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:

Intrinsically dysregulated cellular stress signaling genes and gene networks in postpartum depression

Abstract

Postpartum depression (PPD) is a leading cause of morbidity and mortality among women. Clinically, the administration and withdrawal of supraphysiologic estradiol and progesterone (E2 + P) can cause affective symptom reoccurrence in women with a history of PPD, but not matched controls. To investigate the cellular basis underlying this differential affective response, lymphoblastoid cell lines (LCLs) were derived from women with and without past PPD and compared transcriptomically in hormone conditions mimicking pregnancy and parturition: supraphysiologic E2 + P-addback; supraphysiologic E2 + P-withdrawal; and no added E2 + P (Baseline). RNA-sequencing identified unique differentially expressed genes (DEGs) in all hormone conditions, but the majority tended to be downregulated in PPD and observed in E2 + P-addback. Two of these DEGs were evolutionarily conserved cellular stress regulators: IMPACT, an integrative response protein maintaining translational homeostasis, and WWTR1, a transcriptional coactivator in the ‘Hippo’ pathway mediating cell proliferation and survival. Correspondingly, significant gene network modules were linked to cell cycle progression, estrogen response, and immune dysregulation, suggesting innate differences in intracellular signaling in PPD. In certain hormone conditions, PPD LCLs displayed increased GATA3 expression (an upstream regulator of IMPACT and WWTR1) and differentially phosphorylated eiF2α (the ultimate downstream target of IMPACT). Taken together, these transcriptomic data primarily implicate innately dysregulated cellular responses as potentially influencing mood and/or escalating PPD risk. Furthermore, the intrinsic downregulation of IMPACT’s translation and WWTR1’s transcription networks may suggest a novel link between PPD and a compromised ability to maintain homeostasis in the context of cellular stress occurring during pregnancy and parturition.

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: Transcriptomics reveal differentially expressed genes (DEGs) in PPD LCLs compared to Controls in all steroid hormone conditions (HCs), identifying the most FDR-significant DEGs in E2+P-Addback and a bias toward transcript downregulation in PPD.
Fig. 2: IMPACT and WWTR1 (TAZ), cellular stress signaling genes, are decreased in PPD LCLs.
Fig. 3: Transcription factors GATA3 and MYC, and related gene networks, are dysregulated in PPD.
Fig. 4: eiF2α, a downstream target of IMPACT, is differentially phosphorylated at baseline in PPD LCLs.
Fig. 5: Weighted Gene Correlation Network Analysis (WGCNA) uncovers eight PPD significant modules with enriched functions including IL-2/STAT5, GATA, MYC, estrogen response, and E2F cell cycle regulation.

Similar content being viewed by others

References

  1. Hahn-Holbrook J, Cornwell-Hinrichs T, Anaya I. Economic and Health Predictors of National Postpartum Depression Prevalence: A Systematic Review, Meta-analysis, and Meta-Regression of 291 Studies from 56 Countries. Front Psychiatry. 2017;8:248.

    Article  PubMed  Google Scholar 

  2. Meltzer-Brody S, Howard LM, Bergink V, Vigod S, Jones I, Munk-Olsen T, et al. Postpartum psychiatric disorders. Nat Rev Dis Prim. 2018;4:18022.

    Article  PubMed  Google Scholar 

  3. Gavin NI, Gaynes BN, Lohr KN, Meltzer-Brody S, Gartlehner G, Swinson T. Perinatal depression: a systematic review of prevalence and incidence. Obstet Gynecol. 2005;106:1071–83.

    Article  PubMed  Google Scholar 

  4. Oates M. Perinatal psychiatric disorders: a leading cause of maternal morbidity and mortality. Br Med Bull. 2003;67:219–29.

    Article  PubMed  Google Scholar 

  5. Josefsson A, Sydsjo G. A follow-up study of postpartum depressed women: recurrent maternal depressive symptoms and child behavior after four years. Arch Women’s Ment Health. 2007;10:141–5.

