Altered estradiol-dependent cellular Ca2+ homeostasis and endoplasmic reticulum stress response in Premenstrual Dysphoric Disorder

Howard J Li, Allison Goff, Sarah A Rudzinskas, Yonwoo Jung, Neelima Dubey, Jessica Hoffman, Dion Hipolito, Maria Mazzu, David R Rubinow, Peter J Schmidt, David Goldman, Howard J Li, Allison Goff, Sarah A Rudzinskas, Yonwoo Jung, Neelima Dubey, Jessica Hoffman, Dion Hipolito, Maria Mazzu, David R Rubinow, Peter J Schmidt, David Goldman

Abstract

Premenstrual Dysphoric Disorder (PMDD) is characterized by debilitating mood symptoms in the luteal phase of the menstrual cycle. Prior studies of affected women have implicated a differential response to ovarian steroids. However, the molecular basis of these patients' differential response to hormone remains poorly understood. We performed transcriptomic analyses of lymphoblastoid cell lines (LCLs) derived from women with PMDD and asymptomatic controls cultured under untreated (steroid-free), estradiol-treated (E2), and progesterone-treated (P4) conditions. Weighted gene correlation network analysis (WGCNA) of transcriptomes identified four gene modules with significant diagnosis x hormone interactions, including one enriched for neuronal functions. Next, in a gene-level analysis comparing transcriptional response to hormone across diagnoses, a generalized linear model identified 1522 genes differentially responsive to E2 (E2-DRGs). Among the top 10 E2-DRGs was a physically interacting network (NUCB1, DST, GCC2, GOLGB1) involved in endoplasmic reticulum (ER)-Golgi function. qRT-PCR validation reproduced a diagnosis x E2 interaction (F(1,24)=7.01, p = 0.014) for NUCB1, a regulator of cellular Ca2+ and ER stress. Finally, we used a thapsigargin (Tg) challenge assay to test whether E2 induces differences in Ca2+ homeostasis and ER stress response in PMDD. PMDD LCLs had a 1.36-fold decrease in Tg-induced XBP1 splicing response compared to controls, and a 1.62-fold decreased response (p = 0.005), with a diagnosis x treatment interaction (F(3,33)=3.51, p = 0.026) in the E2-exposed condition. Altered hormone-dependent in cellular Ca2+ dynamics and ER stress may contribute to the pathophysiology of PMDD.

Trial registration: ClinicalTrials.gov NCT00001259 NCT00001322.

Conflict of interest statement

DRR serves on the Clinical Advisory Board of Sage Therapeutics, and reports consulting fees and travel reimbursement from Sage Therapeutics.

© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.

