1
|
Leung YY, Lee W, Kuzma AB, Nicaretta H, Valladares O, Gangadharan P, Qu L, Zhao Y, Ren Y, Cheng P, Kuksa PP, Wang H, White H, Katanic Z, Bass L, Saravanan N, Greenfest‐Allen E, Kirsch M, Cantwell L, Iqbal T, Wheeler NR, Farrell JJ, Zhu C, Turner SL, Gunasekaran TI, Mena PR, Jin Y, Carter L, Alzheimer's Disease Sequencing Project, Zhang X, Vardarajan BN, Toga A, Cuccaro M, Hohman TJ, Bush WS, Naj AC, Martin E, Dalgard CL, Kunkle BW, Farrer LA, Mayeux RP, Haines JL, Pericak‐Vance MA, Schellenberg GD, Wang L. Alzheimer's Disease Sequencing Project release 4 whole genome sequencing dataset. Alzheimers Dement 2025; 21:e70237. [PMID: 40407102 PMCID: PMC12100500 DOI: 10.1002/alz.70237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 05/26/2025]
Abstract
INTRODUCTION The Alzheimer's Disease Sequencing Project (ADSP) is a national initiative to understand the genetic architecture of Alzheimer's disease and related dementias (ADRD) by integrating whole genome sequencing (WGS) with other genetic, phenotypic, and harmonized datasets from diverse populations. METHODS The Genome Center for Alzheimer's Disease (GCAD) uniformly processed WGS from 36,361 ADSP samples, including 35,014 genetically unique participants of which 45% are from non-European ancestry, across 17 cohorts in 14 countries in this fourth release (R4). RESULTS This sequencing effort identified 387 million bi-allelic variants, 42 million short insertions/deletions, and 6.8 million structural variants. Annotations and quality control data are available for all variants and samples. Additionally, detailed phenotypes from 15,927 participants across 10 domains are also provided. A linkage disequilibrium panel was created using unrelated AD cases and controls. DISCUSSION Researchers can access and analyze the genetic data via the National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site (NIAGADS) Data Sharing Service, the VariXam, or NIAGADS GenomicsDB. HIGHLIGHTS We detailed the genetic architecture and quality of the Alzheimer's Disease Sequencing Project release 4 whole genome sequences. We identified 435 million single nucleotide polymorphisms, insertions and deletions, and structural variants from diverse genomes. We harmonized extensive phenotypes, linkage disequilibrium reference panel on subset of samples. Data is publicly available at NIAGADS Data Storage Site, variants and annotations are browsable on two different websites.
Collapse
Grants
- HHSN268201100012C NHLBI NIH HHS
- Alzheimer's Therapeutic Research Institute
- P30 AG10161 Religious Orders Study
- NS039764 Mayo Parkinson's Disease
- P20 AG068082 NIA NIH HHS
- R01AG064614 Alzheimer's Therapeutic Researc Institute
- U01AG052410 Alzheimer's Therapeutic Researc Institute
- R01 AG042188 NIA NIH HHS
- P30 AG066444 NIA NIH HHS
- RC2 HL102419 NHLBI NIH HHS
- Austrian Stroke Prevention Study
- P30 AG013854 NIA NIH HHS
- P30 AG072975 NIA NIH HHS
- R37AG015473 Alzheimer's Disease Centers
- Alzheimer's Disease Sequencing Project (ADSP)
- Welfare and Sports
- U01AG057659 The Longitudinal Aging Study in India
- RC2AG036528 Arizona Department of Health Services
- R01AG057777 Darrell K Royal Texas Alzheimer's Initiative
- HHSN268201100009I NHLBI NIH HHS
- Medical Research Council
- R01 AG018016 NIA NIH HHS
- A2011048 University of Miami
- R01 AG060747 NIA NIH HHS
- 5R37AG015473 Estudio Familiar de Influencia Genetica en Alzheimer
- P01AG003991 Darrell K Royal Texas Alzheimer's Initiative
- R01 AG064877 NIA NIH HHS
- NS071674 Mayo Parkinson's Disease
- U01 AG046152 NIA NIH HHS
- P30 AG010124 NIA NIH HHS
- EuroImmun
- P30 AG072946 NIA NIH HHS
- R01AG051125 The Longitudinal Aging Study in India
- American Genome Center
- R01 NS017950 NINDS NIH HHS
- P30 AG066518 NIA NIH HHS
- R01 AG025711 Darrell K Royal Texas Alzheimer's Initiative
- Netherlands Consortium for Healthy Aging
- RC2 AG036528 NIA NIH HHS
- R01 AG019771 NIA NIH HHS
- P30 AG028377 NIA NIH HHS
- Alzheimer's Association "Identification of Rare Variants in Alzheimer Disease"
- Biogen
- P30 AG066507 NIA NIH HHS
- RF1AG054014 Arizona Department of Health Services
- U01 AG058654 NIA NIH HHS
- AG041718 Arizona Department of Health Services
- U01 HL096812 NHLBI NIH HHS
- R01 AG057777 NIA NIH HHS
- Johnson & Johnson Pharmaceutical Research & Development LLC.