    Article  CAS  Google Scholar 

  6. Lindahl V, Pearson JL, Colpe L. Prevalence of suicidality during pregnancy and the postpartum. Arch Women’s Ment Health. 2005;8:77–87.

    Article  CAS  Google Scholar 

  7. Segre LS, O’Hara MW, Arndt S, Stuart S. The prevalence of postpartum depression: the relative significance of three social status indices. Soc Psychiatry Psychiatr Epidemiol. 2007;42:316–21.

    Article  PubMed  Google Scholar 

  8. Meltzer-Brody S, Colquhoun H, Riesenberg R, Epperson CN, Deligiannidis KM, Rubinow DR, et al. Brexanolone injection in post-partum depression: two multicentre, double-blind, randomised, placebo-controlled, phase 3 trials. Lancet. 2018;392:1058–70.

    Article  CAS  PubMed  Google Scholar 

  9. Melon L, Hammond R, Lewis M, Maguire J. A Novel, Synthetic, Neuroactive Steroid Is Effective at Decreasing Depression-Like Behaviors and Improving Maternal Care in Preclinical Models of Postpartum Depression. Front Endocrinol (Lausanne). 2018;9:703.

    Article  PubMed  Google Scholar 

  10. O’Hara MW, Schlechte JA, Lewis DA, Wright EJ. Prospective study of postpartum blues. Biologic and psychosocial factors. Arch Gen Psychiatry. 1991;48:801–6.

    Article  PubMed  Google Scholar 

  11. Harris B. Biological and hormonal aspects of postpartum depressed mood. Br J Psychiatry. 1994;164:288–92.

    Article  CAS  PubMed  Google Scholar 

  12. Bloch M, Schmidt PJ, Danaceau M, Murphy J, Nieman L, Rubinow DR. Effects of gonadal steroids in women with a history of postpartum depression. Am J Psychiatry. 2000;157:924–30.

    Article  CAS  PubMed  Google Scholar 

  13. Schiller CE, Meltzer-Brody S, Rubinow DR. The role of reproductive hormones in postpartum depression. CNS Spectr. 2015;20:48–59.

    Article  PubMed  Google Scholar 

  14. Bloch M, Daly RC, Rubinow DR. Endocrine factors in the etiology of postpartum depression. Compr Psychiatry. 2003;44:234–46.

    Article  PubMed  Google Scholar 

  15. Schiller CE, Walsh E, Eisenlohr-Moul TA, Prim J, Dichter GS, Schiff L, et al. Effects of gonadal steroids on reward circuitry function and anhedonia in women with a history of postpartum depression. J Affect Disord. 2022;314:176–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Plana-Ripoll O, Pedersen CB, Holtz Y, Benros ME, Dalsgaard S, de Jonge P, et al. Exploring Comorbidity Within Mental Disorders Among a Danish National Population. JAMA Psychiatry. 2019;76:259–70.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Munk-Olsen T, Ingstrup KG, Johannsen BM, Liu X. Population-Based Assessment of the Recurrence Risk of Postpartum Mental Disorders: Will It Happen Again? JAMA Psychiatry. 2020;77:213–4.

    Article  PubMed  Google Scholar 

  18. Di Florio A, Gordon-Smith K, Forty L, Kosorok MR, Fraser C, Perry A, et al. Stratification of the risk of bipolar disorder recurrences in pregnancy and postpartum. Br J Psychiatry. 2018;213:542–7.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Guintivano J, Arad M, Gould TD, Payne JL, Kaminsky ZA. Antenatal prediction of postpartum depression with blood DNA methylation biomarkers. Mol Psychiatry. 2014;19:560–7.

    Article  CAS  PubMed  Google Scholar 

  20. Kaminsky Z, Payne J. Seeing the future: epigenetic biomarkers of postpartum depression. Neuropsychopharmacology. 2014;39:233–4.