Figures

Fig. 1. Functional analysis of gene modules…
Fig. 1. Functional analysis of gene modules from hormone-exposed PMDD LCLs.
a Cluster dendrogram of gene modules. In WGCNA analysis of 46 input transcriptomes, 21,746 unique gene features (“genes”) were clustered into 65 co-expression modules using gene–gene correlation patterns across all input transcriptomes. b Module eigengenes showing significant diagnosis x hormone interactions. Eigengenes (defined as the first principal component of a given module) of four modules (Brown, Pink, Turquoise, and Magenta) showed differential regulation by hormone (ANOVA interaction P < 0.05). Closed circles denote controls. Open circles denote PMDD. Note: An extreme outlier in the E2-treated control group was excluded in the eigengene plot of the Magenta module for scaling purposes. Hormone x diagnosis interaction was significant both before and after exclusion of this outlier. P-values are not adjusted for multiple comparisons. c Module membership of ESC/E(Z) complex genes. Nearly half (6/13) of ESC/E(Z) complex genes were assigned to the Turquoise module. Remaining ESC/E(Z) genes were clustered into the Blue, AntiqueWhite4 (aw4), DarkOliveGreen (dog), Cyan (cy), Green (g), and SkyBlue2 (sb2) modules. d ESC/E(Z) complex genes in the Turquoise module. Genes plotted according to module membership and strength of association with diagnosis. ESC/E(Z) complex genes (SUZ12, EED, AEBP2, HDAC2, MTF2, SIRT2) are more likely to have high module membership within the Turquoise module. e Word cloud representations of GSEA results. Top 20 GSEA results were used to generate word clouds for each differentially regulated module. Word size is proportional to frequency of word among GO terms identified by GSEA results. Modules were enriched for terms relating to MAP Kinase and GTPase signaling (Brown, top left), chromatin remodeling and proteasome function (Turquoise, top right), mitochondrial function (Pink, bottom left), and neuronal and synaptic function (Magenta, bottom right). Full, quantitative GSEA results are tabulated in Supplemental Table S3. Word clouds generated via worditout.com.
Fig. 2. Top E2-DRGs implicate ER–Golgi function…
Fig. 2. Top E2-DRGs implicate ER–Golgi function and Ca2+ homeostasis in PMDD.
a GeneMANIA visualization of select E2-DRGs. Key genes relating to ER–Golgi function and intracellular calcium homeostasis (NUCB1, DST, GCC2, GOLGB1) were selected from among the top 10 E2-DRGs and included as inputs for GeneMANIA network visualization (genemania.org, accessed January 2019). The resulting output revealed a structural network of Golgi–ER-associated genes, with physical interactions comprising 67.6% of the weighted network. b qRT-PCR validation of select E2-DRGs. NUCB1, DHCR7, PPP2R5D, and GOLGB1 were selected for qRT-PCR validation in an independent replication cohort of 13 cases and 13 controls. NUCB1 qRT-PCR results revealed a significant main effect of diagnosis (F(1,24)=4.61, p = 0.042) and interaction of diagnosis and hormone (F(1,24)=7.01, p = 0.014). In post-hoc testing, E2-treated PMDD samples had significantly reduced NUCB1 expression compared to control samples (p = 0.003). qRT-PCR results for DHCR7 did not reveal significant effects of diagnosis, hormone, or their interaction, but in multiple comparisons testing, DHCR7 expression was significantly reduced in E2-treated PMDD samples compared to E2-treated control samples (p = 0.040). No significant differences for PPP2R5D or GOLGB1 were found in qRT-PCR validation.
Fig. 3. Thapsigargin (Tg) challenge differentiates PMDD…
Fig. 3. Thapsigargin (Tg) challenge differentiates PMDD and Control LCLs.
a Experimental design of Tg challenge assays. PMDD and control LCLs were cultured in phenol red-free RPMI for 5–6 days and treated in four arms: (1) no treatment (vehicle), (2) E2 (100 nM) for 24 h, (3) Tg (0.2 uM) and CaCl2 (0.8 mM) for 4 h, and (4) E2 (100 nM) for 24 h with Tg (0.2 uM) and CaCl2 (0.8 mM) for the last 4 h. Cells were harvested and assayed for the spliced-to-unspliced ratio of XBP1 by qRT-PCR. b XBP1 S/US ratios in Tg challenge assays (by diagnosis group). Control LCLs showed an increase in XBP1 S/US ratio after Tg treatment. Pre-treatment with E2 potentiated this response. Both of these effects were blunted or absent in PMDD LCLs. A significant diagnosis x treatment interaction (ANOVA-RM F(3,33) = 3.508, p = 0.026) in XBP1 splicing response to Tg was observed. Post-hoc analysis revealed a significant difference between control and PMDD LCLs only when cells were treated with the combination of E2 + Tg (p = 0.005). E2 had a significant modulatory effect on Tg response in control cells (p = 0.007) but not in PMDD cells (p = 0.693) (see Supplemental Table S6). c S/US XBP1 ratios in Tg challenge assays (by individual cell line). Examining S/US XBP1 ratios of individual cell lines across treatment conditions shows qualitatively distinct trajectories between control and PMDD groups, most notably, the loss of E2-mediated potentiation of XBP1 splicing in PMDD LCLs. However, significant within-group heterogeneity among cell lines is also appreciated.