- U54 AG052427 NIA NIH HHS
- Washington University Genome Institute
- R01AG058501 Darrell K Royal Texas Alzheimer's Initiative
- Framingham Heart Study
- R01 AG046949 NIA NIH HHS
- R01 AG042210 NIA NIH HHS
- P50 AG008671 NIA NIH HHS
- R01 AG054076 NIA NIH HHS
- U54 HG003067 NHGRI NIH HHS
- P50 AG005142 NIA NIH HHS
- R01 AG057909 NIA NIH HHS
- R01 AG058501 NIA NIH HHS
- 2014-A-004-NET Hillblom Aging Network
- Ministry of Education
- U01AG062602 Gwangju Alzheimer and Related Dementias Study
- HHSN268201100010C NHLBI NIH HHS
- RF1 AG057440 NIA NIH HHS
- European Special Populations Research Network
- R01AG044829 Longevity Genes Project
- U01 AG058922 NIA NIH HHS
- P20 AG068053 NIA NIH HHS
- R01 AG009029 NIA NIH HHS
- K08 AG034290 NIA NIH HHS
- R01 AG015928 NIA NIH HHS
- R01 AG044546 NIA NIH HHS
- P01 NS026630 NINDS NIH HHS
- P30 AG010133 NIA NIH HHS
- U24 AG021886 NIA NIH HHS
- Netherlands Organization for Scientific Research
- R01 AG009956 NIA NIH HHS
- HHSN268201100008C NHLBI NIH HHS
- U01 HL080295 NHLBI NIH HHS
- P30 AG72975 Religious Orders Study
- IIRG09133827 University of Miami
- R01NS29993 Northern Manhattan Study
- Culture and Science
- 2R01AG09029 Multi-Institutional Research in Alzheimer's Genetic Epidemiology
- 5R37AG015473 Alzheimer's Therapeutic Researc Institute
- Victorian Forensic Institute of Medicine
- P50 AG005131 NIA NIH HHS
- P30AG038072 Longevity Genes Project
- R01 NS069719 NINDS NIH HHS
- U24AG056270 Alzheimer's Disease Centers
- CIHR
- NU38CK000480 Case Western Reserve University Rapid Decline
- UF1AG047133 NIH HHS
- CurePSP Foundation
- HHSN268201500001C NHLBI NIH HHS
- Reta Lila Weston Institute for Neurological Studies
- P50 AG005146 NIA NIH HHS
- Atherosclerosis Risk in Communities
- HHSN268201100005G NHLBI NIH HHS
- RF1AG053303 Alzheimer's Therapeutic Researc Institute
- K25 AG041906 NIA NIH HHS
- P30 AG066512 NIA NIH HHS
- U01 AG049507 NIA NIH HHS
- U01 HL096917 NHLBI NIH HHS
- P50 AG016574 NIA NIH HHS
- R01AG025259 Multi-Institutional Research in Alzheimer's Genetic Epidemiology
- P30 AG066511 NIA NIH HHS
- RF1AG053303 Darrell K Royal Texas Alzheimer's Initiative
- U24 AG072122 NIA NIH HHS
- Erasmus University
- R01 AG11101 Arizona Department of Health Services
- P01 AG017586 NIA NIH HHS
- RF1 AG054014 NIA NIH HHS
- HHSN268201100008I NHLBI NIH HHS
- R01 AG15819 Religious Orders Study
- European Commission
- Alzheimer's Drug Discovery Foundation
- RF1AG058501 Darrell K Royal Texas Alzheimer's Initiative
- P50 AG05681 Darrell K Royal Texas Alzheimer's Initiative
- U01 AG052411 NIA NIH HHS
- R01AG064614 Darrell K Royal Texas Alzheimer's Initiative
- R01 AG019085 NIA NIH HHS
- Genetic and Environmental Risk Factors
- P30 AG066515 NIA NIH HHS
- RF1 AG053303 NIA NIH HHS
- U01 HL130114 NHLBI NIH HHS
- R01 AG017216 Darrell K Royal Texas Alzheimer's Initiative
- R01 AG11101 Chicago Health and Aging Project
- U01 AG032984 NIA NIH HHS
- R01AG032289 Hillblom Aging Network
- P30 AG062421 NIA NIH HHS
- R01 AG030146 NIA NIH HHS
- U01 AG046170 NIA NIH HHS
- R01 AG057907 NIA NIH HHS
- R01 AG09029 Multi-Institutional Research in Alzheimer's Genetic Epidemiology Study
- U01 AG024904 NIA NIH HHS
- European Research Council
- P50 AG008702 NIA NIH HHS
- Lumosity
- R01AG028786 University of Miami Brain Endowment Bank
- HHSN268201100007C NHLBI NIH HHS
- HHSN268200800007C NHLBI NIH HHS
- R01 AG048234 NIA NIH HHS
- RF1AG015473 Estudio Familiar de Influencia Genetica en Alzheimer
- Ministry for Health
- U01AG065958 The Longitudinal Aging Study in India
- Piramal Imaging
- R01 AG043617 NIA NIH HHS
- Takeda Pharmaceutical Company
- U01 AG068057 NIA NIH HHS
- W81XWH-12-2-0012 National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site
- Hussman Institute for Human Genomics
- U54HG003067 Broad Institute Genome Center
- P01AG017586 Tau Consortium
- HHSN268201100011I NHLBI NIH HHS
- QLG2-CT-2002- 01254 Quality of Life and Management of the Living Resources
- HHSN268201100011C NHLBI NIH HHS
- Alzheimer's Association
- RF1 AG054074 NIA NIH HHS
- Erasmus Rucphen Family Study
- AG064877 Arizona Department of Health Services
- U01 AG016976 NIA NIH HHS
- U54AG052427 Genome Center for Alzheimer's Disease
- P50 NS039764 NINDS NIH HHS
- P30AG066462 Darrell K Royal Texas Alzheimer's Initiative
- P30 AG066508 NIA NIH HHS
- R01 AG18023 Arizona Department of Health Services
- Genentech, Inc.
- R01 AG003949 Darrell K Royal Texas Alzheimer's Initiative
- P50 AG005681 NIA NIH HHS
- P01 AG003991 NIA NIH HHS
- Study Investigators institutions
- R56AG051876 Alzheimer's Therapeutic Researc Institute
- R01AG041797 University of Washington Families
- U24 AG056270 NIA NIH HHS
- P30 AG072978 NIA NIH HHS
- HEALTH-F4- 2007-201413 European Community's Seventh Framework
- P01 AG026276 NIA NIH HHS
- R01 AG017917 NIA NIH HHS
- RC2 HG005605 NHGRI NIH HHS
- U24 AG21886 The Longitudinal Aging Study in India
- R01AG046949 Longevity Genes Project
- RF1 AG058501 NIA NIH HHS
- P30 AG062429 NIA NIH HHS
- P01 AG03991 Darrell K Royal Texas Alzheimer's Initiative
- P30 AG19610 MND Victoria
- U01 HL096902 NHLBI NIH HHS
- P30 AG013846 NIA NIH HHS
- Rotterdam
- Netherlands Organization for Scientific Research and the Russian Foundation for Basic Research
- U01AG032984 Transition Therapeutics
- R56 AG051876 NIA NIH HHS
- U54NS100693 Arizona Department of Health Services
- R01AG064877 Darrell K Royal Texas Alzheimer's Initiative
- R01AG054047 Wisconsin Registry for Alzheimer's Prevention
- R01 AG028786 NIA NIH HHS
- Anniversary Fund
- N01HC55222 NHLBI NIH HHS
- U54 HG003273 NHGRI NIH HHS
- R01 AG049607 NIA NIH HHS
- Araclon Biotech
- P30 AG066519 NIA NIH HHS
- UF1 AG047133 NIA NIH HHS
- U01 AG057659 NIA NIH HHS
- KL2 RR024151 NCRR NIH HHS
- Medical University of Graz
- U24 AG074855 NIA NIH HHS
- R01 AG061155 NIA NIH HHS
- Novartis Pharmaceuticals Corporation
- P50 AG005136 NIA NIH HHS
- R01AG025259 Alzheimer Disease Among African Americans Study
- Meso Scale Diagnostics, LLC.
- CereSpir, Inc.
- P30 AG012300 NIA NIH HHS
- NU38 CK000480 NCEZID CDC HHS
- R01 AG42210 Religious Orders Study
- Northern California Institute for Research and Education
- Center for Genome Technology
- N01HC85086 NHLBI NIH HHS
- BioClinica, Inc.
