In vitro model of perimenopausal depression implicates steroid metabolic and proinflammatory genes

Sarah Rudzinskas, Jessica F Hoffman, Pedro Martinez, David R Rubinow, Peter J Schmidt, David Goldman, Sarah Rudzinskas, Jessica F Hoffman, Pedro Martinez, David R Rubinow, Peter J Schmidt, David Goldman

Abstract

The estimated 20-30% of women who develop perimenopausal depression (PMD) are at an increased risk of cardiovascular and all-cause mortality. The therapeutic benefits of estradiol (E2) and symptom-provoking effects of E2-withdrawal (E2-WD) suggest that a greater sensitivity to changes in E2 at the cellular level contribute to PMD. We developed an in vitro model of PMD with lymphoblastoid cell lines (LCLs) derived from participants of a prior E2-WD clinical study. LCLs from women with past PMD (n = 8) or control women (n = 9) were cultured in three experimental conditions: at vehicle baseline, during E2 treatment, and following E2-WD. Transcriptome analysis revealed significant differences in transcript expression in PMD in all experimental conditions, and significant overlap in genes that were changed in PMD regardless of experimental condition. Of these, chemokine CXCL10, previously linked to cardiovascular disease, was upregulated in women with PMD, but most so after E2-WD (p < 1.55 × 10-5). CYP7B1, an enzyme intrinsic to DHEA metabolism, was upregulated in PMD across experimental conditions (F(1,45) = 19.93, p < 0.0001). These transcripts were further validated via qRT-PCR. Gene networks dysregulated in PMD included inflammatory response, early/late E2-response, and cholesterol homeostasis. Our results provide evidence that differential behavioral responsivity to E2-WD in PMD reflects intrinsic differences in cellular gene expression. Genes such as CXCL10, CYP7B1, and corresponding proinflammatory and steroid biosynthetic gene networks, may represent biomarkers and molecular targets for intervention in PMD. Finally, this in vitro model allows for future investigations into the mechanisms of genes and gene networks involved in the vulnerability to, and consequences of, PMD.

Trial registration: ClinicalTrials.gov NCT00060736 NCT00001231.

Conflict of interest statement

The authors have no potential conflicts of interest or financial support regarding this manuscript.

© 2020. The Author(s), under exclusive licence to Springer Nature Limited.

