Transcriptome-wide mega-analyses reveal joint dysregulation of immunologic genes and transcription regulators in brain and blood in schizophrenia

Jonathan L Hess, Daniel S Tylee, Rahul Barve, Simone de Jong, Roel A Ophoff, Nishantha Kumarasinghe, Paul Tooney, Ulrich Schall, Erin Gardiner, Natalie Jane Beveridge, Rodney J Scott, Surangi Yasawardene, Antionette Perera, Jayan Mendis, Vaughan Carr, Brian Kelly, Murray Cairns, Neurobehavioural Genetics Unit, Ming T Tsuang, Stephen J Glatt, Jonathan L Hess, Daniel S Tylee, Rahul Barve, Simone de Jong, Roel A Ophoff, Nishantha Kumarasinghe, Paul Tooney, Ulrich Schall, Erin Gardiner, Natalie Jane Beveridge, Rodney J Scott, Surangi Yasawardene, Antionette Perera, Jayan Mendis, Vaughan Carr, Brian Kelly, Murray Cairns, Neurobehavioural Genetics Unit, Ming T Tsuang, Stephen J Glatt

Abstract

The application of microarray technology in schizophrenia research was heralded as paradigm-shifting, as it allowed for high-throughput assessment of cell and tissue function. This technology was widely adopted, initially in studies of postmortem brain tissue, and later in studies of peripheral blood. The collective body of schizophrenia microarray literature contains apparent inconsistencies between studies, with failures to replicate top hits, in part due to small sample sizes, cohort-specific effects, differences in array types, and other confounders. In an attempt to summarize existing studies of schizophrenia cases and non-related comparison subjects, we performed two mega-analyses of a combined set of microarray data from postmortem prefrontal cortices (n=315) and from ex-vivo blood tissues (n=578). We adjusted regression models per gene to remove non-significant covariates, providing best-estimates of transcripts dysregulated in schizophrenia. We also examined dysregulation of functionally related gene sets and gene co-expression modules, and assessed enrichment of cell types and genetic risk factors. The identities of the most significantly dysregulated genes were largely distinct for each tissue, but the findings indicated common emergent biological functions (e.g. immunity) and regulatory factors (e.g., predicted targets of transcription factors and miRNA species across tissues). Our network-based analyses converged upon similar patterns of heightened innate immune gene expression in both brain and blood in schizophrenia. We also constructed generalizable machine-learning classifiers using the blood-based microarray data. Our study provides an informative atlas for future pathophysiologic and biomarker studies of schizophrenia.

Keywords: Blood; Brain; Gene expression; Innate immunity; Random forests; Schizophrenia; Support vector machine; Transcriptome.

Conflict of interest statement

The authors have no conflicts of interest to declare.

Published by Elsevier B.V.