- R01 AG027161 NIA NIH HHS
- RF1 AG054023 NIA NIH HHS
- R01 AG054047 NIA NIH HHS
- 5R01AG009956 Ibadan Study of Aging
- Meso Scale Diagnostics, LLC
- R01AG060747 Religious Orders Study
- U24AG041689 The Longitudinal Aging Study in India
- P30 AG072973 NIA NIH HHS
- U54AG052427 CurePSP
- P30 AG062422 NIA NIH HHS
- RF1 AG054080 NIA NIH HHS
- U19 AG024904 NIA NIH HHS
- U01 AG062943 NIA NIH HHS
- RF1 AG051504 NIA NIH HHS
- GE Healthcare
- P50 AG016573 NIA NIH HHS
- GHR Foundation
- R01 HL105756 NHLBI NIH HHS
- R01 AG079280 NIA NIH HHS
- Large Scale Sequencing and Analysis Centers
- K25 AG041906-01 Arizona Department of Health Services
- Research Institute for Diseases in the Elderly
- Austrian Science Fond
- RF1AG058501 Alzheimer's Disease Centers
- P30AG066444 Alzheimer's Therapeutic Researc Institute
- U.S. Department of Health and Human Services
- R01AG031272 Cache County Study
- Cardiovascular Health Study
- Medical Research Council UK
- RF1 AG058066 NIA NIH HHS
- Parkinson's Victoria
- R01AG042188 Longevity Genes Project
- Departments of Neurology and Psychiatry at Washington University School of Medicine
- 3U01AG052410 CubanAmerican Alzheimer's Disease Initiative
- U24 AG041689 NIA NIH HHS
- U01AG052410 University of Miami Brain Endowment Bank
- P50 AG005134 NIA NIH HHS
- U01 AG006781 NIH HHS
- RF1AG054080 Darrell K Royal Texas Alzheimer's Initiative
- P30 AG008017 NIA NIH HHS
- HHSN268201100006C NHLBI NIH HHS
- R01AG044546 Alzheimer's Therapeutic Researc Institute
- R01 AG044829 NIA NIH HHS
- P30 AG066462 NIA NIH HHS
- R01NS069719 University of Washington Families
- U54AG052427 The Longitudinal Aging Study in India
- R01AG027161 Wisconsin Registry for Alzheimer's Prevention
- P30 AG010161 NIA NIH HHS
- P30 AG066530 NIA NIH HHS
- R01 AG033193 NIA NIH HHS
- R01AG057909 Longevity Genes Project
- Austrian National Bank
- HHSN268201200036C NHLBI NIH HHS
- Victorian Brain Bank
- R01 AG036042 NIA NIH HHS
- U01 AG058589 NIA NIH HHS
- P50 AG025688 NIA NIH HHS
- HHSN268201100005I NHLBI NIH HHS
- R01 AG032990 NIA NIH HHS
- Netherlands Organization
- AbbVie
- 2R01AG048927 Multi-Institutional Research in Alzheimer's Genetic Epidemiology
- U01AG052410 Research in African American Alzheimer Disease Initiative
- P50AG008012 Case Western Reserve University Brain Bank
- Austrian Science Fund
- R37 AG015473 NIA NIH HHS
- R01AG21136 Cache County Study
- U01 NS041588 NINDS NIH HHS
- U01AG057659 Uniformed Services University of the Health Sciences
- R01 NS080820 NINDS NIH HHS
- Department of Internal Medicine
- R01 AG059716 NIA NIH HHS
- HHSN268201500001I NHLBI NIH HHS
- P01AG03991 Alzheimer's Therapeutic Researc Institute
- U01 AG049508 NIA NIH HHS
- U01AG052410 Darrell K Royal Texas Alzheimer's Initiative
- P50 AG008012 NIA NIH HHS
- P01AG03991 Darrell K Royal Texas Alzheimer's Initiative
- P50 NS072187 NINDS NIH HHS
- U24AG041689 National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site
- R01AG046170 Arizona Department of Health Services
- U01AG058922 Alzheimer's Therapeutic Researc Institute
- R01 AG15819 Arizona Department of Health Services
- Avid and Cogstate
- R01AG018016 Mexican Health and Aging Study
- RF1AG015473 Alzheimer's Therapeutic Researc Institute
- U01 HL096814 NHLBI NIH HHS
- Mayo Clinic Florida
- U01AG032984 Arizona Department of Health Services
- RC2 AG036547 NIA NIH HHS
- Steiermärkische Krankenanstalten Gesellschaft
- U24AG041689 Transition Therapeutics
- U01AG058922 Alzheimer's Disease Centers
- Cogstate
- U01 AG052410 NIA NIH HHS
- 5RC2HG005605 Mayo Parkinson's Disease
- P30 AG066509 NIA NIH HHS
- Erasmus MC
- U01 AG006781 NIA NIH HHS
- R01 AG041797 NIA NIH HHS
- EU Joint Programme - Neurodegenerative Disease Research
- NIBIB NIH HHS
- K23 AG030944 NIA NIH HHS
- U54NS100693 CurePSP
- P20 AG068077 NIA NIH HHS
- U01 AG062602 NIA NIH HHS
- R01 NS029993 NINDS NIH HHS
- R01AG044546 Darrell K Royal Texas Alzheimer's Initiative
- Johnson & Johnson Pharmaceutical Research & Development LLC
- RC4 AG039085 NIA NIH HHS
- Reasons for Geographic and Racial Differences in Stroke
- HHSN271201300031C NIDA NIH HHS
- P30 AG066546 NIA NIH HHS
- P30AG066444 Darrell K Royal Texas Alzheimer's Initiative
- R01 AG064614 NIA NIH HHS
- BRIDGET
- P30 AG079280 Alzheimer's Disease Research Centers
- P30 AG038072 NIA NIH HHS
- Erasmus Medical Center
- R01 AG032289 NIA NIH HHS
- R01 AG048927 NIA NIH HHS
- U54 HG003079 NHGRI NIH HHS
- R01 AG019757 NIA NIH HHS
- U01 AG052409 NIA NIH HHS
- U01 AG046139 NIA NIH HHS
- R01 AG033040 NIA NIH HHS
- RF1 AG054052 NIA NIH HHS
- R01 AG007584 Genetic Differences
- R01 AG021547 NIA NIH HHS
- RF1AG054080 Alzheimer's Therapeutic Researc Institute
- R01AG11380 Cache County Study
- U01 AG006576 Darrell K Royal Texas Alzheimer's Initiative
- R01 AG051125 NIA NIH HHS
- Accelerating Medicines Partnership
- R56 AG064877 NIA NIH HHS
- National Institutes of Health-National Institute on Aging
- Elan Pharmaceuticals, Inc.
