Skip to main content

Advertisement

Log in

Maternal androgen excess increases the risk of pre-diabetes mellitus in male offspring in later life: a long-term population-based follow-up study

  • Original Article
  • Published:
Journal of Endocrinological Investigation Aims and scope Submit manuscript

Abstract

Purpose

Prenatal androgen exposure could be a source of early programming, leading to the development of cardiometabolic diseases in later life. In this study, we aimed to examine cardiometabolic disturbances in males exposed to maternal androgen excess during their prenatal life.

Methods

In this prospective population-based study, 409 male offspring with maternal hyperandrogenism (MHA), and 954 male offspring without MHA, as controls, were included. Both groups of male offspring were followed from the baseline to the date of the incidence of events, censoring, or end of the study period, whichever came first. Age-scaled unadjusted and adjusted Cox regression models were applied to assess the hazard ratios (HR) and 95% confidence intervals (CIs) for the association between MHA with pre-diabetes mellitus (Pre-DM), type 2 diabetes mellitus (T2DM), pre-hypertension (Pre-HTN), hypertension (HTN), dyslipidemia, overweight, and obesity in the offspring of both groups. Statistical analysis was performed using the STATA software package; the significance level was set at P < 0.05.

Results

A higher risk of Pre-DM (adjusted HR: 1.46 (1.20, 1.78)) was observed in male offspring with MHA after adjustment for potential confounders, including body mass index, education, and physical activity. However, no significant differences were observed in the risk of T2DM, Pre-HTN, HTN, dyslipidemia, overweight, and obesity in males with MHA compared to controls in both the unadjusted and adjusted models.

Conclusion

Maternal androgen excess increases the risk of Pre-DM in male offspring in later life. More longitudinal studies with long enough follow-up are needed to clarify the effects of MHA on the cardiometabolic risk factors of male offspring in later life.

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

Additional data may reasonably be requested from the corresponding author.

References

  1. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, De Ferranti S, Després JP, Fullerton HJ, Howard VJ, Huffman MD (2015) Heart disease and stroke statistics–2015 update: a report from the American Heart Association. Circulation 131:e29–e322. https://doi.org/10.1161/CIR.0000000000000152

    Article  PubMed  Google Scholar 

  2. Kwon EJ, Kim YJ (2017) What is fetal programming?: a lifetime health is under the control of in utero health. Obstet Gynecol Sci 60:506–519. https://doi.org/10.5468/ogs.2017.60.6.506

    Article  PubMed  PubMed Central  Google Scholar 

  3. Recabarren SE, Petermann T, Maliqueo M, Lobos A, Rojas-García P (2006) Prenatal exposure to androgens as a factor of fetal programming. Rev Med Chile 134:101–108. https://doi.org/10.4067/S0034-98872006000100015

    Article  CAS  PubMed  Google Scholar 

  4. Hakim C, Padmanabhan V, Vyas AK (2017) Gestational hyperandrogenism in developmental programming. Endocrinology 158:199–212. https://doi.org/10.1210/en.2016-1801

    Article  CAS  PubMed  Google Scholar 

  5. Puttabyatappa M, Sargis RM, Padmanabhan V (2020) Developmental programming of insulin resistance: are androgens the culprits? J Endocrinol 245:R23–R48. https://doi.org/10.1530/JOE-20-0044

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Carrasco A, Recabarren MP, Rojas-García PP, Silva N, Fuenzalida J, Sir-Petermann T, Recabarren SE (2021) Insulin sensitivity in male sheep born to ewes treated with testosterone during pregnancy. J Dev Orig Health Dis 12:456–464. https://doi.org/10.1017/S2040174420000665

    Article  CAS  PubMed  Google Scholar 

  7. Carrasco A, Recabarren MP, Rojas-García PP, Gutiérrez M, Morales K, Petermann T, Recabarren SE (2020) Prenatal testosterone exposure disrupts insulin secretion and promotes insulin resistance. Sci Rep 10:1–7. https://doi.org/10.1038/s41598-019-57197-x