Figures

Figure 1. Cross-Tissue Comparison of Significantly Dysregulated…
Figure 1. Cross-Tissue Comparison of Significantly Dysregulated Gene Sets (Bonferroni p <0.05)
The results of the permutation-based gene-set analyses are shown (Panel A), highlighting gene sets that were significantly dysregulated in both tissues. A total of 745 gene sets were dysregulated (out of 9254 tested) based on single-gene test statistics from the brain mega-analysis. A total of 526 gene sets were dysregulated (out of 9256 tested) based on single-gene test statistics from the blood mega-analysis. For the purpose of cross-tissue comparison, gene sets with either an absolute effect (i.e., all genes in the target set) or a mixed effect (i.e., a subset of the genes in the target set) were considered to be directionally dysregulated. Among the dysregulated gene sets, 263 were common to both tissues (Bonferroni-corrected hypergeometric p < 1.7×10−158, and among these, 255 showed evidence of up-regulation across tissues (p < 2.8×10−198), while only 4 showed evidence of down-regulation across tissues (p < 4.6×10−4). Detailed methods for gene set analysis and the reference databases can be found in the Supplementary Methods. Among dysregulated gene sets corresponding to the Molecular Signature Database’s (Broad Institute) Hallmark category, we assessed whether SZ cases showed significant evidence for heterogeneity using a previously developed approach described in the Supplementary Methods. Within the brain data, we observed significant 2-group clustering of SZ cases based on the expression values corresponding to 5 gene sets showing a main-effect of upregulation in SZ (Panel B); cases are shown such that the individuals belonging to the cluster driving the up-regulation effect are depicted in red. These results suggest that different SZ cases contribute to the observed dysregulation in distinct biological pathways. Within the blood data, we observed significant 2-group clustering for a single gene set which showed a main-effect of down-regulation among SZ cases (Panel C).
Figure 2. Cross-Tissue Comparison of Significantly Dysregulated…
Figure 2. Cross-Tissue Comparison of Significantly Dysregulated Genes (FDR q <0.05)
(A) Based on the mega-analyses, we identified the most significantly dysregulated genes in brain (n = 92) and blood (n = 2238) at a relatively conservative threshold (FDR q < .10). A total of 10 genes were common to both lists; 7 genes were coordinately up-regulated and 1 gene was coordinately down-regulated across tissues. The degree of cross-tissue overlap for each of the displayed intersections was non-significant based on hypergeometric test statistics.
Figure 3. Co-expression Modules Nominally Associated with…
Figure 3. Co-expression Modules Nominally Associated with SZ in Brain (p < 0.05)
Comparison of module eigengene expression values (unadjusted for covariates) between SZ cases and unaffected comparisons within the “green” and “salmon” co-expression modules identified by the WGCNA R package (A and E, respectively), which were nominally associated with SZ from linear mixed model (uncorrected p < 0.05). We cross-referenced the set of dysregulated genes identified in the brain mega-analysis (q < .1) with the top 25 genes in each module ranked by intramodular connectivity (overlaps denoted by asterisk *) (B and F). In panels B and F, the top 5 “hub” genes are found in the innermost circle. Modules were biologically characterized by testing for enrichment of brain cell-type signatures (C and G) and annotations by a pathway-based approach (D and H). In panels D and H, annotations that surpassed a BH p < 0.05 (cutoff depicted by vertical dotted line) from hypergeometric tests are shown (represented as −log10[P]).
Figure 4. A Co-expression Module that was…
Figure 4. A Co-expression Module that was Significantly Associated with SZ in Blood (BH p < 0.05)
Comparison of module eigengene expression values (unadjusted for covariates) between SZ cases and unaffected comparisons within the “darkolivegreen” co-expression module identified by the WGCNA R package (A), which was significantly associated with SZ based on linear mixed model test (BH p < 4.4×10−6). We cross-referenced the set of dysregulated genes identified in the blood mega-analysis (q < .1) with the top 25 genes ranked by intramodular connectivity in this module (overlaps denoted by asterisk *) (B). In panel B, the top 5 “hub” genes are found in the innermost circle. To biological characterize this module, a pathway-based approach was used to test for significant enrichment biological annotations mapping to “darkolivegreen” genes (C). In panel C, annotations that surpassed a BH p < 0.05 (cutoff depicted by vertical dotted line) from hypergeometric tests are shown (represented as −log10[P]).