- P30 AG072977 NIA NIH HHS
- R01AG064877 Alzheimer's Therapeutic Researc Institute
- R01 AG020098 NIA NIH HHS
- public-private-philanthropic partnership
- P30 AG062677 NIA NIH HHS
- N01HC85082 NHLBI NIH HHS
- Rainwater Charitable Foundation
- RF1AG054074 Puerto Rican Alzheimer Disease Initiative
- Health Research and Development
- R01 HL070825 NHLBI NIH HHS
- N01HC85083 NHLBI NIH HHS
- RF1AG057440 Arizona Department of Health Services
- R01 AG027944 NIA NIH HHS
- HHSN268201100005C NHLBI NIH HHS
- P30 AG072958 NIA NIH HHS
- R01 AG025259 NIA NIH HHS
- R01 AG030653 NIA NIH HHS
- HHSN268201100009C NHLBI NIH HHS
- P20 AG068024 NIA NIH HHS
- Eli Lilly and Company
- P01 AG003949 NIA NIH HHS
- RF1 AG057519 NIA NIH HHS
- U01 HL096899 NHLBI NIH HHS
- P30 AG062715 NIA NIH HHS
- RF1 AG015473 NIA NIH HHS
- P50AG005136 University of Washington Families
- U01AG058922 Darrell K Royal Texas Alzheimer's Initiative
- Prospective Dementia Registry-Austria
- P30 AG072976 NIA NIH HHS
- P30 AG010129 NIA NIH HHS
- HHSN268201100007I NHLBI NIH HHS
- R01AG064614 The Longitudinal Aging Study in India
- U01 AG049506 NIA NIH HHS
- U24 NS072026 NINDS NIH HHS
- Wellcome Trust
- R01 AG011101 NIA NIH HHS
- R01 AG17917 Arizona Department of Health Services
- U24 AG21886 Arizona Department of Health Services
- P30 AG066506 NIA NIH HHS
- U24-AG041689 University of Pennsylvania
- P30 AG066468 NIA NIH HHS
- R01 AG021136 NIA NIH HHS
- P30 AG019610 NIA NIH HHS
- IXICO Ltd.
- R01 AG041718 NIA NIH HHS
- Arizona Alzheimer's Disease Core Center
- UG3 NS104095 NINDS NIH HHS
- RF1AG058267 Case Western Reserve University Rapid Decline
- P30AG066462 Alzheimer's Therapeutic Researc Institute
- R01AG057907 Arizona Department of Health Services
- R01AG061155 Longevity Genes Project
- Hussman Institute for Human Genomics Brain Bank
- NeuroRx Research
- P30 AG072947 NIA NIH HHS
- P50 AG025711 NIA NIH HHS
- U01 AG058635 NIA NIH HHS
- Longitudinal Evaluation of Amyloid Risk and Neurodegeneration
- P30 AG072931 NIA NIH HHS
- Merck & Co., Inc.
- RF1AG053303 Alzheimer's Disease Centers
- RF1AG058066 Amish Protective Variant
- #NS072187 Morris K. Udall Parkinson's Disease Research Center of Excellence
- P30 AG072972 NIA NIH HHS
- U01 ES017155 NIEHS NIH HHS
- R01 AG30146 Arizona Department of Health Services
- R01 AG023629 NIA NIH HHS
- N01HC85079 NHLBI NIH HHS
- German Center for Neurodegenerative Diseases
- RF1AG054052 Cache County Study
- Janssen Alzheimer Immunotherapy Research & Development, LLC
- RF1AG058501 Alzheimer's Therapeutic Researc Institute
- R01AG064877 Alzheimer's Disease Centers
- P30 AG066514 NIA NIH HHS
- R56AG051876 Estudio Familiar de Influencia Genetica en Alzheimer
- U19 AG066567 NIH HHS
- U54AG052427 Transition Therapeutics
- P30 AG028383 NIA NIH HHS
- U19 AG066567 NIA NIH HHS
- P01 AG017216 NIA NIH HHS
- N01HC85080 NHLBI NIH HHS
- P30 AG072959 NIA NIH HHS
- P30-AG066468 Arizona Department of Health Services
- R01 AG031272 NIA NIH HHS
- Austrian Research Promotion agency
- P01AG026276 Darrell K Royal Texas Alzheimer's Initiative
- R01 AG011380 NIA NIH HHS
- Netherlands Genomics Initiative
- R01AG048234 Hillblom Aging Network
- P01AG026276 Alzheimer's Therapeutic Researc Institute
- P50 NS071674 NINDS NIH HHS
- R01 AG018023 NIA NIH HHS
- Neurotrack Technologies
- R01 AG17917 Memory and Aging Project
- Fujirebio
- AG030653 Arizona Department of Health Services
- Lundbeck
- R01AG044546 Alzheimer's Disease Centers
- University of Toronto (UT)
- U01 AG006786 NIA NIH HHS
- U54 NS100693 NINDS NIH HHS
- 2R01AG09029 Alzheimer Disease Among African Americans Study
- Netherlands Organization of Scientific Research NWO Investments
- RF1AG054074 Peru Alzheimer's Disease Initiative
- Rotterdam Study
- P30 AG072979 NIA NIH HHS
- R01 AG015819 NIA NIH HHS
- R01 AG036836 NIA NIH HHS
- Eisai Inc.
- RF1 AG058267 NIA NIH HHS
- 2R01AG048927 Alzheimer Disease Among African Americans Study
- P30 AG10161 Arizona Department of Health Services
- U19AG024904) Alzheimer's Disease Neuroimaging Initiative
- U54AG052427 Arizona Department of Health Services
- N01HC85081 NHLBI NIH HHS
- National Institute on Aging Alzheimer's Disease Data Storage Site
- U01 AG049505 NIA NIH HHS
- University of Pennsylvania
- National Institute on Aging
- National Human Genome Research Institute
- National Heart, Lung, and Blood Institute
- National Institutes of Health
- Mayo Clinic
- University of Miami
- Case Western Reserve University
- Alzheimer's Association
- Wellcome Trust
- Medical Research Council
- Canadian Institutes of Health Research
- Framingham Heart Study
- Medical University of Graz
- Austrian Science Fund
- EU Joint Programme – Neurodegenerative Disease Research
- Austrian National Bank
- National Institute of Neurological Disorders and Stroke
- Erasmus Medical Center
- Ministry of Education
- European Commission
- Erasmus MC
- U.S. Department of Health and Human Services
- Alzheimer's Disease Neuroimaging Initiative
- European Research Council
- Baylor College of Medicine
- Uniformed Services University of the Health Sciences
- Eli Lilly and Company
- GHR Foundation
- University of Southern California
- National Institute of Biomedical Imaging and Bioengineering
- AbbVie
- Alzheimer's Drug Discovery Foundation
- BioClinica, Inc.