Figures

Figure 1 –. PMD vs. Control LCLs:…
Figure 1 –. PMD vs. Control LCLs: in vitro estradiol (E2) exposures
LCLs were subjected to the following experimental treatment time course, creating three treatment groups: baseline, E2, and E2-withdrawal, per diagnostic category (control or PMD). 100nM E2 or vehicle (DMSO) was spiked into the cultures every 24 hours for a total of 72 hours of exposure, with an additional 24 hours of vehicle for LCLs in the E2-withdrawal treatment group. Two LCL cohorts were used: Cohort 1 was a smaller, subject-matched discovery cohort in which RNA-sequencing was performed with resulting differential gene expression (DEG) and gene network analysis, E2 quantification, and ELISAs on LCL culture media. Cohort 2 was an independent replication cohort that was used for qRT-PCR. For Cohort 1, all 8 PMD LCLs were derived from women with past PMD that was demonstrated to be E2-sensitive. For Cohort 2, all 18 PMD LCLs were derived from women with past PMD that underwent a similar psychiatric screening to the Cohort 1 PMD women but were not tested for E2-sensitivity. Thus, Cohort 2 represents an independent and broader PMD phenotype.
Figure 2 –. Significant Overlap between DEGs…
Figure 2 –. Significant Overlap between DEGs in all experimental treatment groups
Venn Diagrams indicating the overlap between genes that are differentially expressed (either up- or down- regulated) (p−41, Jaccard Exact Test). B) Overlap between transcripts that are upregulated (DEGs p−40, Jaccard Exact Test). C) Overlap between transcripts that are downregulated (DEGs p−31, Jaccard Exact Test).
Figure 3 –. CXCL10, a proinflammatory chemokine,…
Figure 3 –. CXCL10, a proinflammatory chemokine, is significantly increased in LCLs from women with PMD, particularly after E2-Withdrawal
A) Two-way ANOVA of LCL gene expression (RNA-seq); diagnosis x experimental treatment. CXCL10 expression significantly increased in PMD LCLs compared to controls (F1,45=15.82, p=0.0003), and in response to E2 treatment (F2,45=6.041, p=0.0047). Sidak posthoc t-tests show a significant increase specifically after E2-withdrawal (t45=4.06, p<0.0006, ***). B, C, D) qRT-PCR validation of RNAseq CXCL10 expression confirmed significant increases in transcript expression in PMD LCLs at baseline (t(37)=2.155, p=0.038, *), E2 (t(36)=2.046, p=0.048, *) and E2-withdrawal (t(37)=2.757, p<0.009, **). E, F) ELISA on the cell culture media on the day of LCL collection from a subset (n=6 controls, n=5 PMD) of the RNA-seq samples. Levels of the chemokine CXCL10 were significantly upregulated in the cell culture supernatant of LCLs from women with PMD compared to controls at baseline (t(9)=3.438, p=0.007, **) and after E2-withdrawal (t(9)=2.984, p=0.015, *).
Figure 4 –. CYP7B1 , the gene…
Figure 4 –. CYP7B1, the gene encoding the enzyme involved in DHEA metabolism, is significantly increased in LCLs from women with PMD, and significantly correlates to decreased plasma DHEA levels
A) A two-way ANOVA of gene expression (RNA-seq) demonstrated a significant main effect of diagnosis in PMD vs controls (F1,45=19.93, p<0.0001, ***), but showed no significant main effect specifically in response to E2-treatment (F2,45=0.358, p=0.701, ns), nor a significant interaction (F2,45=0.437, p=0.648, ns). B, C, D) qRT-PCR validation of the expression levels revealed that CYP7B1 was significantly increased at baseline (t(37)=2.345, p=0.025, *), as well as after E2-withdrawal (t(35)=2.378, p=0.023, *), and trended toward significance with E2 (t(35)=2.02, p=0.051, ns). E) Available plasma DHEA levels on a subset (n=7 control, n=7 PMD) of women whose LCLs were in the RNA-sequencing cohort revealed significantly decreased DHEA in PMD women (t(13)=3.322, p=0.006, **) F) CYP7B1 E2-withdrawal RPKMs (Reads Per Kilobase per Million mapped reads from RNA-seq) demonstrated a significant negative correlation to plasma DHEA levels (r2=0.46, p<0.008). Small black dots represent the 95% confidence intervals of the linear regression line.