References

    1. Abbas AR, Wolslegel K, Seshasayee D, Modrusan Z, Clark HF. Deconvolution of blood microarray data identifies cellular activation patterns in systemic lupus erythematosus. PLoS One. 2009;4(7):e6098.
    1. Åberg K, Saetre P, Lindholm E, Ekholm B, Pettersson U, Adolfsson R, Jazin E. Human QKI, a new candidate gene for schizophrenia involved in myelination. Am. J. Med. Genet. - Neuropsychiatr. Genet. 2006;141 B(1):84–90.
    1. Arion D, Corradi JP, Tang S, Datta D, Boothe F, He a, Cacace a M, Zaczek R, Albright CF, Tseng G, Lewis Da. Distinctive transcriptome alterations of prefrontal pyramidal neurons in schizophrenia and schizoaffective disorder. Mol. Psychiatry. 2015;20(11):1397–1405.
    1. Arnedo J, Svrakic DM, Del Val C, Romero-Zaliz R, Hernendez-Cuervo H, Fanous AH, Pato MT, Pato CN, De Erausquin GA, Robert Cloninger C, Zwir I. Uncovering the hidden risk architecture of the schizophrenias: Confirmation in three independent genome-wide association studies. Am. J. Psychiatry. 2015;172(2):139–153.
    1. Bergon A, Belzeaux R, Comte M, Pelletier F, Herve M, Gardiner EJ, Beveridge NJ, Liu B, Carr V, Scott RJ, Kelly B, Cairns MJ, Kumarasinghe N, Schall U, Blin O, Boucraut J, Tooney PA, Fakra E, Ibrahim EC. CX3CR1 is dysregulated in blood and brain from schizophrenia patients. Schizophr. Res. 2015;168(1–2):434–443.
    1. Cotter D, Mackay D, Chana G, Beasley C, Landau S, Everall IP. Reduced neuronal size and glial cell density in area 9 of the dorsolateral prefrontal cortex in subjects with major depressive disorder. Cereb. Cortex. 2002;12(4):386–394.
    1. Dean B. Understanding the role of inflammatory-related pathways in the pathophysiology and treatment of psychiatric disorders: evidence from human peripheral studies and CNS studies. Int. J. Neuropsychopharmacol. 2011;14(7):997–1012.
    1. Devor EJ, Waziri R. A familial/genetic study of plasma serine and glycine concentrations. Biol. Psychiatry. 1993;34(4):221–225.
    1. Dougherty JD, Schmidt EF, Nakajima M, Heintz N. Analytical approaches to RNA profiling data for the identification of genes enriched in specific cells. Nucleic Acids Res. 2010;38(13):4218–4230.
    1. Duncan LE, Holmans PA, Lee PH, O’Dushlaine CT, Kirby AW, Smoller JW, Öngür D, Cohen BM. Pathway analyses implicate glial cells in schizophrenia. PLoS One. 2014;9(2):e89441.
    1. Eaton WW, Byrne M, Ewald H, Mors O, Chen C-Y, Agerbo E, Mortensen PB. Association of schizophrenia and autoimmune diseases: linkage of Danish national registers. Am. J. Psychiatry. 2006;163(3):521–528.
    1. Ellman LM, Deicken RF, Vinogradov S, Kremen WS, Poole JH, Kern DM, Tsai WY, Schaefer CA, Brown AS. Structural brain alterations in schizophrenia following fetal exposure to the inflammatory cytokine interleukin-8. Schizophr. Res. 2010;121(1–3):46–54.
    1. Fillman SG, Cloonan N, Catts VS, Miller LC, Wong J, McCrossin T, Cairns M, Weickert CS. Increased inflammatory markers identified in the dorsolateral prefrontal cortex of individuals with schizophrenia. Mol. Psychiatry. 2013;18(2):206–214.
    1. Fineberg AM, Ellman LM. Inflammatory cytokines and neurological and neurocognitive alterations in the course of schizophrenia. Biol. Psychiatry. 2013;73(10):951–966.
    1. Fung SJ, Joshi D, Fillman SG, Weickert CS. High white matter neuron density with elevated cortical cytokine expression in schizophrenia. Biol. Psychiatry. 2014;75(4):e5–e7.
    1. Glatt SJ, Tsuang MT, Winn M, Chandler SD, Collins M, Lopez L, Weinfeld M, Carter C, Schork N, Pierce K, Courchesne E. Blood-based gene expression signatures of infants and toddlers with autism. J. Am. Acad. Child Adolesc. Psychiatry. 2012;51(9):934–944.