- Biogen
- Genentech, Inc.
- Fujirebio
- GE Healthcare
- Lundbeck
- Merck & Co., Inc.
- Novartis Pharmaceuticals Corporation
- Takeda Pharmaceutical Company
- Northern California Institute for Research and Education
- Arizona Department of Health Services
- Rainwater Charitable Foundation
- CurePSP
- Tau Consortium
- Victorian Brain Bank
- German Center for Neurodegenerative Diseases
Collapse
Affiliation(s)
- Yuk Yee Leung
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Wan‐Ping Lee
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Amanda B. Kuzma
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Heather Nicaretta
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Otto Valladares
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Prabhakaran Gangadharan
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Liming Qu
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Yi Zhao
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Youli Ren
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Po‐Liang Cheng
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Pavel P. Kuksa
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Hui Wang
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Heather White
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Zivadin Katanic
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Lauren Bass
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Naveen Saravanan
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Emily Greenfest‐Allen
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Maureen Kirsch
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Laura Cantwell
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Taha Iqbal
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biostatistics, Epidemiology, and InformaticsUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Nicholas R. Wheeler
- Department of Population and Quantitative Health SciencesCase Western Reserve UniversityClevelandOhioUSA
- Department of Genetics and Genome SciencesSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - John J. Farrell
- Departments of Medicine (Biomedical Genetics)Boston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Congcong Zhu
- Departments of Medicine (Biomedical Genetics)Boston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Shannon L. Turner
- Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Tamil I. Gunasekaran
- Columbia University Irving Medical CenterNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterTaub Institute for Research on the Aging BrainDepartments of Neurology, Psychiatry, and EpidemiologyCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Pedro R. Mena
- Department of Human Genetics and John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Yumi Jin
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Luke Carter
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Xiaoling Zhang
- Departments of Medicine (Biomedical Genetics)Boston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Badri N. Vardarajan
- Columbia University Irving Medical CenterNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterTaub Institute for Research on the Aging BrainDepartments of Neurology, Psychiatry, and EpidemiologyCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Arthur Toga
- Laboratory of Neuro ImagingUSC Stevens Neuroimaging and Informatics InstituteKeck School of Medicine of USCUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Michael Cuccaro
- Department of Human Genetics and John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Timothy J. Hohman
- Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - William S. Bush
- Department of Population and Quantitative Health SciencesCase Western Reserve UniversityClevelandOhioUSA
- Department of Genetics and Genome SciencesSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Adam C. Naj
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biostatistics, Epidemiology, and InformaticsUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Eden Martin
- Department of Human Genetics and John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology and GeneticsSchool of MedicineUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Brian W. Kunkle
- Department of Human Genetics and John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Lindsay A. Farrer
- Departments of Medicine (Biomedical Genetics)Boston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of OphthalmologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| | - Richard P. Mayeux
- Columbia University Irving Medical CenterNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterTaub Institute for Research on the Aging BrainDepartments of Neurology, Psychiatry, and EpidemiologyCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Jonathan L. Haines
- Department of Population and Quantitative Health SciencesCase Western Reserve UniversityClevelandOhioUSA
- Department of Genetics and Genome SciencesSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Margaret A. Pericak‐Vance
- Department of Human Genetics and John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Li‐San Wang
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Leung YY, Lee WP, Kuzma AB, Nicaretta H, Valladares O, Gangadharan P, Qu L, Zhao Y, Ren Y, Cheng PL, Kuksa PP, Wang H, White H, Katanic Z, Bass L, Saravanan N, Greenfest-Allen E, Kirsch M, Cantwell L, Iqbal T, Wheeler NR, Farrell JJ, Zhu C, Turner SL, Gunasekaran TI, Mena PR, Jin J, Carter L, Alzheimer’s Disease Sequencing Project, Zhang X, Vardarajan BN, Toga A, Cuccaro M, Hohman TJ, Bush WS, Naj AC, Martin E, Dalgard C, Kunkle BW, Farrer LA, Mayeux RP, Haines JL, Pericak-Vance MA, Schellenberg GD, Wang LS. Alzheimer's Disease Sequencing Project Release 4 Whole Genome Sequencing Dataset. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.03.24317000. [PMID: 39677464 PMCID: PMC11643159 DOI: 10.1101/2024.12.03.24317000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The Alzheimer's Disease Sequencing Project (ADSP) is a national initiative to understand the genetic architecture of Alzheimer's Disease and Related Dementias (AD/ADRD) by sequencing whole genomes of affected participants and age-matched cognitive controls from diverse populations. The Genome Center for Alzheimer's Disease (GCAD) processed whole-genome sequencing data from 36,361 ADSP participants, including 35,014 genetically unique participants of which 45% are from non-European ancestry, across 17 cohorts in 14 countries in this fourth release (R4). This sequencing effort identified 387 million bi-allelic variants, 42 million short insertions/deletions, and 2.2 million structural variants. Annotations and quality control data are available for all variants and samples. Additionally, detailed phenotypes from 15,927 participants across 10 domains are also provided. A linkage disequilibrium panel was created using unrelated AD cases and controls. Researchers can access and analyze the genetic data via NIAGADS Data Sharing Service, the VariXam tool, or NIAGADS GenomicsDB.