References

    1. Halbreich U, Borenstein J, Pearlstein T, Kahn LS. The prevalence, impairment, impact, and burden of premenstrual dysphoric disorder (PMS/PMDD) Psychoneuroendocrinology. 2003;28:1–23.
    1. Hofmeister S, Bodden S. Premenstrual syndrome and premenstrual dysphoric disorder. Am Fam Physician. 2016;94:236–240.
    1. Schiller CE, Johnson SL, Abate AC, Schmidt PJ, Rubinow DR. Reproductive steroid regulation of mood and behavior. Compr Physiol. 2016;6:1135–1160.
    1. Rubinow DR, Schmidt PJ, Roca CA. Hormone measures in reproductive endocrine-related mood disorders: diagnostic issues. Psychopharmacol Bull. 1998;34:289–290.
    1. Schmidt PJ, Nieman LK, Danaceau MA, Adams LF, Rubinow DR. Differential behavioral effects of gonadal steroids in women with and in those without premenstrual syndrome. N Engl J Med. 1998;338:209–216.
    1. Baller EB, Wei S-M, Kohn PD, Rubinow DR, Alarcón G, Schmidt PJ, et al. Abnormalities of dorsolateral prefrontal function in women with premenstrual dysphoric disorder: a multimodal neuroimaging study. Am J Psychiatry. 2013;170:305–314.
    1. Toffoletto S, Lanzenberger R, Gingnell M, Sundström-Poromaa I, Comasco E. Emotional and cognitive functional imaging of estrogen and progesterone effects in the female human brain: a systematic review. Psychoneuroendocrinology. 2014;50:28–52.
    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;22:1172–1184.
    1. American Psychiatric Association. Diagnostic and statistical manual of mental disorders (DSM-5®). American Psychiatric Pub; 2013.
    1. Oh H-M, Oh J-M, Choi S-C, Kim S-W, Han W-C, Kim T-H, et al. An efficient method for the rapid establishment of Epstein-Barr virus immortalization of human B lymphocytes. Cell Prolif. 2003;36:191–197.
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinforma. 2008;9:559.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550.
    1. Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44:W90–97.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinforma Oxf Engl. 2010;26:139–140.
    1. Warde-Farley D, Donaldson S, Comes O, Zuberi K, Badrawi R, Chao P, et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010;38. . Accessed 16 Sept 2020.
    1. Kaufman RJ. Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls. Genes Dev. 1999;13:1211–1233.
    1. Chen ZF, Paquette AJ, Anderson DJ. NRSF/REST is required in vivo for repression of multiple neuronal target genes during embryogenesis. Nat Genet. 1998;20:136–142.
    1. Einat H, Yuan P, Gould TD, Li J, Du J, Zhang L, et al. The role of the extracellular signal-regulated kinase signaling pathway in mood modulation. J Neurosci Off. J Soc Neurosci. 2003;23:7311–7316.
    1. Duric V, Banasr M, Licznerski P, Schmidt HD, Stockmeier CA, Simen AA, et al. A negative regulator of MAP kinase causes depressive behavior. Nat Med. 2010;16:1328–1332.
    1. Zhang L, Li B Shing, Zhao W, Chang YH, Ma W, Dragan M, et al. Sex-related differences in MAPKs activation in rat astrocytes: effects of estrogen on cell death. Brain Res Mol Brain Res. 2002;103:1–11.
    1. Shimamoto A, Rappeneau V. Sex-dependent mental illnesses and mitochondria. Schizophr Res. 2017;187:38–46.
    1. Allen J, Romay-Tallon R, Brymer KJ, Caruncho HJ, Kalynchuk LE. Mitochondria and mood: mitochondrial dysfunction as a key player in the manifestation of depression. Front Neurosci. 2018;12:386.
    1. Segars JH, Driggers PH. Estrogen action and cytoplasmic signaling cascades. Part I: membrane-associated signaling complexes. Trends Endocrinol Metab Tem. 2002;13:349–354.
    1. Nilsen J, Brinton RD. Mitochondria as therapeutic targets of estrogen action in the central nervous system. Curr Drug Targets CNS Neurol Disord. 2004;3:297–313.
