Skip to main content

Advertisement

Log in

The influence of growth and sex hormones on risk of alzheimer’s disease: a mendelian randomization study

  • NEURO-EPIDEMIOLOGY
  • Published:
European Journal of Epidemiology Aims and scope Submit manuscript

Abstract

Alzheimer’s disease is more prevalent in women, possibly due to sex or growth hormones but existing evidence is inconclusive. We investigated whether genetically predicted sex and growth hormones are associated with risk of Alzheimer’s disease. Genetic variants strongly and independently predicting insulin-like growth factor 1 (IGF-1), testosterone and sex hormone-binding globulin (SHBG) were obtained from large, published genome wide associations studies (GWAS) and applied to GWAS of Alzheimer’s disease based on clinical diagnosis (cases = 21,982, control = 41,944) from the International Genomics of Alzheimer’s Project and the UK Biobank parental (maternal cases = 27,696; paternal cases = 14,338) and siblings’ diagnosis (cases = 2,171) as proxy cases. Published GWAS summary statistics were used in our analyses. Estimates were obtained from inverse variance weighting with sensitivity analysis (i.e., MR-Egger, weighted median and MR-PRESSO). Multivariable analyses adjusted for pleiotropic effects and possible sources of selection bias were also performed. Genetically predicted higher total testosterone may reduce the risk of paternal Alzheimer’s disease (odds ratio (OR) 0.86, 95% confidence interval (CI) 0.76 to 0.97, per SD increase in testosterone) and in meta-analysis for women (OR 0.92, 95% CI 0.87, 0.98) with directionally similar results from other analyses. SHBG were not associated with Alzheimer’s disease. IGF-1 in women was inversely associated with risk of clinical Alzheimer’s disease in sensitivity analysis but not in the main analysis. These results suggest genetically predicted higher total testosterone may lower risk of Alzheimer’s disease. The role of testosterone and the immune system in Alzheimer’s disease could be further investigated.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Data Availability

All data used are publicly available and can be found in the references and supplementary file.

References

  1. Association A. 2019 Alzheimer’s disease facts and figures. Alzheimer’s Dement [Internet]. John Wiley & Sons, Ltd; 2019;15:321–87. Available from: https://doi.org/10.1016/j.jalz.2019.01.010.

  2. Snyder HM, Asthana S, Bain L, Brinton R, Craft S, Dubal DB, et al. Sex biology contributions to vulnerability to Alzheimer’s disease: a think tank convened by the women’s Alzheimer’s Research Initiative. Alzheimers Dement United States. 2016;12:1186–96.

    Article  Google Scholar 

  3. Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA United States. 1997;278:1349–56.

    CAS  Google Scholar 

  4. Hogervorst E, Williams J, Budge M, Barnetson L, Combrinck M, Smith AD. Serum total testosterone is lower in men with Alzheimer’s disease. Neuro Endocrinol Lett Sweden. 2001;22:163–8.

    CAS  Google Scholar 

  5. Vest RS, Pike CJ. Gender, sex steroid hormones, and Alzheimer’s disease. Horm Behav [Internet]. 2013;63:301–7. Available from: https://www.sciencedirect.com/science/article/pii/S0018506X12001134.

  6. Zhang Z, Kang D, Li H. Testosterone and cognitive impairment or dementia in Middle-Aged or aging males: causation and intervention, a systematic review and Meta-analysis. J Geriatr Psychiatry Neurol United States. 2021;34:405–17.

    Article  Google Scholar 

  7. Lei Y, Renyuan Z. Effects of Androgens on the Amyloid-β protein in Alzheimer’s Disease. Endocrinol United States. 2018;159:3885–94.

    CAS  Google Scholar 

  8. Muller M, Schupf N, Manly JJ, Mayeux R, Luchsinger JA. Sex hormone binding globulin and incident Alzheimer’s disease in elderly men and women. Neurobiol Aging United States. 2010;31:1758–65.

    Article  CAS  Google Scholar 

  9. Deijen JB, Arwert LI, Drent ML. The GH/IGF-I Axis and cognitive changes across a 4-Year period in healthy adults. ISRN Endocrinol United States. 2011;2011:249421.