Collapse
Affiliation(s)
- Yuk Yee Leung
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Wan-Ping Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Amanda B Kuzma
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Heather Nicaretta
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Otto Valladares
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Prabhakaran Gangadharan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Liming Qu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Yi Zhao
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Youli Ren
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Po-Liang Cheng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Pavel P Kuksa
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Hui Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Heather White
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Zivadin Katanic
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Lauren Bass
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Naveen Saravanan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Emily Greenfest-Allen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Maureen Kirsch
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Laura Cantwell
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Taha Iqbal
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nicholas R Wheeler
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - John J. Farrell
- Department of Medicine, Biostatistics & Bioinformatics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Congcong Zhu
- Department of Medicine, Biostatistics & Bioinformatics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Shannon L Turner
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tamil I Gunasekaran
- Columbia University Irving Medical Center, New York, NY, USA
- Gertrude H. Sergievsky Center, Taub Institute for Research on the Aging Brain, Departments of Neurology, Psychiatry, and Epidemiology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Pedro R Mena
- Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jimmy Jin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Luke Carter
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | | | - Xiaoling Zhang
- Department of Medicine, Biostatistics & Bioinformatics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Badri N Vardarajan
- Columbia University Irving Medical Center, New York, NY, USA
- Gertrude H. Sergievsky Center, Taub Institute for Research on the Aging Brain, Departments of Neurology, Psychiatry, and Epidemiology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Arthur Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California
| | - Michael Cuccaro
- Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Timothy J Hohman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Adam C Naj
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eden Martin
- Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Clifton Dalgard
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Brian W Kunkle
- Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lindsay A Farrer
- Department of Medicine, Biostatistics & Bioinformatics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Richard P Mayeux
- Columbia University Irving Medical Center, New York, NY, USA
- Gertrude H. Sergievsky Center, Taub Institute for Research on the Aging Brain, Departments of Neurology, Psychiatry, and Epidemiology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Margaret A Pericak-Vance
- Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
3
|
Koskeridis F, Fancy N, Tan PF, Meena D, Evangelou E, Elliott P, Wang D, Matthews PM, Dehghan A, Tzoulaki I. Multi-trait association analysis reveals shared genetic loci between Alzheimer's disease and cardiovascular traits. Nat Commun 2024; 15:9827. [PMID: 39537608 PMCID: PMC11561119 DOI: 10.1038/s41467-024-53452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Several cardiovascular traits and diseases co-occur with Alzheimer's disease. We mapped their shared genetic architecture using multi-trait genome-wide association studies. Subsequent fine-mapping and colocalisation highlighted 16 genetic loci associated with both Alzheimer's and cardiovascular diseases. We prioritised rs11786896, which colocalised with Alzheimer's disease, atrial fibrillation and expression of PLEC in the heart left ventricle, and rs7529220, which colocalised with Alzheimer's disease, atrial fibrillation and expression of C1Q family genes. Single-cell RNA-sequencing data, co-expression network and protein-protein interaction analyses provided evidence for different mechanisms of PLEC, which is upregulated in left ventricular endothelium and cardiomyocytes with heart failure and in brain astrocytes with Alzheimer's disease. Similar common mechanisms are implicated for C1Q in heart macrophages with heart failure and in brain microglia with Alzheimer's disease. These findings highlight inflammatory and pleomorphic risk determinants for the co-occurrence of Alzheimer's and cardiovascular diseases and suggest PLEC, C1Q and their interacting proteins as potential therapeutic targets.
Collapse
Affiliation(s)
- Fotios Koskeridis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece.
- UK Dementia Research Institute, Imperial College London, London, UK.
| | - Nurun Fancy
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Pei Fang Tan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Devendra Meena
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Dennis Wang
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Paul M Matthews
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
- Systems Biology, Biomedical Research Institute of the Academy of Athens, Athens, Greece
| |
Collapse
|
4
|
Ghatak S, Diedrich JK, Talantova M, Bhadra N, Scott H, Sharma M, Albertolle M, Schork NJ, Yates JR, Lipton SA. Single-Cell Patch-Clamp/Proteomics of Human Alzheimer's Disease iPSC-Derived Excitatory Neurons Versus Isogenic Wild-Type Controls Suggests Novel Causation and Therapeutic Targets. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400545. [PMID: 38773714 PMCID: PMC11304297 DOI: 10.1002/advs.202400545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/03/2024] [Indexed: 05/24/2024]
Abstract
Standard single-cell (sc) proteomics of disease states inferred from multicellular organs or organoids cannot currently be related to single-cell physiology. Here, a scPatch-Clamp/Proteomics platform is developed on single neurons generated from hiPSCs bearing an Alzheimer's disease (AD) genetic mutation and compares them to isogenic wild-type controls. This approach provides both current and voltage electrophysiological data plus detailed proteomics information on single-cells. With this new method, the authors are able to observe hyperelectrical activity in the AD hiPSC-neurons, similar to that observed in the human AD brain, and correlate it to ≈1400 proteins detected at the single neuron level. Using linear regression and mediation analyses to explore the relationship between the abundance of individual proteins and the neuron's mutational and electrophysiological status, this approach yields new information on therapeutic targets in excitatory neurons not attainable by traditional methods. This combined patch-proteomics technique creates a new proteogenetic-therapeutic strategy to correlate genotypic alterations to physiology with protein expression in single-cells.
Collapse
Affiliation(s)
- Swagata Ghatak
- Neurodegeneration New Medicines CenterThe Scripps Research InstituteLa JollaCA92037USA
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
- Present address:
School of Biological SciencesNational Institute of Science Education and Research (NISER)‐Bhubaneswar, an OCC of Homi Bhabha National InstituteJataniOdisha752050India
| | - Jolene K. Diedrich
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
| | - Maria Talantova
- Neurodegeneration New Medicines CenterThe Scripps Research InstituteLa JollaCA92037USA
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
| | - Nivedita Bhadra
- Quantitative Medicine and Systems BiologyThe Translational Genomics Research InstitutePhoenixAZ85004USA
| | - Henry Scott
- Neurodegeneration New Medicines CenterThe Scripps Research InstituteLa JollaCA92037USA
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
| | - Meetal Sharma
- Neurodegeneration New Medicines CenterThe Scripps Research InstituteLa JollaCA92037USA
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
| | - Matthew Albertolle
- Neurodegeneration New Medicines CenterThe Scripps Research InstituteLa JollaCA92037USA
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
- Present address:
Drug Metabolism and Pharmacokinetics DepartmentTakeda Development Center AmericasSan DiegoCA92121USA
| | - Nicholas J. Schork
- Quantitative Medicine and Systems BiologyThe Translational Genomics Research InstitutePhoenixAZ85004USA
| | - John R. Yates
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
| | - Stuart A. Lipton
- Neurodegeneration New Medicines CenterThe Scripps Research InstituteLa JollaCA92037USA
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
- Department of NeurosciencesSchool of MedicineUniversity of California, San DiegoLa JollaCA92093USA
| |
Collapse
|
5
|
Chen H, King FJ, Zhou B, Wang Y, Canedy CJ, Hayashi J, Zhong Y, Chang MW, Pache L, Wong JL, Jia Y, Joslin J, Jiang T, Benner C, Chanda SK, Zhou Y. Drug target prediction through deep learning functional representation of gene signatures. Nat Commun 2024; 15:1853. [PMID: 38424040 PMCID: PMC10904399 DOI: 10.1038/s41467-024-46089-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
Many machine learning applications in bioinformatics currently rely on matching gene identities when analyzing input gene signatures and fail to take advantage of preexisting knowledge about gene functions. To further enable comparative analysis of OMICS datasets, including target deconvolution and mechanism of action studies, we develop an approach that represents gene signatures projected onto their biological functions, instead of their identities, similar to how the word2vec technique works in natural language processing. We develop the Functional Representation of Gene Signatures (FRoGS) approach by training a deep learning model and demonstrate that its application to the Broad Institute's L1000 datasets results in more effective compound-target predictions than models based on gene identities alone. By integrating additional pharmacological activity data sources, FRoGS significantly increases the number of high-quality compound-target predictions relative to existing approaches, many of which are supported by in silico and/or experimental evidence. These results underscore the general utility of FRoGS in machine learning-based bioinformatics applications. Prediction networks pre-equipped with the knowledge of gene functions may help uncover new relationships among gene signatures acquired by large-scale OMICs studies on compounds, cell types, disease models, and patient cohorts.
