoh SW, Akbarian S, Striker R, Assimes TL, Schadt EE, Merad M, tenOever BR, Charney AW, Brennand KJ, Lynch JA, Fullard JF, Roussos P. IL10RB as a key regulator of COVID-19 host susceptibility and severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.05.31.21254851. [PMID: 34100031 PMCID: PMC8183086 DOI: 10.1101/2021.05.31.21254851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Recent efforts have identified genetic loci that are associated with coronavirus disease 2019 (COVID-19) infection rates and disease outcome severity. Translating these genetic findings into druggable genes and readily available compounds that reduce COVID-19 host susceptibility is a critical next step. METHODS We integrate COVID-19 genetic susceptibility variants, multi-tissue genetically regulated gene expression (GReX) and perturbargen signatures to identify candidate genes and compounds that reverse the predicted gene expression dysregulation associated with COVID-19 susceptibility. The top candidate gene is validated by testing both its GReX and observed blood transcriptome association with COVID-19 severity, as well as by in vitro perturbation to quantify effects on viral load and molecular pathway dysregulation. We validate the in silico drug repositioning analysis by examining whether the top candidate compounds decrease COVID-19 incidence based on epidemiological evidence. RESULTS We identify IL10RB as the top key regulator of COVID-19 host susceptibility. Predicted GReX up-regulation of IL10RB and higher IL10RB expression in COVID-19 patient blood is associated with worse COVID-19 outcomes. In vitro IL10RB overexpression is associated with increased viral load and activation of immune-related molecular pathways. Azathioprine and retinol are prioritized as candidate compounds to reduce the likelihood of testing positive for COVID-19. CONCLUSIONS We establish an integrative data-driven approach for gene target prioritization. We identify and validate IL10RB as a suitable molecular target for modulation of COVID-19 host susceptibility. Finally, we provide evidence for a few readily available medications that would warrant further investigation as drug repositioning candidates.
Collapse
Affiliation(s)
- Georgios Voloudakis
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Gabriel Hoffman
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Sanan Venkatesh
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Kyung Min Lee
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Kristina Dobrindt
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - James M Vicari
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Wen Zhang
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Noam D Beckmann
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Shan Jiang
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Daisy Hoagland
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Jiantao Bian
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Lina Gao
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - André Corvelo
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Kelly Cho
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Jennifer S Lee
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Sudha K Iyengar
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Shiuh-Wen Luoh
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Schahram Akbarian
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Robert Striker
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Themistocles L Assimes
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Eric E Schadt
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Miriam Merad
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Benjamin R tenOever
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Alexander W Charney
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Kristen J Brennand
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Julie A Lynch
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - John F Fullard
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| | - Panos Roussos
- Department of Psychiatry (G.V., S.V., K.D., J.V., W.Z., S.J., S.A., A.W.C., K.J.B., P.R.), Pamela Sklar Division of Psychiatric Genomics (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., A.W.C., J. F.F., K.J.B., P.R.), Friedman Brain Institute (G.V., G.H., S.V., K.D., J.V., W.Z., S.J., S.A., J.F.F., K.J.B., P.R.), Department of Genetics and Genomic Science (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A., E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Icahn Institute for Data Science and Genomic Technology (G.V., G.H., S.V., K.D., J.V., W.Z., N.D.B., S.J., S.A, E.E.S., A.W.C., J.F.F., K.J.B., P.R.), Nash Family Department of Neuroscience (K.D., S.A, A.W.C., K.J.B.), Department of Microbiology (D.H., B.R.T.), Virus Engineering Center for Therapeutics and Research (D.H., B.R.T.), Global Health and Emerging Pathogens Institute (D.H., B.R.T.), Mount Sinai Clinical Intelligence Center (N.D.B., A.W.C.), Department of Oncological Sciences (M.M.), Precision Immunology Institute (M.M.) and Tisch Cancer Institute (M.M.) of the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; the Mental Illness Research, Education, and Clinical Center (VISN 2 South) of the James J. Peters VA Medical Center (G.V., S.V., P.R.), Bronx, NY 10468, USA; VA Informatics and Computing Infrastructure (K.M.L., J.B., J.A.L.), VA Salt Lake City Health Care System, Salt Lake City, UT; Department of Psychiatry (K.D., K.J.B.) of the Yale University, New Haven, CT 06511, USA; Division of Epidemiology (J.B., J.A.L.), University of Utah, Salt Lake City, UT 84108, USA; New York Genome Center (A.C.), New York, NY 10013, USA; Biostatistics Shared Resources (L.G.) and Department of Medicine (S.