    Article  PubMed  Google Scholar 

  21. Mehta D, Rex-Haffner M, Sondergaard HB, Pinborg A, Binder EB, Frokjaer VG. Evidence for oestrogen sensitivity in perinatal depression: pharmacological sex hormone manipulation study. Br J Psychiatry. 2019;215:519–27.

    Article  PubMed  Google Scholar 

  22. Bauer AE, Maegbaek ML, Liu X, Wray NR, Sullivan PF, Miller WC, et al. Familiality of Psychiatric Disorders and Risk of Postpartum Psychiatric Episodes: A Population-Based Cohort Study. Am J Psychiatry. 2018;175:783–91.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Forty L, Jones L, Macgregor S, Caesar S, Cooper C, Hough A, et al. Familiality of postpartum depression in unipolar disorder: results of a family study. Am J Psychiatry. 2006;163:1549–53.

    Article  PubMed  Google Scholar 

  24. Corwin EJ, Kohen R, Jarrett M, Stafford B. The heritability of postpartum depression. Biol Res Nurs. 2010;12:73–83.

    Article  CAS  PubMed  Google Scholar 

  25. Treloar SA, Martin NG, Bucholz KK, Madden PA, Heath AC. Genetic influences on post-natal depressive symptoms: findings from an Australian twin sample. Psychol Med. 1999;29:645–54.

    Article  CAS  PubMed  Google Scholar 

  26. Viktorin A, Meltzer-Brody S, Kuja-Halkola R, Sullivan PF, Landén M, Lichtenstein P, et al. Heritability of Perinatal Depression and Genetic Overlap With Nonperinatal Depression. Am J Psychiatry. 2016;173:158–65.

    Article  PubMed  Google Scholar 

  27. Mahon PB, Payne JL, MacKinnon DF, Mondimore FM, Goes FS, Schweizer B, et al. Genome-wide linkage and follow-up association study of postpartum mood symptoms. Am J Psychiatry. 2009;166:1229–37.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Kiewa J, Meltzer-Brody S, Milgrom J, Guintivano J, Hickie IB, Whiteman DC, et al. Perinatal depression is associated with a higher polygenic risk for major depressive disorder than non-perinatal depression. Depress Anxiety. 2022;39:182–91.

    Article  PubMed  Google Scholar 

  29. Pan D, Xu Y, Zhang L, Su Q, Chen M, Li B, et al. Gene expression profile in peripheral blood mononuclear cells of postpartum depression patients. Sci Rep. 2018;8:10139.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Mehta D, Newport DJ, Frishman G, Kraus L, Rex-Haffner M, Ritchie JC, et al. Early predictive biomarkers for postpartum depression point to a role for estrogen receptor signaling. Psychol Med. 2014;44:2309–22.

    Article  CAS  PubMed  Google Scholar 

  31. Mehta D, Grewen K, Pearson B, Wani S, Wallace L, Henders AK, et al. Genome-wide gene expression changes in postpartum depression point towards an altered immune landscape. Transl Psychiatry. 2021;11:155.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Dubey N, Hoffman JF, Schuebel K, Yuan Q, Martinez PE, Nieman LK, et al. The ESC/E(Z) complex, an effector of response to ovarian steroids, manifests an intrinsic difference in cells from women with premenstrual dysphoric disorder. Mol Psychiatry. 2017;22:1172–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Li HJ, Goff A, Rudzinskas SA, Jung Y, Dubey N, Hoffman J, et al. Altered estradiol-dependent cellular Ca(2+) homeostasis and endoplasmic reticulum stress response in Premenstrual Dysphoric Disorder. Mol Psychiatry. 2021;26:6963–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rudzinskas S, Hoffman JF, Martinez P, Rubinow DR, Schmidt PJ, Goldman D. In vitro model of perimenopausal depression implicates steroid metabolic and proinflammatory genes. Mol Psychiatry. 2021;26:3266–76.