References

    1. Freeman EW, Sammel MD, Lin H, Nelson DB. Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry 2006; 63(4): 375–382.
    1. Freeman EW, Sammel MD, Liu L, Gracia CR, Nelson DB, Hollander L. Hormones and menopausal status as predictors of depression in women in transition to menopause. Arch Gen Psychiatry 2004; 61(1): 62–70.
    1. Cohen LS, Soares CN, Vitonis AF, Otto MW, Harlow BL. Risk for new onset of depression during the menopausal transition: the Harvard study of moods and cycles. Arch Gen Psychiatry 2006; 63(4): 385–390.
    1. Bromberger JT, Kravitz HM, Chang YF, Cyranowski JM, Brown C, Matthews KA. Major depression during and after the menopausal transition: Study of Women’s Health Across the Nation (SWAN). Psychol Med 2011; 41(9): 1879–1888.
    1. Bromberger JT, Matthews KA, Schott LL, Brockwell S, Avis NE, Kravitz HM et al. Depressive symptoms during the menopausal transition: the Study of Women’s Health Across the Nation (SWAN). J Affect Disord 2007; 103(1–3): 267–272.
    1. Wariso BA, Guerrieri GM, Thompson K, Koziol DE, Haq N, Martinez PE et al. Depression during the menopause transition: impact on quality of life, social adjustment, and disability. Arch Womens Ment Health 2017; 20(2): 273–282.
    1. Terauchi M, Hiramitsu S, Akiyoshi M, Owa Y, Kato K, Obayashi S et al. Associations among depression, anxiety and somatic symptoms in peri- and postmenopausal women. J Obstet Gynaecol Res 2013; 39(5): 1007–1013.
    1. Wassertheil-Smoller S, Shumaker S, Ockene J, Talavera GA, Greenland P, Cochrane B et al. Depression and cardiovascular sequelae in postmenopausal women. The Women’s Health Initiative (WHI). Arch Intern Med 2004; 164(3): 289–298.
    1. Burger HG. The endocrinology of the menopause. Maturitas 1996; 23(2): 129–136.
    1. Freeman EW. Depression in the menopause transition: risks in the changing hormone milieu as observed in the general population. Womens Midlife Health 2015; 1: 2.
    1. Daly RC, Danaceau MA, Rubinow DR, Schmidt PJ. Concordant restoration of ovarian function and mood in perimenopausal depression. Am J Psychiatry 2003; 160(10): 1842–1846.
    1. Tepper PG, Randolph JF Jr., McConnell DS, Crawford SL, El Khoudary SR, Joffe H et al. Trajectory clustering of estradiol and follicle-stimulating hormone during the menopausal transition among women in the Study of Women’s Health across the Nation (SWAN). J Clin Endocrinol Metab 2012; 97(8): 2872–2880.
    1. Santoro N, Brown JR, Adel T, Skurnick JH. Characterization of reproductive hormonal dynamics in the perimenopause. J Clin Endocrinol Metab 1996; 81(4): 1495–1501.
    1. Schmidt PJ, Nieman L, Danaceau MA, Tobin MB, Roca CA, Murphy JH et al. Estrogen replacement in perimenopause-related depression: a preliminary report. Am J Obstet Gynecol 2000; 183(2): 414–420.
    1. Soares CN, Almeida OP, Joffe H, Cohen LS. Efficacy of estradiol for the treatment of depressive disorders in perimenopausal women: a double-blind, randomized, placebo-controlled trial. Arch Gen Psychiatry 2001; 58(6): 529–534.
    1. Gordon JL, Rubinow DR, Eisenlohr-Moul TA, Xia K, Schmidt PJ, Girdler SS. Efficacy of Transdermal Estradiol and Micronized Progesterone in the Prevention of Depressive Symptoms in the Menopause Transition: A Randomized Clinical Trial. JAMA Psychiatry 2018; 75(2): 149–157.
    1. Ockene JK, Barad DH, Cochrane BB, Larson JC, Gass M, Wassertheil-Smoller S et al. Symptom experience after discontinuing use of estrogen plus progestin. JAMA 2005; 294(2): 183–193.