    1. Glatt SJ, Tylee DS, Chandler SD, Pazol J, Nievergelt CM, Woelk CH, Baker DG, Lohr JB, Kremen WS, Litz BT, Tsuang MT. Blood-based gene-expression predictors of PTSD risk and resilience among deployed marines: a pilot study. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 2013;162B(4):313–326.
    1. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009;4(1):44–57.
    1. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4(2):249–264.
    1. Kumarasinghe N, Beveridge NJ, Gardiner E, Scott RJ, Yasawardene S, Perera A, Mendis J, Suriyakumara K, Schall U, Tooney PA. Gene expression profiling in treatment-naive schizophrenia patients identifies abnormalities in biological pathways involving AKT1 that are corrected by antipsychotic medication. Int. J. Neuropsychopharmacol. 2013;16(7):1483–1503.
    1. Langfelder P, Luo R, Oldham MC, Horvath S. Is my network module preserved and reproducible? PLoS Comput. Biol. 2011;7(1):e1001057.
    1. Lottaz C, Toedling J, Spang R. Annotation-based distance measures for patient subgroup discovery in clinical microarray studies. Bioinformatics. 2007;23(17):2256–2264.
    1. Maino K, Gruber R, Riedel M, Seitz N, Schwarz M, Müller N. T- and B-lymphocytes in patients with schizophrenia in acute psychotic episode and the course of the treatment. Psychiatry Res. 2007;152(2–3):173–180.
    1. Mamdani F, Berlim MT, Beaulieu M-M, Labbe A, Merette C, Turecki G. Gene expression biomarkers of response to citalopram treatment in major depressive disorder. Transl. Psychiatry. 2011;1:e13.
    1. Middleton FA, Mirnics K, Pierri JN, Lewis DA, Levitt P. Gene expression profiling reveals alterations of specific metabolic pathways in schizophrenia. J. Neurosci. 2002;22(7):2718–2729.
    1. Miller BJ, Buckley P, Seabolt W, Mellor A, Kirkpatrick B. Meta-analysis of cytokine alterations in schizophrenia: Clinical status and antipsychotic effects. Biol. Psychiatry. 2011;70(7):663–671.
    1. Mirnics K, Levitt P, Lewis DA. Critical appraisal of DNA microarrays in psychiatric genomics. Biol. Psychiatry. 2006;60(2):163–176.
    1. Mistry M, Gillis J, Pavlidis P. Meta-analysis of gene coexpression networks in the post-mortem prefrontal cortex of patients with schizophrenia and unaffected controls. BMC Neurosci. 2013a;14:105.
    1. Mistry M, Gillis J, Pavlidis P. Genome-wide expression profiling of schizophrenia using a large combined cohort. Mol. Psychiatry. 2013b;18(2):215–225.
    1. Mistry M, Pavlidis P. A cross-laboratory comparison of expression profiling data from normal human postmortem brain. Neuroscience. 2010;167(2):384–395.
    1. Müller N, Schwarz MJ. Immune System and Schizophrenia. Curr. Immunol. Rev. 2010;6(3):213–220.
    1. National Center for Biotechnology Information [NCBI] eQTL Browser. US Natl. Libr. Med. .
    1. Pérez-Santiago J, Diez-Alarcia R, Callado LF, Zhang JX, Chana G, White CH, Glatt SJ, Tsuang MT, Everall IP, Meana JJ, Woelk CH. A combined analysis of microarray gene expression studies of the human prefrontal cortex identifies genes implicated in schizophrenia. J. Psychiatr. Res. 2012;46(11):1464–1474.
    1. Potvin S, Stip E, Sepehry AA, Gendron A, Bah R, Kouassi E. Inflammatory Cytokine Alterations in Schizophrenia: A Systematic Quantitative Review. Biol. Psychiatry. 2008;63(8):801–808.
    1. Rao JS, Kim H-W, Harry GJ, Rapoport SI, Reese EA. Increased neuroinflammatory and arachidonic acid cascade markers, and reduced synaptic proteins, in the postmortem frontal cortex from schizophrenia patients. Schizophr. Res. 2013;147(1):24–31.