    Google Scholar 

  10. Nashiro K, Guevara-Aguirre J, Braskie MN, Hafzalla GW, Velasco R, Balasubramian P et al. Brain structure and function associated with younger adults in growth hormone receptor deficient humans. J Neurosci [Internet]. 2017;1916–29. Available from: http://www.jneurosci.org/content/early/2017/01/10/JNEUROSCI.1929-16.2016.abstract.

  11. Carro E, Trejo JL, Gomez-Isla T, LeRoith D, Torres-Aleman I. Serum insulin-like growth factor I regulates brain amyloid-beta levels. Nat Med United States. 2002;8:1390–7.

    Article  CAS  Google Scholar 

  12. Hammer GP, du Prel J-B, Blettner M. Avoiding bias in observational studies: part 8 in a series of articles on evaluation of scientific publications. Dtsch Arztebl Int Germany. 2009;106:664–8.

    Google Scholar 

  13. Davies NM, Holmes MV, Davey Smith G. Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. BMJ [Internet]. 2018;362:k601. Available from: http://www.bmj.com/content/362/bmj.k601.abstract.

  14. Syed AA, He L, Shi Y. The Potential Effect of Aberrant Testosterone Levels on Common Diseases: A Mendelian Randomization Study. Genes (Basel). 2020.

  15. Williams DM, Karlsson IK, Pedersen NL, Hägg S. Circulating insulin-like growth factors and Alzheimer disease: a mendelian randomization study. Neurol United States. 2018;90:e291–7.

    CAS  Google Scholar 

  16. Ruth KS, Day FR, Tyrrell J, Thompson DJ, Wood AR, Mahajan A et al. Using human genetics to understand the disease impacts of testosterone in men and women. Nat Med [Internet]. 2020;26:252–8. Available from: https://doi.org/10.1038/s41591-020-0751-5.

  17. Neale Lab. GWAS RESULTS. 2018; Available from: http://www.nealelab.is/uk-biobank/.

  18. Kunkle BW, Grenier-Boley B, Sims R, Bis JC, Damotte V, Naj AC, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat Genet. 2019;51:414–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Marioni RE, Harris SE, Zhang Q, McRae AF, Hagenaars SP, Hill WD, et al. GWAS on family history of Alzheimer’s disease. Transl Psychiatry Nature Publishing Group. 2018;8:1–7.

    Google Scholar 

  20. Pan-UKB team. Pan-ancestry genetic analysis of the UK Biobank [Internet]. 2020. Available from: https://pan.ukbb.broadinstitute.org.

  21. Schooling CM, Lopez PM, Yang Z, Zhao JV, Au Yeung SL, Huang JV. Use of Multivariable Mendelian Randomization to Address Biases Due to Competing Risk Before Recruitment [Internet]. Front. Genet. 2021. Available from: https://www.frontiersin.org/articles/https://doi.org/10.3389/fgene.2020.610852.

  22. Schooling CM, Zhao JV, Au Yeung SL, Kwok MK. Letter in response to “Bias in two-sample Mendelian randomization when using heritable covariable-adjusted summary associations’-’Interpreting Mendelian randomization studies pre-adjusted for the heritable covariable survival to recruitment”. Int. J. Epidemiol. England; 2021. p. 1744–5.

  23. Schooling CM, Fei K, Terry MB. Reassessing the causal role of early-life adiposity in breast cancer: could the apparent inverse associations be a manifestation of survival bias? Int J Epidemiol. 2023 Mar;13:dyad027. https://doi.org/10.1093/ije/dyad027.

  24. Lawlor DA, Commentary. Two-sample Mendelian randomization: opportunities and challenges. Int J Epidemiol [Internet]. 2016;45:908–15. Available from: https://doi.org/10.1093/ije/dyw127.

  25. Keevil BG, Adaway J. Assessment of free testosterone concentration. J Steroid Biochem Mol Biol England. 2019;190:207–11.

    Article  CAS  Google Scholar 

  26. Huang Y, Weisgraber KH, Mucke L, Mahley RW, Apolipoprotein E. Diversity of cellular origins, structural and biophysical properties, and effects in Alzheimer’s disease. J Mol Neurosci United States. 2004;23:189–204.