    Article  CAS  Google Scholar 

  8. Recabarren SE, Smith R, Rios R, Maliqueo M, Echiburu B, Codner E, Cassorla F, RojasP Sir-Petermann T (2008) Metabolic profile in sons of women with polycystic ovary syndrome. J Clin Endocrinol Metab 93:1820–1826. https://doi.org/10.1210/jc.2007-2256

    Article  CAS  PubMed  Google Scholar 

  9. Sherman SB, Sarsour N, Salehi M, Schroering A, Mell B, Joe B, Hill JW (2018) Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes 9:400–421. https://doi.org/10.1080/19490976.2018.1441664

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Azizi F, Madjid M, Rahmani M, Emami H, Mirmiran P, Hadjipour R (2000) Tehran Lipid and Glucose Study (TLGS): rationale and design. Int J Endocrinol Metab 2:77–86

    Google Scholar 

  11. Tehrani FR, Behboudi-Gandevani S, Dovom MR, Farahmand M, Minooee S, Noroozzadeh M, Amiri M, Nazarpour S, Azizi F (2018) Reproductive assessment: findings from 20 years of the Tehran Lipid and Glucose Study. Int J Endocrinol Metab 16:84786. https://doi.org/10.5812/ijem.84786

    Article  Google Scholar 

  12. Escobar-Morreale HF, Carmina E, Dewailly D, Gambineri A, Kelestimur F, Moghetti P, Pugeat M, Qiao J, Wijeyaratne CN, Witchel SF, Norman RJ (2012) Epidemiology, diagnosis and management of hirsutism: a consensus statement by the Androgen Excess and Polycystic Ovary Syndrome Society. Hum Reprod Update. 18:146–70. https://doi.org/10.1093/humupd/dmr042

    Article  CAS  PubMed  Google Scholar 

  13. Tan AU, Schlosser BJ, Paller AS (2018) A review of diagnosis and treatment of acne in adult female patients. Int J Womens Dermatol. 4:56–71. https://doi.org/10.1016/j.ijwd.2017.10.006

    Article  CAS  PubMed  Google Scholar 

  14. Association EAD (2013) Diagnosis and classification of diabetes mellitus. Diabetes Care 36:S67–S74. https://doi.org/10.2337/dc13-S067

    Article  Google Scholar 

  15. Romero-Corral A, Somers VK, Sierra-Johnson J, Thomas RJ, Collazo-Clavell ML, Korinek JE, Allison TG, Batsis JA, Sert-Kuniyoshi FH, Lopez-Jimenez F (2008) Accuracy of body mass index in diagnosing obesity in the adult general population. Int J Obes 32:959–966. https://doi.org/10.1038/ijo.2008.11

    Article  CAS  Google Scholar 

  16. Chobanian AV (2003) National heart, lung, and blood institute; national high blood pressure education program coordinating committee. seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure. Hypertension 42:1206–1252. https://doi.org/10.1161/01.hyp.0000107251.49515.c2

    Article  CAS  PubMed  Google Scholar 

  17. Griffin BA, Anderson GL, Shih RA, Whitsel EA (2012) Use of alternative time scales in Cox proportional hazard models: implications for time-varying environmental exposures. Stat Med 31:3320–3327. https://doi.org/10.1002/sim.5347

    Article  PubMed  PubMed Central  Google Scholar 

  18. Zhang Z (2016) Multiple imputation for time series data with Amelia package. Ann Transl Med 4:56. https://doi.org/10.3978/j.issn.2305-5839.2015.12.60

    Article  PubMed  PubMed Central  Google Scholar 

  19. Skogen JC, Øverland S (2012) The fetal origins of adult disease: a narrative review of the epidemiological literature. JRSM 3:1–7. https://doi.org/10.1258/shorts.2012.012048

    Article  Google Scholar 

  20. Barker DJP (2004) Developmental origins of well being. Philos Trans Royal Soc London 359:1359–66. https://doi.org/10.1098/rstb.2004.1518