    1. Ripke S, Neale BM, Corvin A, Walters JTR, Farh K-H, Holmans PA, Lee P, Bulik-Sullivan B, Collier DA, Huang H, Pers TH, Agartz I, Agerbo E, Albus M, Alexander M, Amin F, Bacanu SA, Begemann M, Belliveau RA, Jr, Bene J, Bergen SE, Bevilacqua E, Bigdeli TB, Black DW, Bruggeman R, Buccola NG, Buckner RL, Byerley W, Cahn W, Cai G, Campion D, Cantor RM, Carr VJ, Carrera N, Catts SV, Chambert KD, Chan RCK, Chen RYL, Chen EYH, Cheng W, Cheung EFC, Ann Chong S, Robert Cloninger C, Cohen D, Cohen N, Cormican P, Craddock N, Crowley JJ, Curtis D, Davidson M, Davis KL, Degenhardt F, Del Favero J, Demontis D, Dikeos D, Dinan T, Djurovic S, Donohoe G, Drapeau E, Duan J, Dudbridge F, Durmishi N, Eichhammer P, Eriksson J, Escott-Price V, Essioux L, Fanous AH, Farrell MS, Frank J, Franke L, Freedman R, Freimer NB, Friedl M, Friedman JI, Fromer M, Genovese G, Georgieva L, Giegling I, Giusti-Rodríguez P, Godard S, Goldstein JI, Golimbet V, Gopal S, Gratten J, de Haan L, Hammer C, Hamshere ML, Hansen M, Hansen T, Haroutunian V, Hartmann AM, Henskens FA, Herms S, Hirschhorn JN, Hoffmann P, Hofman A, Hollegaard MV, Hougaard DM, Ikeda M, Joa I, Julià A, Kahn RS, Kalaydjieva L, Karachanak-Yankova S, Karjalainen J, Kavanagh D, Keller MC, Kennedy JL, Khrunin A, Kim Y, Klovins J, Knowles JA, Konte B, Kucinskas V, Ausrele Kucinskiene Z, Kuzelova-Ptackova H, Kähler AK, Laurent C, Lee Chee Keong J, Hong Lee S, Legge SE, Lerer B, Li M, Li T, Liang K-Y, Lieberman J, Limborska S, Loughland CM, Lubinski J, Lönnqvist J, Macek Jr M, Magnusson PKE, Maher BS, Maier W, Mallet J, Marsal S, Mattheisen M, Mattingsdal M, McCarley RW, McDonald C, McIntosh AM, Meier S, Meijer CJ, Melegh B, Melle I, Mesholam-Gately RI, Metspalu A, Michie PT, Milani L, Milanova V, Mokrab Y, Morris DW, Mors O, Murphy KC, Murray RM, Myin-Germeys I, Müller-Myhsok B, Nelis M, Nenadic I, Nertney DA, Nestadt G, Nicodemus KK, Nikitina-Zake L, Nisenbaum L, Nordin A, O’Callaghan E, O’Dushlaine C, O’Neill FA, Oh S-Y, Olincy A, Olsen L, Van Os J, Endophenotypes International Consortium P, Pantelis C, Papadimitriou GN, Papiol S, Parkhomenko E, Pato MT, Paunio T, Pejovic-Milovancevic M, Perkins DO, Pietiläinen O, Pimm J, Pocklington AJ, Powell J, Price A, Pulver AE, Purcell SM, Quested D, Rasmussen HB, Reichenberg A, Reimers MA, Richards AL, Roffman JL, Roussos P, Ruderfer DM, Salomaa V, Sanders AR, Schall U, Schubert CR, Schulze TG, Schwab SG, Scolnick EM, Scott RJ, Seidman LJ, Shi J, Sigurdsson E, Silagadze T, Silverman JM, Sim K, Slominsky P, Smoller JW, So H-C, Spencer CA, Stahl EA, Stefansson H, Steinberg S, Stogmann E, Straub RE, Strengman E, Strohmaier J, Scott Stroup T, Subramaniam M, Suvisaari J, Svrakic DM, Szatkiewicz JP, Söderman E, Thirumalai S, Toncheva D, Tosato S, Veijola J, Waddington J, Walsh D, Wang D, Wang Q, Webb BT, Weiser M, Wildenauer DB, Williams NM, Williams S, Witt SH, Wolen AR, Wong EHM, Wormley BK, Simon Xi H, Zai CC, Zheng X, Zimprich F, Wray NR, Stefansson K, Visscher PM, Trust Case-Control Consortium W, Adolfsson R, Andreassen OA, Blackwood DHR, Bramon E, Buxbaum JD, Børglum AD, Cichon S, Darvasi A, Domenici E, Ehrenreich H, Esko T, Gejman PV, Gill M, Gurling H, Hultman CM, Iwata N, Jablensky AV, Jönsson EG, Kendler KS, Kirov G, Knight J, Lencz T, Levinson DF, Li QS, Liu J, Malhotra AK, McCarroll SA, McQuillin A, Moran JL, Mortensen PB, Mowry BJ, Nöthen MM, Ophoff RA, Owen MJ, Palotie A, Pato CN, Petryshen TL, Posthuma D, Rietschel M, Riley BP, Rujescu D, Sham PC, Sklar P, St Clair D, Weinberger DR, Wendland JR, Werge T, Daly MJ, Sullivan PF, O’Donovan MC. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511(7510):421–427.
    1. Roussos P, Katsel P, Davis KL, Giakoumaki SG, Lencz T, Malhotra AK, Siever LJ, Bitsios P, Haroutunian V. Convergent findings for abnormalities of the NF-κB signaling pathway in schizophrenia. Neuropsychopharmacology. 2013;38(3):533–539.