Collapse
Affiliation(s)
- Hao Chen
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA.
- Department of Computer Science and Engineering, University of California, Riverside, 900 University Avenue, Riverside, CA, 92521, USA.
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| | - Frederick J King
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Bin Zhou
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Yu Wang
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Carter J Canedy
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Joel Hayashi
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Yang Zhong
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Max W Chang
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Lars Pache
- NCI Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Julian L Wong
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Yong Jia
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - John Joslin
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Tao Jiang
- Department of Computer Science and Engineering, University of California, Riverside, 900 University Avenue, Riverside, CA, 92521, USA
| | - Christopher Benner
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Sumit K Chanda
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, 92037, USA
| | - Yingyao Zhou
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
6
|
Zhang Z, Liu X, Zhang S, Song Z, Lu K, Yang W. A review and analysis of key biomarkers in Alzheimer's disease. Front Neurosci 2024; 18:1358998. [PMID: 38445255 PMCID: PMC10912539 DOI: 10.3389/fnins.2024.1358998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects over 50 million elderly individuals worldwide. Although the pathogenesis of AD is not fully understood, based on current research, researchers are able to identify potential biomarker genes and proteins that may serve as effective targets against AD. This article aims to present a comprehensive overview of recent advances in AD biomarker identification, with highlights on the use of various algorithms, the exploration of relevant biological processes, and the investigation of shared biomarkers with co-occurring diseases. Additionally, this article includes a statistical analysis of key genes reported in the research literature, and identifies the intersection with AD-related gene sets from databases such as AlzGen, GeneCard, and DisGeNet. For these gene sets, besides enrichment analysis, protein-protein interaction (PPI) networks utilized to identify central genes among the overlapping genes. Enrichment analysis, protein interaction network analysis, and tissue-specific connectedness analysis based on GTEx database performed on multiple groups of overlapping genes. Our work has laid the foundation for a better understanding of the molecular mechanisms of AD and more accurate identification of key AD markers.
Collapse
Affiliation(s)
- Zhihao Zhang
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Xiangtao Liu
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Suixia Zhang
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Zhixin Song
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Ke Lu
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
| | - Wenzhong Yang
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
| |
Collapse
|
7
|
Gaunt JR, Zainolabidin N, Yip AKK, Tan JM, Low AYT, Chen AI, Ch'ng TH. Cytokine enrichment in deep cerebellar nuclei is contributed by multiple glial populations and linked to reduced amyloid plaque pathology. J Neuroinflammation 2023; 20:269. [PMID: 37978387 PMCID: PMC10656954 DOI: 10.1186/s12974-023-02913-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023] Open
Abstract
Alzheimer's disease (AD) pathology and amyloid-beta (Aβ) plaque deposition progress slowly in the cerebellum compared to other brain regions, while the entorhinal cortex (EC) is one of the most vulnerable regions. Using a knock-in AD mouse model (App KI), we show that within the cerebellum, the deep cerebellar nuclei (DCN) has particularly low accumulation of Aβ plaques. To identify factors that might underlie differences in the progression of AD-associated neuropathology across regions, we profiled gene expression in single nuclei (snRNAseq) across all cell types in the DCN and EC of wild-type (WT) and App KI male mice at age 7 months. We found differences in expression of genes associated with inflammatory activation, PI3K-AKT signalling, and neuron support functions between both regions and genotypes. In WT mice, the expression of interferon-response genes in microglia is higher in the DCN than the EC and this enrichment is confirmed by RNA in situ hybridisation, and measurement of inflammatory cytokines by protein array. Our analyses also revealed that multiple glial populations are responsible for establishing this cytokine-enriched niche. Furthermore, homogenates derived from the DCN induced inflammatory gene expression in BV2 microglia. We also assessed the relationship between the DCN microenvironment and Aβ pathology by depleting microglia using a CSF1R inhibitor PLX5622 and saw that, surprisingly, the expression of a subset of inflammatory cytokines was increased while plaque abundance in the DCN was further reduced. Overall, our study revealed the presence of a cytokine-enriched microenvironment unique to the DCN that when modulated, can alter plaque deposition.
Collapse
Affiliation(s)
- Jessica R Gaunt
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Norliyana Zainolabidin
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Alaric K K Yip
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Jia Min Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Aloysius Y T Low
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Albert I Chen
- Center for Aging Research, Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA, 92121, USA.
- Molecular Neurobiology Laboratory, Salk Institute, La Jolla, CA, 92037, USA.
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore.
- School of Biological Science, Nanyang Technological University, Singapore, 63755, Singapore.
| |
Collapse
|
8
|
Essayan-Perez S, Südhof TC. Neuronal γ-secretase regulates lipid metabolism, linking cholesterol to synaptic dysfunction in Alzheimer's disease. Neuron 2023; 111:3176-3194.e7. [PMID: 37543038 PMCID: PMC10592349 DOI: 10.1016/j.neuron.2023.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/16/2023] [Accepted: 07/10/2023] [Indexed: 08/07/2023]
Abstract
Presenilin mutations that alter γ-secretase activity cause familial Alzheimer's disease (AD), whereas ApoE4, an apolipoprotein for cholesterol transport, predisposes to sporadic AD. Both sporadic and familial AD feature synaptic dysfunction. Whether γ-secretase is involved in cholesterol metabolism and whether such involvement impacts synaptic function remains unknown. Here, we show that in human neurons, chronic pharmacological or genetic suppression of γ-secretase increases synapse numbers but decreases synaptic transmission by lowering the presynaptic release probability without altering dendritic or axonal arborizations. In search of a mechanism underlying these synaptic impairments, we discovered that chronic γ-secretase suppression robustly decreases cholesterol levels in neurons but not in glia, which in turn stimulates neuron-specific cholesterol-synthesis gene expression. Suppression of cholesterol levels by HMG-CoA reductase inhibitors (statins) impaired synaptic function similar to γ-secretase inhibition. Thus, γ-secretase enables synaptic function by maintaining cholesterol levels, whereas the chronic suppression of γ-secretase impairs synapses by lowering cholesterol levels.