    Article  CAS  Google Scholar 

  21. Fowden AL, Forhead AJ (2004) Endocrine mechanisms of intrauterine programming. Reproduction 127:515–526. https://doi.org/10.1530/rep.1.00033

    Article  CAS  PubMed  Google Scholar 

  22. Salamalekis E, Bakas P, Vitoratos N, Eleptheriadis M, Creatsas G (2006) Androgen levels in the third trimester of pregnancy in patients with preeclampsia. Eur J Obstet Gynecol Reprod Biol 126:16–19. https://doi.org/10.1016/j.ejogrb.2005.07.007

    Article  CAS  PubMed  Google Scholar 

  23. Sir-Petermann T, Maliqueo M, Angel B, Lara HE, Perez-Bravo F, Recabarren SE (2002) Maternal serum androgens in pregnant women with polycystic ovarian syndrome: possible implications in prenatal androgenization. Hum Reprod 17:2573–2579. https://doi.org/10.1093/humrep/17.10.2573

    Article  CAS  PubMed  Google Scholar 

  24. Evans DJ, Hoffmann RG, Kalkhoff RK, Kissebah AH (1983) Relationship of androgenic activity to body fat topography, fat cell morphology, and metabolic aberrations in premenopausal women. J Clin Endocrinol Metab 57:304–310. https://doi.org/10.1210/jcem-57-2-304

    Article  CAS  PubMed  Google Scholar 

  25. Guo S (2014) Insulin signaling, resistance, and the metabolic syndrome: insights from mouse models to disease mechanisms. J Endocrinol 220:T1–T23. https://doi.org/10.1530/JOE-13-0327

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nilsson E, Ling C (2017) DNA methylation links genetics, fetal environment, and an unhealthy lifestyle to the development of type 2 diabetes. Clin Epigenetics 9:1–8. https://doi.org/10.1186/s13148-017-0399-2

    Article  CAS  Google Scholar 

  27. Zheng Y, Ley SH, Hu FB (2018) Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol 14:88–98. https://doi.org/10.1038/nrendo.2017.151

    Article  PubMed  Google Scholar 

  28. Franks PW, Pearson E, Florez JC (2013) Gene-environment and gene-treatment interactions in type 2 diabetes: progress, pitfalls, and prospects. Diabetes Care 36:1413–1421. https://doi.org/10.2337/dc12-2211

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Vaiserman A, Lushchak O (2019) Developmental origins of type 2 diabetes: focus on epigenetics. Ageing Res Rev 55:100957. https://doi.org/10.1016/j.arr.2019.100957

    Article  CAS  PubMed  Google Scholar 

  30. Noroozzadeh M, Rahmati M, Behboudi-Gandevani S, Ramezani Tehrani F (2022) Maternal hyperandrogenism is associated with a higher risk of type 2 diabetes mellitus and overweight in adolescent and adult female offspring: a long-term population-based follow-up study. J Endocrinol Invest 45:963–972. https://doi.org/10.1007/s40618-021-01721-2

    Article  CAS  PubMed  Google Scholar 

  31. Tian S, Lin XH, Xiong YM, Liu ME, Yu TT, Lv M, Zhao W, Xu GF, Ding GL, Xu CM, Jin M (2017) Prevalence of prediabetes risk in offspring born to mothers with hyperandrogenism. EBioMedicine 16:275–83. https://doi.org/10.1016/j.ebiom.2017.01.011

    Article  PubMed  PubMed Central  Google Scholar 

  32. Recabarren SE, Smith R, Rios R, Maliqueo M, Echiburu B, Codner E, Cassorla F, Rojas P, Sir-Petermann T (2008) Metabolic profile in sons of women with polycystic ovary syndrome. J Clin Endocrinol Metab 93:1820–1826. https://doi.org/10.1210/jc.2007-2256

    Article  CAS  PubMed  Google Scholar 

  33. Chinnathambi V, Balakrishnan M, Yallampalli C, Sathishkumar K (2012) Prenatal testosterone exposure leads to hypertension that is gonadal hormone-dependent in adult rat male and female offspring. Biology of reproduction. Biol Reprod 86:137–7. https://doi.org/10.1095/biolreprod.103.023689