    Article  CAS  PubMed  Google Scholar 

  35. Oh HM, Oh JM, Choi SC, Kim SW, Han WC, Kim TH, et al. An efficient method for the rapid establishment of Epstein-Barr virus immortalization of human B lymphocytes. Cell Prolif. 2003;36:191–7.

    Article  PubMed  Google Scholar 

  36. Berthois Y, Katzenellenbogen JA, Katzenellenbogen BS. Phenol red in tissue culture media is a weak estrogen: implications concerning the study of estrogen-responsive cells in culture. Proc Natl Acad Sci USA. 1986;83:2496–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Welshons WV, Wolf MF, Murphy CS, Jordan VC. Estrogenic activity of phenol red. Mol Cell Endocrinol. 1988;57:169–78.

    Article  CAS  PubMed  Google Scholar 

  38. Milo GE, Malarkey WB, Powell JE, Blakeslee JR, Yohn DS. Effects of steroid hormones in fetal bovine serum on plating ang cloning of human cells in vitro. Vitro. 1976;12:23–30.

    Article  CAS  Google Scholar 

  39. van der Valk J. Fetal bovine serum-a cell culture dilemma. Science. 2022;375:143–4.

    Article  PubMed  Google Scholar 

  40. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.

    Article  CAS  PubMed  Google Scholar 

  41. Hulsen T, de Vlieg J, Alkema W. BioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genom. 2008;9:488.

    Article  Google Scholar 

  42. Wang M, Zhao Y, Zhang B. Efficient Test and Visualization of Multi-Set Intersections. Sci Rep. 2015;5:16923.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinforma. 2008;9:559.

    Article  Google Scholar 

  44. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinforma. 2013;14:128.

    Article  Google Scholar 

  45. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–8.

    Article  CAS  PubMed  Google Scholar 

  46. Lachmann A, Xu H, Krishnan J, Berger SI, Mazloom AR, Ma’ayan A. ChEA: transcription factor regulation inferred from integrating genome-wide ChIP-X experiments. Bioinformatics. 2010;26:2438–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Baird TD, Wek RC. Eukaryotic initiation factor 2 phosphorylation and translational control in metabolism. Adv Nutr. 2012;3:307–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Meaney MJ. Perinatal Maternal Depressive Symptoms as an Issue for Population Health. Am J Psychiatry. 2018;175:1084–93.

    Article  PubMed  Google Scholar 

  49. Stowe ZN, Hostetter AL, Newport DJ. The onset of postpartum depression: Implications for clinical screening in obstetrical and primary care. Am J Obstet Gynecol. 2005;192:522–6.

    Article  PubMed  Google Scholar 

  50. Stuart-Parrigon K, Stuart S. Perinatal depression: an update and overview. Curr Psychiatry Rep. 2014;16:468.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Katz ER, Stowe ZN, Newport DJ, Kelley ME, Pace TW, Cubells JF, et al. Regulation of mRNA expression encoding chaperone and co-chaperone proteins of the glucocorticoid receptor in peripheral blood: association with depressive symptoms during pregnancy. Psychol Med. 2012;42:943–56.

    Article  CAS  PubMed  Google Scholar 

  52. Poljsak B, Milisav I. Clinical implications of cellular stress responses. Bosn J Basic Med Sci. 2012;12:122–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hilman D, Gat U. The evolutionary history of YAP and the hippo/YAP pathway. Mol Biol Evol. 2011;28:2403–17.

    Article  CAS  PubMed  Google Scholar 

  54. Cambiaghi TD, Pereira CM, Shanmugam R, Bolech M, Wek RC, Sattlegger E, et al. Evolutionarily conserved IMPACT impairs various stress responses that require GCN1 for activating the eIF2 kinase GCN2. Biochem Biophys Res Commun. 2014;443:592–7.