    Article  CAS  Google Scholar 

  27. Deelen J, Evans DS, Arking DE, Tesi N, Nygaard M, Liu X et al. A meta-analysis of genome-wide association studies identifies multiple longevity genes. Nat Commun [Internet]. 2019;10:3669. Available from: https://doi.org/10.1038/s41467-019-11558-2.

  28. Benn M. Rapid response to: Low LDL cholesterol, PCSK9 and HMGCR genetic variation, and risk of Alzheimer’s disease and Parkinson’s disease: Mendelian randomisation study. BMJ [Internet]. 2017; Available from: https://www.bmj.com/content/357/bmj.j1648/rr-1.

  29. Bowden J, Del Greco MF, Minelli C, Davey Smith G, Sheehan NA, Thompson JR. Assessing the suitability of summary data for two-sample Mendelian randomization analyses using MR-Egger regression: the role of the I2 statistic. Int J Epidemiol [Internet]. Oxford University Press; 2016;45:1961–74. Available from: https://pubmed.ncbi.nlm.nih.gov/27616674.

  30. Danaei G, Ding EL, Mozaffarian D, Taylor B, Rehm J, Murray CJL et al. The Preventable Causes of Death in the United States: Comparative Risk Assessment of Dietary, Lifestyle, and Metabolic Risk Factors. PLOS Med [Internet]. Public Library of Science; 2009;6:e1000058. Available from: https://doi.org/10.1371/journal.pmed.1000058.

  31. Eslami A, Naghibi Irvani SS, Ramezankhani A, Fekri N, Asadi K, Azizi F et al. Incidence and associated risk factors for premature death in the Tehran Lipid and Glucose Study cohort, Iran. BMC Public Health [Internet]. 2019;19:719. Available from: https://doi.org/10.1186/s12889-019-7056-y.

  32. Larsson SC, Traylor M, Malik R, Dichgans M, Burgess S, Markus HS. Modifiable pathways in Alzheimer’s disease: Mendelian randomisation analysis. BMJ [Internet]. 2017;359:j5375. Available from: http://www.bmj.com/content/359/bmj.j5375.abstract.

  33. Bhaskaran K, dos-Santos-Silva I, Leon DA, Douglas IJ, Smeeth L. Association of BMI with overall and cause-specific mortality: a population-based cohort study of 3·6 million adults in the UK. Lancet Diabetes Endocrinol [Internet]. Elsevier; 2018;6:944–53. Available from: https://doi.org/10.1016/S2213-8587(18)30288-2

  34. Liu M, Jiang Y, Wedow R, Li Y, Brazel DM, Chen F et al. Association studies of up to 1.2 million individuals yield new insights into the genetic etiology of tobacco and alcohol use. Nat Genet [Internet]. 2019;51:237–44. Available from: https://doi.org/10.1038/s41588-018-0307-5.

  35. Evangelou E, Warren HR, Mosen-Ansorena D, Mifsud B, Pazoki R, Gao H et al. Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nat Genet [Internet]. 2018;50:1412–25. Available from: https://doi.org/10.1038/s41588-018-0205-x.

  36. Timmers PRHJ, Mounier N, Lall K, Fischer K, Ning Z, Feng X et al. Genomics of 1 million parent lifespans implicates novel pathways and common diseases and distinguishes survival chances. Elife [Internet]. eLife Sciences Publications, Ltd; 2019;8:e39856. Available from: https://doi.org/10.7554/eLife.39856.

  37. Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol [Internet]. 2015/06/06. Oxford University Press; 2015;44:512–25. Available from: https://pubmed.ncbi.nlm.nih.gov/26050253.

  38. Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent Estimation in Mendelian Randomization with Some Invalid Instruments Using a Weighted Median Estimator. Genet Epidemiol. 2016/04/12. 2016;40:304–14.

  39. Verbanck M, Chen C-Y, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet [Internet]. 2018;50:693–8. Available from: https://doi.org/10.1038/s41588-018-0099-7.

  40. Burgess S. Sample size and power calculations in mendelian randomization with a single instrumental variable and a binary outcome. Int J Epidemiol Oxford University Press. 2014;43:922–9.

    Google Scholar 

  41. Del Foco GMF, Pichler L, Eller I, Eller P, Benyamin K et al. B,. Serum iron level and kidney function: a Mendelian randomization study. Nephrol Dial Transplant [Internet]. 2017;32:273–8. Available from: https://doi.org/10.1093/ndt/gfw215.

