Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Multi-omics analysis identifies rare variation in leptin/PPAR gene sets and hypermethylation of ABCG1 contribute to antipsychotics-induced metabolic syndromes

Abstract

Antipsychotic-induced metabolic syndrome (APs-induced Mets) is the most common adverse drug reaction, which affects more than 60% of the psychiatric patients. Although the etiology of APs-induced Mets has been extensively investigated, there is a lack of integrated analysis of the genetic and epigenetic factors. In this study, we performed genome-wide, whole-exome sequencing (WES) and epigenome-wide association studies in schizophrenia (SCZ) patients with or without APs-induced Mets to find the underlying mechanisms, followed by in vitro and in vivo functional validations. By population-based omics analysis, we revealed that rare functional variants across in the leptin and peroxisome proliferator-activated receptors (PPARs) gene sets were imbalanced with rare functional variants across the APs-induced Mets and Non-Mets cohort. Besides, we discovered that APs-induced Mets are hypermethylated in ABCG1 (chr21:43642166–43642366, adjusted P < 0.05) than Non-Mets, and hypermethylation of this area was associated with higher TC (total cholesterol) and TG (triglycerides) levels in HepG2 cells. Candidate genes from omics studies were furtherly screened in C. elegans and 17 gene have been verified to associated with olanzapine (OLA) induced fat deposit. Among them, several genes were expressed differentially in Mets cohort and APs-induced in vitro/in vivo models compared to controls, demonstrating the validity of omics study. Overexpression one of the most significant gene, PTPN11, exhibited compromised glucose responses and insulin resistance. Pharmacologic inhibition of PTPN11 protected HepG2 cell from APs-induced insulin resistance. These findings provide important insights into our understanding of the mechanism of the APs-induced Mets.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Details of the genome-wide top hit rs12930712.
Fig. 2: Epigenome-wide association results and enrichment analysis of top-100 CpG sites regarding Mets in schizophrenia cohorts.
Fig. 3: Differentially methylated regions (DMRs) associated with Mets in the schizophrenia cohort.
Fig. 4: RNAi screen identifies genes associated with OLA-induced fat deposit in C. elegans.
Fig. 5: Gene expression level in models of APs-induced Mets and pharmacological inhibition of PTPN11 rescues OLA-induced Mets in vitro.

Similar content being viewed by others

Data availability

Raw data have been deposited in the China National Center for Bioinformation and are available upon request to SQ (chinsir@sjtu.edu.cn). All other data are available in the Supplementary Information.

Code availability

Scripts used to generate the results are available on request from the corresponding author.

References

  1. Arciniegas DB. Psychosis. Continuum: Lifelong Learning in Neurology, 2015 Jun;21(3 Behavioral Neurology and Neuropsychiatry):715–36.

  2. Chang WC, Wong C, Chen E, Lam L, Chan WC, Ng R, et al. Lifetime prevalence and correlates of schizophrenia-spectrum, affective, and other non-affective psychotic disorders in the Chinese adult population. Schizophr Bull. 2017;43:1280–90.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Escudero M, Gutierrez-Rojas L, Lahera G. Second generation antipsychotics monotherapy as maintenance treatment for bipolar disorder: a systematic review of long-term studies. Psychiatr Q. 2020;91:1047–60.

    Article  PubMed  Google Scholar 

  4. Mitchell AJ, Vancampfort D, Sweers K, van Winkel R, Yu W, De Hert M. Prevalence of metabolic syndrome and metabolic abnormalities in schizophrenia and related disorders—a systematic review and meta-analysis. Schizophrenia Bull. 2013;39:306–18.

    Article  Google Scholar 

  5. Chang SC, Goh KK, Lu ML. Metabolic disturbances associated with antipsychotic drug treatment in patients with schizophrenia: state-of-the-art and future perspectives. World J Psychiatry. 2021;11:696–710.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Elder SJ, Lichtenstein AH, Pittas AG, Roberts SB, Fuss PJ, Greenberg AS, et al. Genetic and environmental influences on factors associated with cardiovascular disease and the metabolic syndrome. J Lipid Res. 2009;50:1917–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fanning E, O’Shea D. Genetics and the metabolic syndrome. Clin Dermatol. 2017;1:9–13.