-W.L.), Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97229, USA; VA Boston Healthcare System (K.C.), Boston, MA, USA; Division of Aging (K.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine (J.S.L., T.L.A.), Stanford University School of Medicine, Stanford, CA, 94304, USA; VA Palo Alto Health Care System (J.S.L., T.L.A.), Palo Alto, CA, 94304, USA; Department of Population and Quantitative Health Sciences (S.K.I), and Department of Genetics and Genome Sciences (S.K.I.), School of Medicine, Case Western Reserve University, Cleveland, OH 44106; VA Northeast Ohio Healthcare System (S.K.I.), Cleveland VA Medical Center, Cleveland, OH 44106; VA Portland Health Care System (S.-W.L.), Portland, OR 97239; Division of Infectious Diseases (R.S.), Department of Medicine, University of Wisconsin, Madison, WI; William S. Middleton Memorial Veterans Hospital (R.S.), Madison, WI 53705; and Sema4 (E.E.S.), Stamford, CT 06902, USA
| |
Collapse
|
140
|
Nøhr AK, Lindow M, Forsingdal A, Demharter S, Nielsen T, Buller R, Moltke I, Vitezic M, Albrechtsen A. A large-scale genome-wide gene expression analysis in peripheral blood identifies very few differentially expressed genes related to antidepressant treatment and response in patients with major depressive disorder. Neuropsychopharmacology 2021; 46:1324-1332. [PMID: 33833401 PMCID: PMC8134553 DOI: 10.1038/s41386-021-01002-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/20/2021] [Accepted: 03/09/2021] [Indexed: 11/08/2022]
Abstract
A better understanding of the biological factors underlying antidepressant treatment in patients with major depressive disorder (MDD) is needed. We perform gene expression analyses and explore sources of variability in peripheral blood related to antidepressant treatment and treatment response in patients suffering from recurrent MDD at baseline and after 8 weeks of treatment. The study includes 281 patients, which were randomized to 8 weeks of treatment with vortioxetine (N = 184) or placebo (N = 97). To our knowledge, this is the largest dataset including both gene expression in blood and placebo-controlled treatment response measured by a clinical scale in a randomized clinical trial. We identified three novel genes whose RNA expression levels at baseline and week 8 are significantly (FDR < 0.05) associated with treatment response after 8 weeks of treatment. Among these genes were SOCS3 (FDR = 0.0039) and PROK2 (FDR = 0.0028), which have previously both been linked to depression. Downregulation of these genes was associated with poorer treatment response. We did not identify any genes that were differentially expressed between placebo and vortioxetine groups at week 8 or between baseline and week 8 of treatment. Nor did we replicate any genes identified in previous peripheral blood gene expression studies examining treatment response. Analysis of genome-wide expression variability showed that type of treatment and treatment response explains very little of the variance, a median of <0.0001% and 0.05% in gene expression across all genes, respectively. Given the relatively large size of the study, the limited findings suggest that peripheral blood gene expression might not be the best approach to explore the biological factors underlying antidepressant treatment.
Collapse
Affiliation(s)
- Anne Krogh Nøhr
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen N, Denmark.
- H. Lundbeck A/S, Valby, Copenhagen, Denmark.
| | | | | | | | | | | | - Ida Moltke
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | | | - Anders Albrechtsen
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
141
|
Geanon D, Lee B, Gonzalez‐Kozlova E, Kelly G, Handler D, Upadhyaya B, Leech J, De Real RM, Herbinet M, Magen A, Del Valle D, Charney A, Kim‐Schulze S, Gnjatic S, Merad M, Rahman AH. A streamlined whole blood CyTOF workflow defines a circulating immune cell signature of COVID-19. Cytometry A 2021; 99:446-461. [PMID: 33496367 PMCID: PMC8013522 DOI: 10.1002/cyto.a.24317] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/10/2020] [Accepted: 01/06/2021] [Indexed: 01/21/2023]
Abstract
Mass cytometry (CyTOF) represents one of the most powerful tools in immune phenotyping, allowing high throughput quantification of over 40 parameters at single-cell resolution. However, wide deployment of CyTOF-based immune phenotyping studies are limited by complex experimental workflows and the need for specialized CyTOF equipment and technical expertise. Furthermore, differences in cell isolation and enrichment protocols, antibody reagent preparation, sample staining, and data acquisition protocols can all introduce technical variation that can confound integrative analyses of large data-sets of samples processed across multiple labs. Here, we present a streamlined whole blood CyTOF workflow which addresses many of these sources of experimental variation and facilitates wider adoption of CyTOF immune monitoring across sites with limited technical expertise or sample-processing resources or equipment. Our workflow utilizes commercially available reagents including the Fluidigm MaxPar Direct Immune Profiling Assay (MDIPA), a dry tube 30-marker immunophenotyping panel, and SmartTube Proteomic Stabilizer, which allows for simple and reliable fixation and cryopreservation of whole blood samples. We validate a workflow that allows for streamlined staining of whole blood samples with minimal processing requirements or expertise at the site of sample collection, followed by shipment to a central CyTOF core facility for batched downstream processing and data acquisition. We apply this workflow to characterize 184 whole blood samples collected longitudinally from a cohort of 72 hospitalized COVID-19 patients and healthy controls, highlighting dynamic disease-associated changes in circulating immune cell frequency and phenotype.