    1. Ness J, Aronow WS, Beck G. Menopausal symptoms after cessation of hormone replacement therapy. Maturitas 2006; 53(3): 356–361.
    1. Schmidt PJ, Murphy JH, Haq N, Danaceau MA, St Clair L. Basal plasma hormone levels in depressed perimenopausal women. Psychoneuroendocrinology 2002; 27(8): 907–920.
    1. Schmidt PJ, Ben Dor R, Martinez PE, Guerrieri GM, Harsh VL, Thompson K et al. Effects of Estradiol Withdrawal on Mood in Women With Past Perimenopausal Depression: A Randomized Clinical Trial. JAMA Psychiatry 2015; 72(7): 714–726.
    1. 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(8): 1172–1184.
    1. Steinberg EM, Rubinow DR, Bartko JJ, Fortinsky PM, Haq N, Thompson K et al. A cross-sectional evaluation of perimenopausal depression. J Clin Psychiatry 2008; 69(6): 973–980.
    1. 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(4): 191–197.
    1. 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 U S A 1986; 83(8): 2496–2500.
    1. Welshons WV, Wolf MF, Murphy CS, Jordan VC. Estrogenic activity of phenol red. Mol Cell Endocrinol 1988; 57(3): 169–178.
    1. 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. In Vitro 1976; 12(1): 23–30.
    1. Benjamini Y, Hochberg Y. Controlling the False Discovery Rate - a Practical and Powerful Approach to Multiple Testing. Journal of the Royal Statistical Society Series B-Statistical Methodology 1995; 57(1): 289–300.
    1. Wang M, Zhao Y, Zhang B. Efficient Test and Visualization of Multi-Set Intersections. Sci Rep 2015; 5: 16923.
    1. Turner SD. qqman: an R package for visualizing GWAS results using Q-Q and manhattan plots. bioRxiv 2014: 005165.
    1. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 2015; 1(6): 417–425.
    1. 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 Bioinformatics 2013; 14: 128.
    1. Duan Q, Flynn C, Niepel M, Hafner M, Muhlich JL, Fernandez NF et al. LINCS Canvas Browser: interactive web app to query, browse and interrogate LINCS L1000 gene expression signatures. Nucleic Acids Res 2014; 42: W449–460.
    1. Menard C, Hodes GE, Russo SJ. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience 2016; 321: 138–162.
    1. Hodes GE, Kana V, Menard C, Merad M, Russo SJ. Neuroimmune mechanisms of depression. Nat Neurosci 2015; 18(10): 1386–1393.
    1. Altara R, Mallat Z, Booz GW, Zouein FA. The CXCL10/CXCR3 Axis and Cardiac Inflammation: Implications for Immunotherapy to Treat Infectious and Noninfectious Diseases of the Heart. J Immunol Res 2016; 2016: 4396368.
    1. Altara R, Manca M, Hessel MH, Gu Y, van Vark LC, Akkerhuis KM et al. CXCL10 Is a Circulating Inflammatory Marker in Patients with Advanced Heart Failure: a Pilot Study. J Cardiovasc Transl Res 2016; 9(4): 302–314.
    1. van den Borne P, Quax PH, Hoefer IE, Pasterkamp G. The multifaceted functions of CXCL10 in cardiovascular disease. Biomed Res Int 2014; 2014: 893106.
    1. Le Thuc O, Stobbe K, Cansell C, Nahon JL, Blondeau N, Rovere C. Hypothalamic Inflammation and Energy Balance Disruptions: Spotlight on Chemokines. Front Endocrinol (Lausanne) 2017; 8: 197.
    1. Rotondi M, Chiovato L, Romagnani S, Serio M, Romagnani P. Role of chemokines in endocrine autoimmune diseases. Endocr Rev 2007; 28(5): 492–520.
    1. Bronger H, Kraeft S, Schwarz-Boeger U, Cerny C, Stockel A, Avril S et al. Modulation of CXCR3 ligand secretion by prostaglandin E2 and cyclooxygenase inhibitors in human breast cancer. Breast Cancer Res 2012; 14(1): R30.