    1. Irwin RW, Yao J, Hamilton RT, Cadenas E, Brinton RD, Nilsen J. Progesterone and estrogen regulate oxidative metabolism in brain mitochondria. Endocrinology. 2008;149:3167–3175.
    1. Milanesi E, Voinsky I, Hadar A, Srouji A, Maj C, Shekhtman T, et al. RNA sequencing of bipolar disorder lymphoblastoid cell lines implicates the neurotrophic factor HRP-3 in lithium’s clinical efficacy. World J Biol Psychiatry Off J World Fed Soc Biol Psychiatry. 2017;1–13.
    1. Duan J, Göring HHH, Sanders AR, Moy W, Freda J, Drigalenko EI, et al. Transcriptomic signatures of schizophrenia revealed by dopamine perturbation in an ex vivo model. Transl Psychiatry. 2018;8.
    1. Kittel-Schneider S, Hilscher M, Scholz C-J, Weber H, Grünewald L, Schwarz R, et al. Lithium-induced gene expression alterations in two peripheral cell models of bipolar disorder. World J Biol Psychiatry Off J World Fed Soc Biol Psychiatry. 2017;1–14.
    1. Sanders AR, Göring HHH, Duan J, Drigalenko EI, Moy W, Freda J, et al. Transcriptome study of differential expression in schizophrenia. Hum Mol Genet. 2013;22:5001–5014.
    1. Tylee DS, Espinoza AJ, Hess JL, Tahir MA, McCoy SY, Rim JK, et al. RNA sequencing of transformed lymphoblastoid cells from siblings discordant for autism spectrum disorders reveals transcriptomic and functional alterations: evidence for sex-specific effects. Autism Res. 2017;10:439–455.
    1. Jong S, de, Boks MPM, Fuller TF, Strengman E, Janson E, Kovel CGFde, et al. A gene co-expression network in whole blood of schizophrenia patients is independent of antipsychotic-use and enriched for brain-expressed genes. PLOS One. 2012;7:e39498.
    1. Kos MZ, Duan J, Sanders AR, Blondell L, Drigalenko EI, Carless MA, et al. Dopamine perturbation of gene co-expression networks reveals differential response in schizophrenia for translational machinery. Transl Psychiatry. 2018;8:278.
    1. Lin P, Yao Y, Hofmeister R, Tsien RY, Farquhar MG. Overexpression of CALNUC (Nucleobindin) increases agonist and Thapsigargin releasable Ca2+ storage in the Golgi. J Cell Biol. 1999;145:279–289.
    1. Tsukumo Y, Tomida A, Kitahara O, Nakamura Y, Asada S, Mori K, et al. Nucleobindin 1 controls the unfolded protein response by inhibiting ATF6 activation. J Biol Chem. 2007;282:29264–29272.
    1. Tulke S, Williams P, Hellysaz A, Ilegems E, Wendel M, Broberger C. Nucleobindin 1 (NUCB1) is a Golgi-resident marker of neurons. Neuroscience. 2016;314:179–188.
    1. Sundarrajan L, Blanco AM, Bertucci JI, Ramesh N, Canosa LF, Unniappan S. Nesfatin-1-like peptide encoded in nucleobindin-1 in goldfish is a novel anorexigen modulated by sex steroids, macronutrients and daily rhythm. Sci Rep. 2016;6:28377.
    1. Schalla MA, Stengel A. Current understanding of the role of Nesfatin-1. J Endocr Soc. 2018;2:1188–1206.
    1. Bloem B, Xu L, Morava E, Faludi G, Palkovits M, Roubos EW, et al. Sex-specific differences in the dynamics of cocaine- and amphetamine-regulated transcript and nesfatin-1 expressions in the midbrain of depressed suicide victims vs. controls. Neuropharmacology. 2012;62:297–303.
    1. Hofmann T, Elbelt U, Ahnis A, Rose M, Klapp BF, Stengel A. Sex-specific regulation of NUCB2/nesfatin-1: differential implication in anxiety in obese men and women. Psychoneuroendocrinology. 2015;60:130–137.
    1. Xiao M-M, Li J-B, Jiang L-L, Shao H, Wang B-L. Plasma nesfatin-1 level is associated with severity of depression in Chinese depressive patients. BMC Psychiatry 2018;18.
    1. Sönnichsen B, Lowe M, Levine T, Jämsä E, Dirac-Svejstrup B, Warren G. A role for giantin in docking COPI vesicles to Golgi membranes. J Cell Biol. 1998;140:1013–1021.