Collapse
Affiliation(s)
- Daniel Geanon
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Brian Lee
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Edgar Gonzalez‐Kozlova
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Geoffrey Kelly
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Diana Handler
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Bhaskar Upadhyaya
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - John Leech
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ronaldo M. De Real
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Manon Herbinet
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Assaf Magen
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Diane Del Valle
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Alexander Charney
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Seunghee Kim‐Schulze
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Sacha Gnjatic
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Miriam Merad
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Adeeb H. Rahman
- Human Immune Monitoring CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
142
|
Oh VKS, Li RW. Temporal Dynamic Methods for Bulk RNA-Seq Time Series Data. Genes (Basel) 2021; 12:352. [PMID: 33673721 PMCID: PMC7997275 DOI: 10.3390/genes12030352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Dynamic studies in time course experimental designs and clinical approaches have been widely used by the biomedical community. These applications are particularly relevant in stimuli-response models under environmental conditions, characterization of gradient biological processes in developmental biology, identification of therapeutic effects in clinical trials, disease progressive models, cell-cycle, and circadian periodicity. Despite their feasibility and popularity, sophisticated dynamic methods that are well validated in large-scale comparative studies, in terms of statistical and computational rigor, are less benchmarked, comparing to their static counterparts. To date, a number of novel methods in bulk RNA-Seq data have been developed for the various time-dependent stimuli, circadian rhythms, cell-lineage in differentiation, and disease progression. Here, we comprehensively review a key set of representative dynamic strategies and discuss current issues associated with the detection of dynamically changing genes. We also provide recommendations for future directions for studying non-periodical, periodical time course data, and meta-dynamic datasets.
Collapse
Affiliation(s)
- Vera-Khlara S. Oh
- Animal Genomics and Improvement Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD 20705, USA;
- Department of Computer Science and Statistics, College of Natural Sciences, Jeju National University, Jeju City 63243, Korea
| | - Robert W. Li
- Animal Genomics and Improvement Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD 20705, USA;
| |
Collapse
|
143
|
Crowell HL, Soneson C, Germain PL, Calini D, Collin L, Raposo C, Malhotra D, Robinson MD. muscat detects subpopulation-specific state transitions from multi-sample multi-condition single-cell transcriptomics data. Nat Commun 2020; 11:6077. [PMID: 33257685 PMCID: PMC7705760 DOI: 10.1038/s41467-020-19894-4] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has become an empowering technology to profile the transcriptomes of individual cells on a large scale. Early analyses of differential expression have aimed at identifying differences between subpopulations to identify subpopulation markers. More generally, such methods compare expression levels across sets of cells, thus leading to cross-condition analyses. Given the emergence of replicated multi-condition scRNA-seq datasets, an area of increasing focus is making sample-level inferences, termed here as differential state analysis; however, it is not clear which statistical framework best handles this situation. Here, we surveyed methods to perform cross-condition differential state analyses, including cell-level mixed models and methods based on aggregated pseudobulk data. To evaluate method performance, we developed a flexible simulation that mimics multi-sample scRNA-seq data. We analyzed scRNA-seq data from mouse cortex cells to uncover subpopulation-specific responses to lipopolysaccharide treatment, and provide robust tools for multi-condition analysis within the muscat R package.
Collapse
Affiliation(s)
- Helena L Crowell
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Charlotte Soneson
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
- Friedrich Miescher Institute for Biomedical Research and SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Pierre-Luc Germain
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- D-HEST Institute for Neuroscience, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Daniela Calini
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Ludovic Collin
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Catarina Raposo
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Dheeraj Malhotra
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Mark D Robinson
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.
- SIB Swiss Institute of Bioinformatics, Zurich, Switzerland.