Collapse
Affiliation(s)
- Sofia Essayan-Perez
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
9
|
Wang S, Fang X, Wen X, Yang C, Yang Y, Zhang T. Prioritization of risk genes for Alzheimer's disease: an analysis framework using spatial and temporal gene expression data in the human brain based on support vector machine. Front Genet 2023; 14:1190863. [PMID: 37867597 PMCID: PMC10587557 DOI: 10.3389/fgene.2023.1190863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023] Open
Abstract
Background: Alzheimer's disease (AD) is a complex disorder, and its risk is influenced by multiple genetic and environmental factors. In this study, an AD risk gene prediction framework based on spatial and temporal features of gene expression data (STGE) was proposed. Methods: We proposed an AD risk gene prediction framework based on spatial and temporal features of gene expression data. The gene expression data of providers of different tissues and ages were used as model features. Human genes were classified as AD risk or non-risk sets based on information extracted from relevant databases. Support vector machine (SVM) models were constructed to capture the expression patterns of genes believed to contribute to the risk of AD. Results: The recursive feature elimination (RFE) method was utilized for feature selection. Data for 64 tissue-age features were obtained before feature selection, and this number was reduced to 19 after RFE was performed. The SVM models were built and evaluated using 19 selected and full features. The area under curve (AUC) values for the SVM model based on 19 selected features (0.740 [0.690-0.790]) and full feature sets (0.730 [0.678-0.769]) were very similar. Fifteen genes predicted to be risk genes for AD with a probability greater than 90% were obtained. Conclusion: The newly proposed framework performed comparably to previous prediction methods based on protein-protein interaction (PPI) network properties. A list of 15 candidate genes for AD risk was also generated to provide data support for further studies on the genetic etiology of AD.
Collapse
Affiliation(s)
- Shiyu Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xixian Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xiang Wen
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Beijing, China
| | - Congying Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Ying Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Tianxiao Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Anti-Drug Laboratory Shaanxi Regional Center, Xi’an, China
| |
Collapse
|
10
|
Hou Y, Song Q, Wang Y, Liu J, Cui Y, Zhang X, Zhang J, Fu J, Cao M, Zhang C, Liu C, Wang X, Duan H, Wang P. Downregulation of Krüppel-like factor 14 accelerated cellular senescence and aging. Aging Cell 2023; 22:e13950. [PMID: 37551728 PMCID: PMC10577553 DOI: 10.1111/acel.13950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/02/2023] [Accepted: 07/17/2023] [Indexed: 08/09/2023] Open
Abstract
Aging has been considered as a risk factor in many diseases, thus, comprehensively understanding the cellular and molecular mechanisms of delayed aging is important. Here we investigated whether Krüppel-like factor 14 (KLF14) is a suppressor of cellular senescence and aging. In our research, KLF14 levels significantly decreased not only in the lymphocytes of healthy people but also in the cells and tissues of mice with aging. We performed in vitro and in vivo experiments on cells and mice to reveal the function of KLF14 in aging. KLF14 deficiency facilitates cellular senescence and aging-related pathologies in C57BL/6J mice, whereas KLF14 overexpression attenuates cellular senescence. Mechanistically, KLF14 delays aging by binding to the POLD1 promoter to positively regulate POLD1 expression. Remarkably, cellular senescence mediated by KLF14 downregulation could be alleviated by POLD1 expression. In addition, perhexiline, an agonist of KLF14, could delay cellular senescence and aging-related pathologies in senescence-accelerated P8 mice by inducing POLD1 expression, as perhexiline could enhance the effect of KLF14's transcription activation to POLD1 by elevating the binding level of KLF14 to the POLD1 promoter. Our data indicate that KLF14 might be a critical element in aging by upregulating POLD1 expression, indicating that the activation of KLF14 may delay aging and aging-associated diseases.
Collapse
Affiliation(s)
- Yuli Hou
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Qiao Song
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Yaqi Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Jing Liu
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Yuting Cui
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Xiaomin Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Jingjing Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Jingxuan Fu
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Min Cao
- Department of Clinical LaboratoryBeijing Huairou HospitalBeijingChina
| | - Chi Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Congcong Liu
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Xiaoling Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Huanli Duan
- Departments of Pathology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Peichang Wang
- Department of Clinical Laboratory, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| |
Collapse
|
11
|
Shapiro D, Lee K, Asmussen J, Bourquard T, Lichtarge O. Evolutionary Action-Machine Learning Model Identifies Candidate Genes Associated With Early-Onset Coronary Artery Disease. J Am Heart Assoc 2023; 12:e029103. [PMID: 37642027 PMCID: PMC10547338 DOI: 10.1161/jaha.122.029103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/11/2023] [Indexed: 08/31/2023]
Abstract
Background Coronary artery disease is a primary cause of death around the world, with both genetic and environmental risk factors. Although genome-wide association studies have linked >100 unique loci to its genetic basis, these only explain a fraction of disease heritability. Methods and Results To find additional gene drivers of coronary artery disease, we applied machine learning to quantitative evolutionary information on the impact of coding variants in whole exomes from the Myocardial Infarction Genetics Consortium. Using ensemble-based supervised learning, the Evolutionary Action-Machine Learning framework ranked each gene's ability to classify case and control samples and identified 79 significant associations. These were connected to known risk loci; enriched in cardiovascular processes like lipid metabolism, blood clotting, and inflammation; and enriched for cardiovascular phenotypes in knockout mouse models. Among them, INPP5F and MST1R are examples of potentially novel coronary artery disease risk genes that modulate immune signaling in response to cardiac stress. Conclusions We concluded that machine learning on the functional impact of coding variants, based on a massive amount of evolutionary information, has the power to suggest novel coronary artery disease risk genes for mechanistic and therapeutic discoveries in cardiovascular biology, and should also apply in other complex polygenic diseases.
Collapse
Affiliation(s)
- Dillon Shapiro
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Kwanghyuk Lee
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Jennifer Asmussen
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Thomas Bourquard
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Olivier Lichtarge
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
- Computational & Integrative Biomedical Research CenterBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
12
|
Catumbela CSG, Giridharan VV, Barichello T, Morales R. Clinical evidence of human pathogens implicated in Alzheimer's disease pathology and the therapeutic efficacy of antimicrobials: an overview. Transl Neurodegener 2023; 12:37. [PMID: 37496074 PMCID: PMC10369764 DOI: 10.1186/s40035-023-00369-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023] Open
Abstract
A wealth of pre-clinical reports and data derived from human subjects and brain autopsies suggest that microbial infections are relevant to Alzheimer's disease (AD). This has inspired the hypothesis that microbial infections increase the risk or even trigger the onset of AD. Multiple models have been developed to explain the increase in pathogenic microbes in AD patients. Although this hypothesis is well accepted in the field, it is not yet clear whether microbial neuroinvasion is a cause of AD or a consequence of the pathological changes experienced by the demented brain. Along the same line, the gut microbiome has also been proposed as a modulator of AD. In this review, we focus on human-based evidence demonstrating the elevated abundance of microbes and microbe-derived molecules in AD hosts as well as their interactions with AD hallmarks. Further, the direct-purpose and potential off-target effects underpinning the efficacy of anti-microbial treatments in AD are also addressed.
Collapse
Affiliation(s)
- Celso S G Catumbela
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Vijayasree V Giridharan
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Rodrigo Morales
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA), Universidad Bernardo O'Higgins, 8370993, Santiago, Chile.
| |
Collapse
|