    1. Derby MC, Lieu ZZ, Brown D, Stow JL, Goud B, Gleeson PA. The trans-Golgi network golgin, GCC185, is required for endosome-to-Golgi transport and maintenance of Golgi structure. Traffic Cph Den. 2007;8:758–773.
    1. Young KG, Kothary R. Dystonin/Bpag1 is a necessary endoplasmic reticulum/nuclear envelope protein in sensory neurons. Exp Cell Res. 2008;314:2750–2761.
    1. Ryan SD, Ferrier A, Sato T, O’Meara RW, De Repentigny Y, Jiang SX, et al. Neuronal dystonin isoform 2 is a mediator of endoplasmic reticulum structure and function. Mol Biol Cell. 2012;23:553–566.
    1. Welihinda AA, Tirasophon W, Kaufman RJ. The cellular response to protein misfolding in the endoplasmic reticulum. Gene Expr. 2018;7:293–300.
    1. Carreras-Sureda A, Pihán P, Hetz C. Calcium signaling at the endoplasmic reticulum: fine-tuning stress responses. Cell Calcium. 2018;70:24–31.
    1. Bixo M, Johansson M, Timby E, Michalski L, Bäckström T. Effects of GABA active steroids in the female brain with a focus on the premenstrual dysphoric disorder. J Neuroendocrinol. 2018;30.
    1. Van Wingen GA, Van Broekhoven F, Verkes RJ, Petersson KM, Bäckström T, Buitelaar JK, et al. Progesterone selectively increases amygdala reactivity in women. Mol Psychiatry. 2008;13:325.
    1. Mansour I, Reznikoff-Etievant MF, Netter A. No evidence for the expression of the progesterone receptor on peripheral blood lymphocytes during pregnancy. Hum Reprod Oxf Engl. 1994;9:1546–1549.
    1. Scariano JK, Emery-Cohen AJ, Pickett GG, Morgan M, Simons PC, Alba F. Estrogen receptors alpha (ESR1) and beta (ESR2) are expressed in circulating human lymphocytes. J Recept Signal Transduct Res. 2008;28:285–293.
    1. Dosiou C, Hamilton AE, Pang Y, Overgaard MT, Tulac S, Dong J, et al. Expression of membrane progesterone receptors on human T lymphocytes and Jurkat cells and activation of G-proteins by progesterone. J Endocrinol. 2008;196:67–77.
    1. Pierdominici M, Maselli A, Colasanti T, Giammarioli AM, Delunardo F, Vacirca D, et al. Estrogen receptor profiles in human peripheral blood lymphocytes. Immunol Lett. 2010;132:79–85.
    1. Gross KS, Mermelstein PG. Estrogen receptor signaling through metabotropic glutamate receptors. Vitam Horm. 2020;114:211–232.
    1. Srivastava DP, Waters EM, Mermelstein PG, Kramár EA, Shors TJ, Liu F. Rapid estrogen signaling in the brain: implications for the fine-tuning of neuronal circuitry. J Soc Neurosci. 2011;31:16056–16063.
    1. Bali N, Arimoto JM, Iwata N, Lin SW, Zhao L, Brinton RD, et al. Differential responses of progesterone receptor membrane component-1 (Pgrmc1) and the classical progesterone receptor (Pgr) to 17β-estradiol and progesterone in hippocampal subregions that support synaptic remodeling and neurogenesis. Endocrinology. 2012;153:759–769.
    1. Petersen SL, Intlekofer KA, Moura-Conlon PJ, Brewer DN, Del Pino Sans J, Lopez JA. Novel progesterone receptors: neural localization and possible functions. Front Neurosci. 2013;7:164.
    1. So J, Warsh JJ, Li PP. Impaired endoplasmic reticulum stress response in B-lymphoblasts from patients with bipolar-I disorder. Biol Psychiatry. 2007;62:141–147.
    1. Hayashi A, Kasahara T, Kametani M, Toyota T, Yoshikawa T, Kato T. Aberrant endoplasmic reticulum stress response in lymphoblastoid cells from patients with bipolar disorder. Int J Neuropsychopharmacol. 2009;12:33–43.
    1. Pfaffenseller B, Wollenhaupt-Aguiar B, Fries GR, Colpo GD, Burque RK, Bristot G, et al. Impaired endoplasmic reticulum stress response in bipolar disorder: cellular evidence of illness progression. Int J Neuropsychopharmacol. 2014;17:1453–1463.