  42. Niu H, Qu Y, Li Z, Wang R, Li L, Li M, et al. Smoking and risk for Alzheimer Disease: a Meta-analysis based on both case-control and cohort study. J Nerv Ment Dis United States. 2018;206:680–5.

    Article  Google Scholar 

  43. Kivipelto M, Helkala EL, Laakso MP, Hänninen T, Hallikainen M, Alhainen K, et al. Midlife vascular risk factors and Alzheimer’s disease in later life: longitudinal, population based study. BMJ Engl. 2001;322:1447–51.

    Article  CAS  Google Scholar 

  44. Sanderson E, Davey Smith G, Windmeijer F, Bowden J. An examination of multivariable mendelian randomization in the single-sample and two-sample summary data settings. Int J Epidemiol. 2019;48:713–27.

    Article  PubMed  Google Scholar 

  45. Burgess S, Davies NM, Thompson SG. Bias due to participant overlap in two-sample mendelian randomization. Genet Epidemiol United States. 2016;40:597–608.

    Article  Google Scholar 

  46. Lv W, Du N, Liu Y, Fan X, Wang Y, Jia X, et al. Low testosterone level and risk of Alzheimer’s Disease in the Elderly Men: a systematic review and Meta-analysis. Mol Neurobiol United States. 2016;53:2679–84.

    Article  CAS  Google Scholar 

  47. Lu PH, Masterman DA, Mulnard R, Cotman C, Miller B, Yaffe K, et al. Effects of testosterone on cognition and mood in male patients with mild Alzheimer disease and healthy elderly men. Arch Neurol United States. 2006;63:177–85.

    Article  Google Scholar 

  48. Tan RS, Pu SJ. A pilot study on the effects of testosterone in hypogonadal aging male patients with Alzheimer’s disease. aging male Off J Int Soc Study Aging Male England. 2003;6:13–7.

    Article  CAS  Google Scholar 

  49. Cherrier MM, Matsumoto AM, Amory JK, Asthana S, Bremner W, Peskind ER, et al. Testosterone improves spatial memory in men with Alzheimer disease and mild cognitive impairment. Neurol United States. 2005;64:2063–8.

    CAS  Google Scholar 

  50. Tan S, Sohrabi HR, Weinborn M, Tegg M, Bucks RS, Taddei K, et al. Effects of Testosterone supplementation on separate cognitive domains in cognitively healthy older men: a Meta-analysis of current randomized clinical trials. Am J Geriatr psychiatry Off J Am Assoc Geriatr Psychiatry England. 2019;27:1232–46.

    Article  Google Scholar 

  51. Schooling CM, Luo S, Au Yeung SL, Thompson DJ, Karthikeyan S, Bolton TR, et al. Genetic predictors of testosterone and their associations with cardiovascular disease and risk factors: a mendelian randomization investigation. Int J Cardiol Netherlands. 2018;267:171–6.

    Article  Google Scholar 

  52. Luo S, Au Yeung SL, Zhao JV, Burgess S, Schooling CM. Association of genetically predicted testosterone with thromboembolism, heart failure, and myocardial infarction: mendelian randomisation study in UK Biobank. BMJ [Internet]. 2019;364:l476. Available from: http://www.bmj.com/content/364/bmj.l476.abstract.

  53. Sardi F, Fassina L, Venturini L, Inguscio M, Guerriero F, Rolfo E, et al. Alzheimer’s disease, autoimmunity and inflammation. The good, the bad and the ugly. Autoimmun Rev Netherlands. 2011;11:149–53.

    Article  CAS  Google Scholar 

  54. Wotton CJ, Goldacre MJ. Associations between specific autoimmune diseases and subsequent dementia: retrospective record-linkage cohort study, UK. J Epidemiol Community Health England. 2017;71:576–83.

    Article  Google Scholar 

  55. Hampl R, Bicíková M. Neuroimmunomodulatory steroids in Alzheimer dementia. J Steroid Biochem Mol Biol England. 2010;119:97–104.

    Article  CAS  Google Scholar 

  56. Foo YZ, Nakagawa S, Rhodes G, Simmons LW. The effects of sex hormones on immune function: a meta-analysis. Biol Rev Camb Philos Soc England. 2017;92:551–71.