    1. Sanders AR, Göring HHH, Duan J, Drigalenko EI, Moy W, Freda J, He D, Shi J, Gejman PV. Transcriptome study of differential expression in schizophrenia. Hum. Mol. Genet. 2013;22(24):5001–5014.
    1. Schwieler L, Larsson MK, Skogh E, Kegel ME, Orhan F, Abdelmoaty S, Finn A, Bhat M, Samuelsson M, Lundberg K, Dahl M-L, Sellgren C, Schuppe-Koistinen I, Svensson C, Erhardt S, Engberg G. Increased levels of IL-6 in the cerebrospinal fluid of patients with chronic schizophrenia--significance for activation of the kynurenine pathway. J. Psychiatry Neurosci. 2015;40(2):126–133.
    1. Seifuddin F, Pirooznia M, Judy JT, Goes FS, Potash JB, Zandi PP. Systematic review of genome-wide gene expression studies of bipolar disorder. BMC Psychiatry. 2013;13:213.
    1. Sekar A, Bialas AR, de Rivera H, Davis A, Hammond TR, Kamitaki N, Tooley K, Presumey J, Baum M, Van Doren V, Genovese G, Rose SA, Handsaker RE, Daly MJ, Carroll MC, Stevens B, McCarroll SA. Schizophrenia risk from complex variation of complement component 4. Nature. 2016;530(7589):177–183.
    1. Steiner J, Jacobs R, Panteli B, Brauner M, Schiltz K, Bahn S, Herberth M, Westphal S, Gos T, Walter M, Bernstein H-G, Myint AM, Bogerts B. Acute schizophrenia is accompanied by reduced T cell and increased B cell immunity. Eur. Arch. Psychiatry Clin. Neurosci. 2010;260(7):509–518.
    1. Storey JD. The positive false discovery rate: a Bayesian interpretation and the q -value. Ann. Stat. 2003;31(6):2013–2035.
    1. Sugino H, Futamura T, Mitsumoto Y, Maeda K, Marunaka Y. Atypical antipsychotics suppress production of proinflammatory cytokines and up-regulate interleukin-10 in lipopolysaccharide-treated mice. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2009;33(2):303–307.
    1. Tsuang MT, Faraone SV. The case for heterogeneity in the etiology of schizophrenia. Schizophr. Res. 1995;17(2):161–175.
    1. Tsuang MT, Nossova N, Yager T, Tsuang M-M, Guo S-C, Shyu KG, Glatt SJ, Liew CC. Assessing the validity of blood-based gene expression profiles for the classification of schizophrenia and bipolar disorder: a preliminary report. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 2005;133B(1):1–5.
    1. Tylee DS, Chandler SD, Nievergelt CM, Liu X, Pazol J, Woelk CH, Lohr JB, Kremen WS, Baker DG, Glatt SJ, Tsuang MT. Blood-based gene-expression biomarkers of post-traumatic stress disorder among deployed marines: A pilot study. Psychoneuroendocrinology. 2015;51:472–494.
    1. Tylee DS, Kawaguchi DM, Glatt SJ. On the outside, looking in: a review and evaluation of the comparability of blood and brain “-omes”. Am. J. Med. Genet. B. Neuropsychiatr. Genet. 2013;162B(7):595–603.
    1. Väremo L, Nielsen J, Nookaew I. Enriching the gene set analysis of genome-wide data by incorporating directionality of gene expression and combining statistical hypotheses and methods. Nucleic Acids Res. 2013;41(8):4378–4391.
    1. Venkatasubramanian G, Debnath M. The TRIPS (Toll-like receptors in immuno-inflammatory pathogenesis) Hypothesis: a novel postulate to understand schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2013;44:301–311.
    1. Watkins NA, Gusnanto A, de Bono B, De S, Miranda-Saavedra D, Hardie DL, Angenent WGJ, Attwood AP, Ellis PD, Erber W, Foad NS, Garner SF, Isacke CM, Jolley J, Koch K, Macaulay IC, Morley SL, Rendon A, Rice KM, Taylor N, Thijssen-Timmer DC, Tijssen MR, van der Schoot CE, Wernisch L, Winzer T, Dudbridge F, Buckley CD, Langford CF, Teichmann S, Göttgens B, Ouwehand WH. A HaemAtlas: characterizing gene expression in differentiated human blood cells. Blood. 2009;113(19):e1–e9.

Source: PubMed

3
S'abonner