    Article  CAS  PubMed  Google Scholar 

  34. Bruns CM, Baum ST, Colman RJ, Eisner JR, Kemnitz JW, Weindruch R, Abbott DH (2004) Insulin resistance and impaired insulin secretion in prenatally androgenized male rhesus monkeys. J Clin Endocrinol Metab 89:6218–6223. https://doi.org/10.1210/jc.2004-0918

    Article  CAS  PubMed  Google Scholar 

  35. Lazic M, Aird F, Levine JE, Dunaif A (2011) Prenatal androgen treatment alters body composition and glucose homeostasis in male rats. J Endocrinol 208:293–300. https://doi.org/10.1677/JOE-10-0263

    Article  CAS  PubMed  Google Scholar 

  36. Huang G, Cherkerzian S, Loucks EB, Buka SL, Handa RJ, Lasley BL, Bhasin S, Goldstein JM (2018) Sex differences in the prenatal programming of adult metabolic syndrome by maternal androgens. J Clin Endocrinol Metab 103:3945–3953. https://doi.org/10.1210/jc.2014-2700

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang B, Wang J, Shen S, Liu J, Sun J, Gu T, Ye X, Zhu D, Bi Y (2018) Association of androgen excess with glucose intolerance in women with polycystic ovary syndrome. BioMed Res Int. https://doi.org/10.1155/2018/6869705

    Article  PubMed  PubMed Central  Google Scholar 

  38. Osibogun O, Ogunmoroti O, Michos ED (2020) Polycystic ovary syndrome and cardiometabolic risk: opportunities for cardiovascular disease prevention. Trends Cardiovasc Med 30:399–404. https://doi.org/10.1016/j.tcm.2019.08.010

    Article  PubMed  Google Scholar 

  39. Ramezani Tehrani F, Amiri M, Behboudi-Gandevani S, Bidhendi-Yarandi R, Carmina E (2020) Cardiovascular events among reproductive and menopausal age women with polycystic ovary syndrome: a systematic review and meta-analysis. Gynecol Endocrinol 36:12–23. https://doi.org/10.1080/09513590.2019.1650337

    Article  PubMed  Google Scholar 

  40. Forslund M, Schmidt J, Brännström M, Landin-Wilhelmsen K, Dahlgren E (2022) Morbidity and mortality in PCOS: A prospective follow-up up to a mean age above 80 years. Eur J Obstet Gynecol Reprod Biol 271:195–203. https://doi.org/10.1016/j.ejogrb.2022.02.020

    Article  PubMed  Google Scholar 

  41. Ashraf S, Nabi M, Rashid F, Amin S (2019) Hyperandrogenism in polycystic ovarian syndrome and role of CYP gene variants: a review. EJMHG 20:1. https://doi.org/10.1186/s43042-019-0031-4

    Article  Google Scholar 

  42. Franks S, McCarthy MI, Hardy K (2006) Development of polycystic ovary syndrome: involvement of genetic and environmental factors. Int J Androl 29:278–85. https://doi.org/10.1111/j.1365-2605.2005.00623.x

    Article  CAS  PubMed  Google Scholar 

  43. Barsky M, Merkison J, Hosseinzadeh P, Yang L, Bruno-Gaston J, Dunn J, Gibbons W, Blesson CS (2021) Fetal programming of polycystic ovary syndrome: Effects of androgen exposure on prenatal ovarian development. J Steroid Bioch Mol Biol 207:105830. https://doi.org/10.1016/j.jsbmb.2021.105830

    Article  CAS  Google Scholar 

  44. Cesta CE, Öberg AS, Ibrahimson A, Yusuf I, Larsson H, Almqvist C, D’Onofrio BM, Bulik CM, de la Cruz LF, Mataix-Cols D, Landén M (2020) Maternal polycystic ovary syndrome and risk of neuropsychiatric disorders in offspring: prenatal androgen exposure or genetic confounding? Psychol Med 50:616–24. https://doi.org/10.1017/S0033291719000424