    Article  Google Scholar 

  57. Trumble BC, Blackwell AD, Stieglitz J, Thompson ME, Suarez IM, Kaplan H, et al. Associations between male testosterone and immune function in a pathogenically stressed forager-horticultural population. Am J Phys Anthropol United States. 2016;161:494–505.

    Article  Google Scholar 

  58. Kodama L, Gan L. Do microglial sex differences contribute to sex differences in neurodegenerative Diseases? Trends Mol Med England. 2019;25:741–9.

    Article  Google Scholar 

  59. Ostrowski PP, Barszczyk A, Forstenpointner J, Zheng W, Feng Z-P. Meta-Analysis of Serum Insulin-Like Growth Factor 1 in Alzheimer’s Disease. PLoS One [Internet]. Public Library of Science; 2016;11:e0155733. Available from: https://doi.org/10.1371/journal.pone.0155733.

  60. Hu X, Yang Y, Gong D. Circulating insulin-like growth factor 1 and insulin-like growth factor binding protein-3 level in Alzheimer’s disease: a meta-analysis. Neurol Sci Off J Ital Neurol Soc Ital Soc Clin Neurophysiol Italy. 2016;37:1671–7.

    Google Scholar 

  61. Davey Smith G, Lawlor D, Harbord R, Timpson N, Day I, Ebrahim S. Clustered environments and randomized genes: a fundamental distinction between conventional and genetic epidemiology. PLoS Med. 2008;4:e352.

    Article  Google Scholar 

  62. Belloy ME, Napolioni V, Greicius MD. A quarter century of APOE and Alzheimer’s Disease: progress to date and the path Forward. Neuron United States. 2019;101:820–38.

    Article  CAS  Google Scholar 

  63. Hartwig FP, Tilling K, Davey Smith G, Lawlor DA, Borges MC. Bias in two-sample mendelian randomization when using heritable covariable-adjusted summary associations. Int J Epidemiol. 2021;50:1639–50.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Neale Lab (http://www.nealelab.is/uk-biobank/) for conducting the GWAS and providing the sex-specific genetic associations in the UK Biobank. We thank the International Genomics of Alzheimer’s Project (IGAP) for providing summary results data for these analyses. The investigators within IGAP contributed to the design and implementation of IGAP and/or provided data but did not participate in analysis or writing of this report. IGAP was made possible by the generous participation of the control subjects, the patients, and their families. The i–Select chips was funded by the French National Foundation on Alzheimer’s disease and related disorders. EADI was supported by the LABEX (laboratory of excellence program investment for the future) DISTALZ grant, Inserm, Institut Pasteur de Lille, Université de Lille 2 and the Lille University Hospital. GERAD/PERADES was supported by the Medical Research Council (Grant n° 503480), Alzheimer’s Research UK (Grant n° 503176), the Wellcome Trust (Grant n° 082604/2/07/Z) and German Federal Ministry of Education and Research (BMBF): Competence Network Dementia (CND) grant n° 01GI0102, 01GI0711, 01GI0420. CHARGE was partly supported by the NIH/NIA grant R01 AG033193 and the NIA AG081220 and AGES contract N01–AG–12100, the NHLBI grant R01 HL105756, the Icelandic Heart Association, and the Erasmus Medical Center and Erasmus University. ADGC was supported by the NIH/NIA grants: U01 AG032984, U24 AG021886, U01 AG016976, and the Alzheimer’s Association grant ADGC–10–196728.

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

CHCY, CMS and SLAY designed the study. All authors interpreted the results. CHCY conducted the analyses and wrote the first draft of the article with critical feedback and revision from CMS, SLAY, JVZ and MKK. All authors gave approval for the final version of the article for publication.

Corresponding author

Correspondence to Chris Ho Ching Yeung.

Ethics declarations

Conflict of interest

The authors do not have any conflict or financial interest.

Ethics approval

Only publicly available summary data was used in this study, no specific ethical approval is required.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yeung, C., Au Yeung, S., Kwok, M.K. et al. The influence of growth and sex hormones on risk of alzheimer’s disease: a mendelian randomization study. Eur J Epidemiol 38, 745–755 (2023). https://doi.org/10.1007/s10654-023-01015-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10654-023-01015-2

Keywords

Navigation