    1. Koten K, Hirohata S, Miyoshi T, Ogawa H, Usui S, Shinohata R et al. Serum interferon-gamma-inducible protein 10 level was increased in myocardial infarction patients, and negatively correlated with infarct size. Clin Biochem 2008; 41(1–2): 30–37.
    1. Xanthou G, Duchesnes CE, Williams TJ, Pease JE. CCR3 functional responses are regulated by both CXCR3 and its ligands CXCL9, CXCL10 and CXCL11. Eur J Immunol 2003; 33(8): 2241–2250.
    1. Evans J, Salamonsen LA. Decidualized human endometrial stromal cells are sensors of hormone withdrawal in the menstrual inflammatory cascade. Biol Reprod 2014; 90(1): 14.
    1. Kanda N, Watanabe S. 17beta-estradiol inhibits the production of interferon-induced protein of 10 kDa by human keratinocytes. J Invest Dermatol 2003; 120(3): 411–419.
    1. Cerciat M, Unkila M, Garcia-Segura LM, Arevalo MA. Selective estrogen receptor modulators decrease the production of interleukin-6 and interferon-gamma-inducible protein-10 by astrocytes exposed to inflammatory challenge in vitro. Glia 2010; 58(1): 93–102.
    1. Sentman CL, Meadows SK, Wira CR, Eriksson M. Recruitment of uterine NK cells: induction of CXC chemokine ligands 10 and 11 in human endometrium by estradiol and progesterone. J Immunol 2004; 173(11): 6760–6766.
    1. Muller C, Hennebert O, Morfin R. The native anti-glucocorticoid paradigm. J Steroid Biochem Mol Biol 2006; 100(1–3): 95–105.
    1. Rose KA, Stapleton G, Dott K, Kieny MP, Best R, Schwarz M et al. Cyp7b, a novel brain cytochrome P450, catalyzes the synthesis of neurosteroids 7alpha-hydroxy dehydroepiandrosterone and 7alpha-hydroxy pregnenolone. Proc Natl Acad Sci U S A 1997; 94(10): 4925–4930.
    1. Pak TR, Chung WC, Hinds LR, Handa RJ. Estrogen receptor-beta mediates dihydrotestosterone-induced stimulation of the arginine vasopressin promoter in neuronal cells. Endocrinology 2007; 148(7): 3371–3382.
    1. Handa RJ, Pak TR, Kudwa AE, Lund TD, Hinds L. An alternate pathway for androgen regulation of brain function: activation of estrogen receptor beta by the metabolite of dihydrotestosterone, 5alpha-androstane-3beta,17beta-diol. Horm Behav 2008; 53(5): 741–752.
    1. Maninger N, Wolkowitz OM, Reus VI, Epel ES, Mellon SH. Neurobiological and neuropsychiatric effects of dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEAS). Front Neuroendocrinol 2009; 30(1): 65–91.
    1. Schmidt PJ, Daly RC, Bloch M, Smith MJ, Danaceau MA, St Clair LS et al. Dehydroepiandrosterone monotherapy in midlife-onset major and minor depression. Arch Gen Psychiatry 2005; 62(2): 154–162.
    1. Ben Dor R, Marx CE, Shampine LJ, Rubinow DR, Schmidt PJ. DHEA metabolism to the neurosteroid androsterone: a possible mechanism of DHEA’s antidepressant action. Psychopharmacology (Berl) 2015; 232(18): 3375–3383.
    1. Tang W, Eggertsen G, Chiang JY, Norlin M. Estrogen-mediated regulation of CYP7B1: a possible role for controlling DHEA levels in human tissues. J Steroid Biochem Mol Biol 2006; 100(1–3): 42–51.
    1. Morrison MF, Freeman EW, Lin H, Sammel MD. Higher DHEA-S (dehydroepiandrosterone sulfate) levels are associated with depressive symptoms during the menopausal transition: results from the PENN Ovarian Aging Study. Arch Womens Ment Health 2011; 14(5): 375–382.
    1. Martin C, Bean R, Rose K, Habib F, Seckl J. cyp7b1 catalyses the 7alpha-hydroxylation of dehydroepiandrosterone and 25-hydroxycholesterol in rat prostate. Biochem J 2001; 355(Pt 2): 509–515.
    1. Martin C, Ross M, Chapman KE, Andrew R, Bollina P, Seckl JR et al. CYP7B generates a selective estrogen receptor beta agonist in human prostate. J Clin Endocrinol Metab 2004; 89(6): 2928–2935.