    Article  PubMed  Google Scholar 

  45. Daan NM, Koster MP, Steegers-Theunissen RP, Eijkemans MJ, Fauser BC (2017) Endocrine and cardiometabolic cord blood characteristics of offspring born to mothers with and without polycystic ovary syndrome. Fertil Steril 107:261–8. https://doi.org/10.1016/j.fertnstert.2016.09.042

    Article  CAS  PubMed  Google Scholar 

  46. Mehrabian F, Khani B (2014) Comparison of the metabolic parameters and androgen level of umbilical cord blood in newborns of mothers with polycystic ovary syndrome and controls. Int J Reprod BioMed 12:5

    Google Scholar 

  47. Barry JA, Kay AR, Navaratnarajah R, Iqbal S, Bamfo JE, David AL, Hines M, Hardiman PJ (2010) Umbilical vein testosterone in female infants born to mothers with polycystic ovary syndrome is elevated to male levels. J Obstet Gynaecol 30:444–6. https://doi.org/10.3109/01443615.2010.485254

    Article  CAS  PubMed  Google Scholar 

  48. Palomba S, Russo T, Falbo A, Di Cello A, Tolino A, Tucci L, La Sala GB, Zullo F (2013) Macroscopic and microscopic findings of the placenta in women with polycystic ovary syndrome. Hum Reprod 28:2838–47. https://doi.org/10.1093/humrep/det250

    Article  CAS  PubMed  Google Scholar 

  49. Maliqueo M, Lara HE, Sánchez F, Echiburú B, Crisosto N, Sir-Petermann T (2013) Placental steroidogenesis in pregnant women with polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol 166:151–5. https://doi.org/10.1016/j.ejogrb.2012.10.015

    Article  CAS  PubMed  Google Scholar 

  50. Beckett EM, Astapova O, Steckler TL, Veiga-Lopez A, Padmanabhan V (2014) Developmental programing: impact of testosterone on placental differentiation. Reprod 148:199–209. https://doi.org/10.1530/REP-14-0055

    Article  CAS  Google Scholar 

  51. Padmanabhan V, Veiga-Lopez A (2013) Animal models of the polycystic ovary syndrome phenotype. Steroids 78:734–40. https://doi.org/10.1016/j.steroids.2013.05.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was financially supported by the project (No. 19868) of the Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.

Author information

Authors and Affiliations

Authors

Contributions

M.F.A. contributed substantially to the conception and design, interpretation of data, drafted the article and revised and approved the final version to be published. M.N. contributed substantially to the conception and design, interpretation of data, drafted the article and revised and approved the final version to be published. M.G. contributed substantially to interpretation of data, drafted the article and revised and approved the final version to be published. M.R. contributed substantially to analysis data, drafted the article, and revised and approved the final version to be published. M.S.G.N. contributed substantially to the draft of the article and revised and approved the final version to be published. F.A. contributed substantially to conception and design, revised and approved the final version to be published. F.R.T contributed substantially to conception and design, interpretation of data, drafting the article, and revising and approving the final version to be published.

Corresponding author

Correspondence to M. Noroozzadeh.

Ethics declarations

Conflict of interest

The authors declare that there are no conflicts of interest.

Ethical approval

The ethics review board of the Research Institute for Endocrine Sciences approved the study proposal (approval number: IR.SBMU.ENDOCRINE.REC.1398.125).

Consent to participate

Written informed consent was signed by all participants after a full explanation of the purpose of the study to them. Written consent was obtained from their parents if they were under 18 years old.

Consent for publication

All authors approved the final version of the article for publication.

Additional information

Publisher's Note

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

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

Farhadi-Azar, M., Noroozzadeh, M., Ghahremani, M. et al. Maternal androgen excess increases the risk of pre-diabetes mellitus in male offspring in later life: a long-term population-based follow-up study. J Endocrinol Invest 46, 1775–1785 (2023). https://doi.org/10.1007/s40618-022-01972-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40618-022-01972-7

Keywords

Navigation