    Google Scholar 

  8. Puangpetch A, Unaharassamee W, Jiratjintana N, Koomdee N, Sukasem C. Genetic polymorphisms of HTR2C, LEP and LEPR on metabolic syndromes in patients treated with atypical antipsychotic drugs. J Pharm Pharmacol. 2018;70:536–42.

    Article  CAS  PubMed  Google Scholar 

  9. Yang L, Chen J, Li Y, Wang Y, Liang S, Shi Y, et al. Association between SCAP and SREBF1 gene polymorphisms and metabolic syndrome in schizophrenia patients treated with atypical antipsychotics. World J Biol Psychiatry. 2016;17:467–74.

  10. Cui D, Peng Y, Zhang C, Li Z, Su Y, Qi Y, et al. Macrophage migration inhibitory factor mediates metabolic dysfunction induced by atypical antipsychotic therapy. J Clin Investig. 2018;128:4997–5007.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Yu H, Wang L, Lv L, Ma C, Du B, Lu T, et al. Genome-Wide Association Study suggested the PTPRD polymorphisms were associated with weight gain effects of atypical antipsychotic medications. Schizophrenia Bull. 2016;42:814–23.

    Article  Google Scholar 

  12. Lott SA, Burghardt PR, Burghardt KJ, Bly MJ, Grove TB, Ellingrod VL. The influence of metabolic syndrome, physical activity and genotype on catechol-O-methyl transferase promoter-region methylation in schizophrenia. Pharmacogenom J. 2012;13:264–71.

    Article  Google Scholar 

  13. Burghardt KJ, Pilsner JR, Bly MJ, Ellingrod VL. DNA methylation in schizophrenia subjects: gender and MTHFR 677C/T genotype differences. Epigenomics. 2012;4:261–8.

    Article  CAS  PubMed  Google Scholar 

  14. Burghardt KJ, Goodrich JM, Lines BN, Ellingrod VL. The influence of metabolic syndrome and sex on the DNA methylome in schizophrenia. Int J Genom. 2018;2018:1–12.

    Article  Google Scholar 

  15. Zhu J, Gao R, Zhao S, Lu G, Zhao D, Li J. 2016 Chinese guidelines for the management of dyslipidemia in adults. J Geriatr Cardiol. 2018;15:1–29.

  16. Rio DC, Ares MJ, Hannon GJ, Nilsen TW. Purification of RNA using TRIzol (TRI reagent). Cold Spring Harb Protoc. 2010;2010:t5439.

    Article  Google Scholar 

  17. Carli M, Kolachalam S, Longoni B, Pintaudi A, Baldini M, Aringhieri S, et al. Atypical Antipsychotics and Metabolic Syndrome: From Molecular Mechanisms to Clinical Differences. Pharmaceuticals. 2021;14:238.

  18. Tyagi S, Gupta P, Saini AS, Kaushal C, Sharma S. The peroxisome proliferator-activated receptor: a family of nuclear receptors role in various diseases. J Adv Pharm Technol Res. 2011;2:236–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sárvári AK, Veréb Z, Uray IP, Fésüs L, Balajthy Z. Atypical antipsychotics induce both proinflammatory and adipogenic gene expression in human adipocytes in vitro. Biochem Biophys Res Co. 2014;450:1383–9.

    Article  Google Scholar 

  20. Su Y, Liu X, Lian J, Deng C. Epigenetic histone modulations of PPARγ and related pathways contribute to olanzapine-induced metabolic disorders. Pharmacol Res. 2020;155:104703.

    Article  CAS  PubMed  Google Scholar 

  21. Klok MD, Jakobsdottir S, Drent ML. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: a review. Obes Rev. 2007;8:21–34.

    Article  CAS  PubMed  Google Scholar 

  22. Paz-Filho G, Mastronardi C, Wong ML, Licinio J. Leptin therapy, insulin sensitivity, and glucose homeostasis. Indian J Endocrinol Metab. 2012;16:S549–55.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Potvin S, Zhornitsky S, Stip E. Antipsychotic-induced changes in blood levels of leptin in schizophrenia: a meta-analysis. Can J Psychiatry. 2015;60:S26–34.