    1. Howard DM, Adams MJ, Clarke TK, Hafferty JD, Gibson J, Shirali M et al. Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat Neurosci 2019; 22(3): 343–352.
    1. Nagel M, Jansen PR, Stringer S, Watanabe K, de Leeuw CA, Bryois J et al. Meta-analysis of genome-wide association studies for neuroticism in 449,484 individuals identifies novel genetic loci and pathways. Nat Genet 2018; 50(7): 920–927.
    1. Baselmans BML, Jansen R, Ip HF, van Dongen J, Abdellaoui A, van de Weijer MP et al. Multivariate genome-wide analyses of the well-being spectrum. Nat Genet 2019; 51(3): 445–451.
    1. Slowik A, Lammerding L, Hoffmann S, Beyer C. Brain inflammasomes in stroke and depressive disorders: Regulation by oestrogen. J Neuroendocrinol 2018; 30(2): e12482. 10.1111/jne.12482.
    1. Pfeilschifter J, Koditz R, Pfohl M, Schatz H. Changes in proinflammatory cytokine activity after menopause. Endocr Rev 2002; 23(1): 90–119.
    1. Dulos J, Verbraak E, Bagchus WM, Boots AM, Kaptein A. Severity of murine collagen-induced arthritis correlates with increased CYP7B activity: enhancement of dehydroepiandrosterone metabolism by interleukin-1beta. Arthritis Rheum 2004; 50(10): 3346–3353.
    1. Zare N, Khalifeh S, Khodagholi F, Shahamati SZ, Motamedi F, Maghsoudi N. Geldanamycin Reduces Abeta-Associated Anxiety and Depression, Concurrent with Autophagy Provocation. J Mol Neurosci 2015; 57(3): 317–324.
    1. Binder EB. The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology 2009; 34 Suppl 1: S186–195.
    1. Criado-Marrero M, Rein T, Binder EB, Porter JT, Koren J, 3rd, Blair LJ. Hsp90 and FKBP51: complex regulators of psychiatric diseases. Philos Trans R Soc Lond B Biol Sci 2018; 373(1738).
    1. Tsai YC, Leu SY, Chen SY, Kung CW, Lee YM, Liu YP et al. 17-DMAG, an Hsp90 inhibitor, ameliorates ovariectomy-induced obesity in rats. Life Sci 2019; 232: 116672.
    1. Dome P, Tombor L, Lazary J, Gonda X, Rihmer Z. Natural health products, dietary minerals and over-the-counter medications as add-on therapies to antidepressants in the treatment of major depressive disorder: a review. Brain Res Bull 2019; 146: 51–78.
    1. Abd-Rabo MM, Georgy GS, Saied NM, Hassan WA. Involvement of the serotonergic system and neuroplasticity in the antidepressant effect of curcumin in ovariectomized rats: Comparison with oestradiol and fluoxetine. Phytother Res 2019; 33(2): 387–396.
    1. Bhat A, Mahalakshmi AM, Ray B, Tuladhar S, Hediyal TA, Manthiannem E et al. Benefits of curcumin in brain disorders. Biofactors 2019; 45(5): 666–689.
    1. Miodownik C, Lerner V, Kudkaeva N, Lerner PP, Pashinian A, Bersudsky Y et al. Curcumin as Add-On to Antipsychotic Treatment in Patients With Chronic Schizophrenia: A Randomized, Double-Blind, Placebo-Controlled Study. Clin Neuropharmacol 2019; 42(4): 117–122.
    1. Ng QX, Koh SSH, Chan HW, Ho CYX. Clinical Use of Curcumin in Depression: A Meta-Analysis. J Am Med Dir Assoc 2017; 18(6): 503–508.
    1. Caliskan M, Cusanovich DA, Ober C, Gilad Y. The effects of EBV transformation on gene expression levels and methylation profiles. Hum Mol Genet 2011; 20(8): 1643–1652.
    1. Richards M, Rubinow DR, Daly RC, Schmidt PJ. Premenstrual symptoms and perimenopausal depression. Am J Psychiatry 2006; 163(1): 133–137.
    1. Honigberg MC, Zekavat SM, Aragam K, Finneran P, Klarin D, Bhatt DL et al. Association of Premature Natural and Surgical Menopause With Incident Cardiovascular Disease. JAMA 2019. 10.1001/jama.2019.19191. Online ahead of print.

Source: PubMed

3
Iratkozz fel