Sex differences in the genetic predictors of Alzheimer’s pathology

Author:

Dumitrescu Logan12,Barnes Lisa L3,Thambisetty Madhav4,Beecham Gary56,Kunkle Brian6,Bush William S7,Gifford Katherine A1,Chibnik Lori B89,Mukherjee Shubhabrata10,De Jager Philip L1112,Kukull Walter13,Crane Paul K10,Resnick Susan M4,Keene C Dirk14,Montine Thomas J15,Schellenberg Gerard D16,Deming Yuetiva17,Chao Michael J18,Huentelman Matt19,Martin Eden R56,Hamilton-Nelson Kara6,Shaw Leslie M16,Trojanowski John Q16,Peskind Elaine R20,Cruchaga Carlos17,Pericak-Vance Margaret A6,Goate Alison M18,Cox Nancy J2,Haines Jonathan L7,Zetterberg Henrik21222324,Blennow Kaj2122,Larson Eric B1025,Johnson Sterling C26,Albert Marilyn27,Bennett David A3,Schneider Julie A3,Jefferson Angela L1,Hohman Timothy J12,

Affiliation:

1. Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA

2. Vanderbilt Genetics Institute, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA

3. Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA

4. Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA

5. John T MacDonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA

6. John P. Hussman Institute for Human Genomics, University of Miami School of Medicine, Miami, FL, USA

7. Department of Population and Quantitative Health Sciences, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA

8. Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA

9. Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA

10. Department of Medicine, University of Washington, Seattle, WA, USA

11. Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA

12. Cell Circuits Program, Broad Institute, Cambridge MA, USA

13. Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA

14. Department of Pathology, University of Washington, Seattle, WA, USA

15. Department of Pathology, Stanford University, Stanford, CA, USA

16. Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA

17. Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA

18. Ronald M Loeb Center for Alzheimer’s Disease, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA

19. Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA

20. Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA

21. Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden

22. Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden

23. Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK

24. UK Dementia Research Institute at UCL, London, UK

25. Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA

26. Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA

27. Department of Neurology, the Johns Hopkins University School of Medicine, Baltimore, MD

Abstract

Abstract Autopsy measures of Alzheimer’s disease neuropathology have been leveraged as endophenotypes in previous genome-wide association studies (GWAS). However, despite evidence of sex differences in Alzheimer’s disease risk, sex-stratified models have not been incorporated into previous GWAS analyses. We looked for sex-specific genetic associations with Alzheimer’s disease endophenotypes from six brain bank data repositories. The pooled dataset included 2701 males and 3275 females, the majority of whom were diagnosed with Alzheimer’s disease at autopsy (70%). Sex-stratified GWAS were performed within each dataset and then meta-analysed. Loci that reached genome-wide significance (P < 5 × 10−8) in stratified models were further assessed for sex interactions. Additional analyses were performed in independent datasets leveraging cognitive, neuroimaging and CSF endophenotypes, along with age-at-onset data. Outside of the APOE region, one locus on chromosome 7 (rs34331204) showed a sex-specific association with neurofibrillary tangles among males (P = 2.5 × 10−8) but not females (P = 0.85, sex-interaction P = 2.9 × 10−4). In follow-up analyses, rs34331204 was also associated with hippocampal volume, executive function, and age-at-onset only among males. These results implicate a novel locus that confers male-specific protection from tau pathology and highlight the value of assessing genetic associations in a sex-specific manner.

Funder

Alzheimer’s Association

Intramural Research Program

NIA

NIH

Vanderbilt Memory & Alzheimer’s Center

NIMH

Swedish and European Research Councils

UK Dementia Research Institute

Royal Swedish Academy of Sciences

NACC

Publisher

Oxford University Press (OUP)

Subject

Clinical Neurology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3