    PubMed  PubMed Central  Google Scholar 

  24. Endomba FT, Tankeu AT, Nkeck JR, Tochie JN. Leptin and psychiatric illnesses: does leptin play a role in antipsychotic-induced weight gain? Lipids Health Dis. 2020;19:22.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Choi E, Kikuchi S, Gao H, Brodzik K, Nassour I, Yopp A, et al. Mitotic regulators and the SHP2-MAPK pathway promote IR endocytosis and feedback regulation of insulin signaling. Nat Commun. 2019;10:1473.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Yue X, Han T, Hao W, Wang M, Fu Y. SHP2 knockdown ameliorates liver insulin resistance by activating IRS-2 phosphorylation through the AKT and ERK1/2 signaling pathways. Febs Open Bio. 2020;10:2578–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Paccoud R, Saint-Laurent C, Piccolo E, Tajan M, Dortignac A, Pereira O, et al. SHP2 drives inflammation-triggered insulin resistance by reshaping tissue macrophage populations. Sci Transl Med. 2021;13:eabe2587.

  28. Liu W, Yin Y, Wang M, Fan T, Zhu Y, Shen L, et al. Disrupting phosphatase SHP2 in macrophages protects mice from high-fat diet-induced hepatic steatosis and insulin resistance by elevating IL-18 levels. J Biol Chem. 2020;295:10842–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Dayeh T, Tuomi T, Almgren P, Perfilyev A, Jansson PA, de Mello VD, et al. DNA methylation of loci within ABCG1 and PHOSPHO1 in blood DNA is associated with future type 2 diabetes risk. Epigenetics. 2016;11:482–8.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Dekkers KF, van Iterson M, Slieker RC, Moed MH, Bonder MJ, van Galen M, et al. Blood lipids influence DNA methylation in circulating cells. Genome Biol. 2016;17:138.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Park HS, Lee K, Kim SH, Hong MJ, Jeong NJ, Kim MS. Luteolin improves hypercholesterolemia and glucose intolerance through LXRalpha-dependent pathway in diet-induced obese mice. J Food Biochem. 2020;44:e13358.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Nature Science Foundation of China (81773818, 81273596, 30900799, 81671326, 81503051), National key research and development program (2016YFC0905000, 2016YFC0905002, 2016YFC1200200, 2016YFC0906400), Shanghai science and Technology Innovation Fund (20DZ2202000, 21002411100), Major projects of scientific and technological innovation 2030 (2021ZD0200801), Shanghai Municipal Science and Technology Major Project (Grant No. 2017SHZDZX01), The 4th Three-year Action Plan for Public Health of Shanghai (The Project No.: 15GWZK0101), 111 project, Shanghai Pujiang Program (17PJD020), Natural Science Foundation of Shanghai (22ZR1430300), Shanghai Key Laboratory of Psychotic Disorders (13dz2260500), Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) Program of Shanghai Jiao Tong University (2020GDND04, 2021GDND02), China Postdoctoral Science Foundation (2017M621488), Youth Backbone Teacher Training Project of Jiangsu University (Jing Sun, 2015), Effective & Toxicity Monitoring Innovative Practice Center for Jiangsu University Food Pharmaceutical Specialty (2020).

Author information

Authors and Affiliations

Authors

Contributions

ZW performed the data analysis, experiments and wrote the paper; SJ, LYX, and HC contributed to in vitro and in vivo experiments. YZH, LQY, SCF, ZWL, LCX, WSZ, HS, WH, ZN, ZY, and HS contributed to sample recruitment and preparation; ZW, GYJ, WH, HSJ, ZN, and ZY contributed to sample and library prepare; ZW, HC, SYD, LM, DHH, SRX, and ZJH contributed to data analysis; ZW, WH, ZRS, WLF, and ZYT contributed to C. elegans experiments; HC, LM, CL, HHL, WMY, SXF, and HL revised the paper; QSY designed and supervised the whole study.

Corresponding author

Correspondence to Shengying Qin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Rights and permissions

Springer Nature or its licensor 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

Zhou, W., Sun, J., Huai, C. et al. Multi-omics analysis identifies rare variation in leptin/PPAR gene sets and hypermethylation of ABCG1 contribute to antipsychotics-induced metabolic syndromes. Mol Psychiatry 27, 5195–5205 (2022). https://doi.org/10.1038/s41380-022-01759-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-022-01759-5

Search

Quick links