| |
Collapse
|
144
|
Beckmann ND, Comella PH, Cheng E, Lepow L, Beckmann AG, Mouskas K, Simons NW, Hoffman GE, Francoeur NJ, Del Valle DM, Kang G, Moya E, Wilkins L, Le Berichel J, Chang C, Marvin R, Calorossi S, Lansky A, Walker L, Yi N, Yu A, Hartnett M, Eaton M, Hatem S, Jamal H, Akyatan A, Tabachnikova A, Liharska LE, Cotter L, Fennessey B, Vaid A, Barturen G, Tyler SR, Shah H, Wang YC, Sridhar SH, Soto J, Bose S, Madrid K, Ellis E, Merzier E, Vlachos K, Fishman N, Tin M, Smith M, Xie H, Patel M, Argueta K, Harris J, Karekar N, Batchelor C, Lacunza J, Yishak M, Tuballes K, Scott L, Kumar A, Jaladanki S, Thompson R, Clark E, Losic B, Zhu J, Wang W, Kasarskis A, Glicksberg BS, Nadkarni G, Bogunovic D, Elaiho C, Gangadharan S, Ofori-Amanfo G, Alesso-Carra K, Onel K, Wilson KM, Argmann C, Alarcón-Riquelme ME, Marron TU, Rahman A, Kim-Schulze S, Gnjatic S, Gelb BD, Merad M, Sebra R, Schadt EE, Charney AW. Cytotoxic lymphocytes are dysregulated in multisystem inflammatory syndrome in children. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.08.29.20182899. [PMID: 32909006 PMCID: PMC7480058 DOI: 10.1101/2020.08.29.20182899] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) presents with fever, inflammation and multiple organ involvement in individuals under 21 years following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. To identify genes, pathways and cell types driving MIS-C, we sequenced the blood transcriptomes of MIS-C cases, pediatric cases of coronavirus disease 2019, and healthy controls. We define a MIS-C transcriptional signature partially shared with the transcriptional response to SARS-CoV-2 infection and with the signature of Kawasaki disease, a clinically similar condition. By projecting the MIS-C signature onto a co-expression network, we identified disease gene modules and found genes downregulated in MIS-C clustered in a module enriched for the transcriptional signatures of exhausted CD8 + T-cells and CD56 dim CD57 + NK cells. Bayesian network analyses revealed nine key regulators of this module, including TBX21 , a central coordinator of exhausted CD8 + T-cell differentiation. Together, these findings suggest dysregulated cytotoxic lymphocyte response to SARS-Cov-2 infection in MIS-C.
Collapse
Affiliation(s)
- Noam D. Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Phillip H. Comella
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Esther Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lauren Lepow
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Aviva G. Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Konstantinos Mouskas
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicole W. Simons
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gabriel E. Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nancy J. Francoeur
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Diane Marie Del Valle
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gurpawan Kang
- Department of Medicine, division of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Emily Moya
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lillian Wilkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jessica Le Berichel
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Christie Chang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Marvin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sharlene Calorossi
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alona Lansky
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Laura Walker
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nancy Yi
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alex Yu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Matthew Hartnett
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Melody Eaton
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sandra Hatem
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hajra Jamal
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alara Akyatan
- Department of of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexandra Tabachnikova
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lora E. Liharska
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Liam Cotter
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Brian Fennessey
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Akhil Vaid
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Guillermo Barturen
- Department of Medical Genomics, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government (GENYO), 18007 Urb. los Vergeles, Granada, Spain
| | - Scott R. Tyler
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hardik Shah
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ying-chih Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Shwetha Hara Sridhar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Juan Soto
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Swaroop Bose
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Kent Madrid
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Ethan Ellis
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Elyze Merzier
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Konstantinos Vlachos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Nataly Fishman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Manying Tin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Melissa Smith
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Hui Xie
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Manishkumar Patel
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kimberly Argueta
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jocelyn Harris
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Neha Karekar
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Craig Batchelor
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jose Lacunza
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mahlet Yishak
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kevin Tuballes
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Leisha Scott
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arvind Kumar
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Suraj Jaladanki
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ryan Thompson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
| | - Evan Clark
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bojan Losic
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jun Zhu
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wenhui Wang
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew Kasarskis
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Benjamin S. Glicksberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Girish Nadkarni
- Mount Sinai COVID Informatics Center, New York, NY 10029, USA
- Department of Medicine, Mount Sinai, New York, NY 10029, USA
- Hasso Plattner Institute for Digital Health at Mount Sinai, New York, NY 10029, USA
- Charles Bronfman Institute for Personalized Medicine, New York, NY 10029, USA
| | - Dusan Bogunovic
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Cordelia Elaiho
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sandeep Gangadharan
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - George Ofori-Amanfo
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kasey Alesso-Carra
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenan Onel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karen M. Wilson
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marta E. Alarcón-Riquelme
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Thomas U. Marron
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Adeeb Rahman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Seunghee Kim-Schulze
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bruce D. Gelb
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Departments of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute at Mount Sinai, New York, NY 10029, USA
| | - Miriam Merad
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
- Black Family Stem Cell Institute, New York, NY 10029, USA
- Sema4, a Mount Sinai venture, Stamford CT, 06902, USA
| | - Eric E. Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
- Sema4, a Mount Sinai venture, Stamford CT, 06902, USA
| | - Alexander W. Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Data Science and Genomics Technology, New York, NY 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai COVID Informatics Center, New York, NY 10029, USA
| |
Collapse
|