    1. Hendrick V, Altshuler LL. Recurrent mood shifts of premenstrual dysphoric disorder can be mistaken for rapid-cycling bipolar II disorder. J Clin Psychiatry. 1998;59:479–480.
    1. Margueron R, Reinberg D. The Polycomb complex PRC2 and its mark in life. Nature. 2011;469:343–349.
    1. Viré E, Brenner C, Deplus R, Blanchon L, Fraga M, Didelot C, et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature. 2006;439:871.
    1. Höglinger GU, Melhem NM, Dickson DW, Sleiman PMA, Wang L-S, Klei L, et al. Identification of common variants influencing risk of the tauopathy progressive supranuclear palsy. Nat Genet. 2011;43:699–705.
    1. Latourelle JC, Pankratz N, Dumitriu A, Wilk JB, Goldwurm S, Pezzoli G, et al. Genomewide association study for onset age in Parkinson disease. BMC Med Genet. 2009;10:98.
    1. Andruska N, Zheng X, Yang X, Helferich WG, Shapiro DJ. Anticipatory estrogen activation of the unfolded protein response is linked to cell proliferation and poor survival in estrogen receptor α-positive breast cancer. Oncogene. 2015;34:3760–3769.
    1. Zheng X, Andruska N, Yu L, Mao C, Kim JE, Livezey M, et al. Interplay between steroid hormone activation of the unfolded protein response and nuclear receptor action. Steroids. 2016;114:2–6.
    1. Nilsen J, Diaz Brinton R. Mechanism of estrogen-mediated neuroprotection: regulation of mitochondrial calcium and Bcl-2 expression. Proc Natl Acad Sci USA. 2003;100:2842–2847.
    1. Bäckström T, Bixo M, Johansson M, Nyberg S, Ossewaarde L, Ragagnin G, et al. Allopregnanolone and mood disorders. Prog Neurobiol. 2014;113:88–94.
    1. Wohleb ES, Franklin T, Iwata M, Duman RS. Integrating neuroimmune systems in the neurobiology of depression. Nat Rev Neurosci. 2016;17:497–511.
    1. Brites D, Fernandes A. Neuroinflammation and depression: microglia activation, extracellular microvesicles and microRNA dysregulation. Front Cell Neurosci. 2015;9:476.
    1. Petschner P, Gonda X, Baksa D, Eszlari N, Trivaks M, Juhasz G, et al. Genes linking mitochondrial function, cognitive impairment and depression are associated with endophenotypes serving precision medicine. Neuroscience. 2018;370:207–217.
    1. Zhang K, Kaufman RJ. From endoplasmic-reticulum stress to the inflammatory response. Nature. 2008;454:455–462.
    1. McEwen BS, Milner TA. Understanding the broad influence of sex hormones and sex differences in the brain. J Neurosci Res. 2017;95:24–39.
    1. Brinton RD. The healthy cell bias of estrogen action: mitochondrial bioenergetics and neurological implications. Trends Neurosci. 2008;31:529–537.
    1. Gold PW, Licinio J, Pavlatou MG. Pathological parainflammation and endoplasmic reticulum stress in depression: potential translational targets through the CNS insulin, klotho and PPAR-γ systems. Mol Psychiatry. 2013;18:154–165.
    1. Levy NA, Janicak PG. Calcium channel antagonists for the treatment of bipolar disorder. Bipolar Disord. 2000;2:108–119.
    1. Pollack MH, Rosenbaum JF, Hyman SE. Calcium channel blockers in psychiatry: potential for a growing role in pharmacotherapy. Psychosomatics. 1987;28:356–369.
    1. Sie L, Loong S, Tan EK. Utility of lymphoblastoid cell lines. J Neurosci. Res. 2009;87:1953–1959.
    1. Cai C, Langfelder P, Fuller TF, Oldham MC, Luo R, van den Berg LH, et al. Is human blood a good surrogate for brain tissue in transcriptional studies? BMC Genomics. 2010;11:589.
    1. Gladkevich A, Kauffman HF, Korf J. Lymphocytes as a neural probe: potential for studying psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:559–576.

Source: PubMed

3
Suscribir