    Article  CAS  PubMed  Google Scholar 

  55. Plouffe SW, Lin KC, Moore JL III, Tan FE, Ma S, Ye Z, et al. The Hippo pathway effector proteins YAP and TAZ have both distinct and overlapping functions in the cell. J Biol Chem. 2018;293:11230–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Purdy MP, Ducharme M, Haak AJ, Ravix J, Tan Q, Sicard D, et al. YAP/TAZ are Activated by Mechanical and Hormonal Stimuli in Myometrium and Exhibit Increased Baseline Activation in Uterine Fibroids. Reprod Sci. 2020;27:1074–85.

    Article  CAS  PubMed  Google Scholar 

  57. Stepan J, Anderzhanova E, Gassen NC. Hippo Signaling: Emerging Pathway in Stress-Related Psychiatric Disorders? Front Psychiatry. 2018;9:715.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Zhao L, Bracken MB, DeWan AT. Genome-wide association study of pre-eclampsia detects novel maternal single nucleotide polymorphisms and copy-number variants in subsets of the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study cohort. Ann Hum Genet. 2013;77:277–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhang Y, Ho K, Keaton JM, Hartzel DN, Day F, Justice AE, et al. A genome-wide association study of polycystic ovary syndrome identified from electronic health records. Am J Obstet Gynecol. 2020;223:559 e551–21.

    Article  Google Scholar 

  60. Koric A, Singh B, VanDerslice JA, Stanford JB, Rogers CR, Egan DT, et al. Polycystic ovary syndrome and postpartum depression symptoms: a population-based cohort study. Am J Obstet Gynecol. 2021;224:591 e591–e512.

    Article  Google Scholar 

  61. Sorrentino G, Ruggeri N, Zannini A, Ingallina E, Bertolio R, Marotta C, et al. Glucocorticoid receptor signalling activates YAP in breast cancer. Nat Commun. 2017;8:14073.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Liu S, Miao R, Zhai M, Pang Q, Deng Y, Liu S, et al. Effects and related mechanisms of serotonin on malignant biological behavior of hepatocellular carcinoma via regulation of Yap. Oncotarget. 2017;8:47412–24.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Okamura K, Hagiwara-Takeuchi Y, Li T, Vu TH, Hirai M, Hattori M, et al. Comparative genome analysis of the mouse imprinted gene impact and its nonimprinted human homolog IMPACT: toward the structural basis for species-specific imprinting. Genome Res. 2000;10:1878–89.

    Article  CAS  PubMed  Google Scholar 

  64. Pereira CM, Sattlegger E, Jiang HY, Longo BM, Jaqueta CB, Hinnebusch AG, et al. IMPACT, a protein preferentially expressed in the mouse brain, binds GCN1 and inhibits GCN2 activation. J Biol Chem. 2005;280:28316–23.

    Article  CAS  PubMed  Google Scholar 

  65. Pakos-Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A, Gorman AM. The integrated stress response. EMBO Rep. 2016;17:1374–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kosaki K, Suzuki T, Kosaki R, Yoshihashi H, Itoh M, Goto Y, et al. Human homolog of the mouse imprinted gene Impact resides at the pericentric region of chromosome 18 within the critical region for bipolar affective disorder. Mol Psychiatry. 2001;6:87–91.

    Article  CAS  PubMed  Google Scholar 

  67. Bittencourt S, Pereira CM, Avedissian M, Delamano A, Mello LE, Castilho BA. Distribution of the protein IMPACT, an inhibitor of GCN2, in the mouse, rat, and marmoset brain. J Comp Neurol. 2008;507:1811–30.

    Article  CAS  PubMed  Google Scholar 

  68. Pereira CM, Filev R, Dubiela FP, Brandao BB, Queiroz CM, Ludwig RG, et al. The GCN2 inhibitor IMPACT contributes to diet-induced obesity and body temperature control. PLoS ONE. 2019;14:e0217287.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Viger RS, Guittot SM, Anttonen M, Wilson DB, Heikinheimo M. Role of the GATA family of transcription factors in endocrine development, function, and disease. Mol Endocrinol. 2008;22:781–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Yoon NK, Maresh EL, Shen D, Elshimali Y, Apple S, Horvath S, et al. Higher levels of GATA3 predict better survival in women with breast cancer. Hum Pathol. 2010;41:1794–801.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Blais L, Salah Ahmed SI, Beauchesne MF, Forget A, Kettani FZ, Lavoie KL. Risk of Postpartum Depression Among Women with Asthma. J Allergy Clin Immunol Pr. 2019;7:925–33.e922.

    Article  Google Scholar 

  72. de Kluiver H, Jansen R, Milaneschi Y, Penninx B. Involvement of inflammatory gene expression pathways in depressed patients with hyperphagia. Transl Psychiatry. 2019;9:193.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Wong ML, Dong C, Maestre-Mesa J, Licinio J. Polymorphisms in inflammation-related genes are associated with susceptibility to major depression and antidepressant response. Mol Psychiatry. 2008;13:800–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Reimer D, Sadr S, Wiedemair A, Goebel G, Concin N, Hofstetter G, et al. Expression of the E2F family of transcription factors and its clinical relevance in ovarian cancer. Ann N. Y Acad Sci. 2006;1091:270–81.

    Article  CAS  PubMed  Google Scholar 

  75. Davis JN, Wojno KJ, Daignault S, Hofer MD, Kuefer R, Rubin MA, et al. Elevated E2F1 Inhibits Transcription of the Androgen Receptor in Metastatic Hormone-Resistant Prostate Cancer. Cancer Res. 2006;66:11897–906.

    Article  CAS  PubMed  Google Scholar 

  76. Roffe M, Hajj GN, Azevedo HF, Alves VS, Castilho BA. IMPACT is a developmentally regulated protein in neurons that opposes the eukaryotic initiation factor 2alpha kinase GCN2 in the modulation of neurite outgrowth. J Biol Chem. 2013;288:10860–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Carr JR, Kiefer MM, Park HJ, Li J, Wang Z, Fontanarosa J, et al. FoxM1 regulates mammary luminal cell fate. Cell Rep. 2012;1:715–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Ching T, Huang S, Garmire LX. Power analysis and sample size estimation for RNA-Seq differential expression. RNA. 2014;20:1684–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank Cheryl Marietta, Longina Akhtar, Howard Li, Alan Meyers, and Damien Islek of NIH/NIAAA for their technical assistance, support, and expertize in conducting this study. In addition, we thank Erin Richardson and Laura Lundegard of UNC for assisting with study administration and specimen collection. This work was written as part of PJS’s and DG’s official duties as government employees. This research was supported by the Office of Clinical Research Bench to Bedside Program Funds Award #480670 and the Intramural Research Programs of the NIMH/NIH and NIAAA/NIH; Protocols 95-M-0097 and 03-M-0138; NIMH Project #MH002865 and NIAAA Project #AA000301. Extramural funding from the Foundation of Hope, Brain & Behavior Research Foundation, and NIH R21MH101409. Clinical Trial: NCT01762943.

Author information

Authors and Affiliations

Authors

Contributions

SR designed, planned, performed, and analyzed the experiments and data. AG contributed the computational framework for data analysis. MM helped perform the experiments. CES, and SM-B designed the protocols that provided the clinical samples used. DR, PS, and DG conceived the research questions and contributed to the interpretation of the results. SR led the drafting of the paper in direct consultation with PS and DG. All authors gave critical input and approved the final paper.

Corresponding author

Correspondence to Peter J. Schmidt.

Ethics declarations

Competing interests

The authors declare no competing interests or financial support regarding this paper. SM-B receives grant support from Janssen Research and Development and Sage Therapeutics, Inc., awarded to the University of North Carolina (Chapel Hill, NC), but these grants are outside the submitted work.

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rudzinskas, S.A., Goff, A.C., Mazzu, M.A. et al. Intrinsically dysregulated cellular stress signaling genes and gene networks in postpartum depression. Mol Psychiatry 28, 3023–3032 (2023). https://doi.org/10.1038/s41380-023-01985-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-023-01985-5

This article is cited by

Search

Quick links