Replicable and Coupled Changes in Innate and Adaptive Immune Gene Expression in Two Case-Control Studies of Blood Microarrays in Major Depressive Disorder

Gwenaël G R Leday, Petra E Vértes, Sylvia Richardson, Jonathan R Greene, Tim Regan, Shahid Khan, Robbie Henderson, Tom C Freeman, Carmine M Pariante, Neil A Harrison, MRC Immunopsychiatry Consortium, V Hugh Perry, Wayne C Drevets, Gayle M Wittenberg, Edward T Bullmore, Edward Bullmore, Petra Vertes, Rudolf Cardinal, Sylvia Richardson, Gwenael Leday, Tom Freeman, Tim Regan, David Hume, Zhaozong Wu, Carmine Pariante, Annamaria Cattaneo, Patricia Zunszain, Alessandra Borsini, Robert Stewart, David Chandran, Livia Carvalho, Joshua Bell, Luis Souza-Teodoro, Hugh Perry, Neil Harrison, Wayne Drevets, Gayle Wittenberg, Declan Jones, Edward Bullmore, Shahid Khan, Annie Stylianou, Robbie Henderson, Gwenaël G R Leday, Petra E Vértes, Sylvia Richardson, Jonathan R Greene, Tim Regan, Shahid Khan, Robbie Henderson, Tom C Freeman, Carmine M Pariante, Neil A Harrison, MRC Immunopsychiatry Consortium, V Hugh Perry, Wayne C Drevets, Gayle M Wittenberg, Edward T Bullmore, Edward Bullmore, Petra Vertes, Rudolf Cardinal, Sylvia Richardson, Gwenael Leday, Tom Freeman, Tim Regan, David Hume, Zhaozong Wu, Carmine Pariante, Annamaria Cattaneo, Patricia Zunszain, Alessandra Borsini, Robert Stewart, David Chandran, Livia Carvalho, Joshua Bell, Luis Souza-Teodoro, Hugh Perry, Neil Harrison, Wayne Drevets, Gayle Wittenberg, Declan Jones, Edward Bullmore, Shahid Khan, Annie Stylianou, Robbie Henderson

Abstract

Background: Peripheral inflammation is often associated with major depressive disorder (MDD), and immunological biomarkers of depression remain a focus of investigation.

Methods: We used microarray data on whole blood from two independent case-control studies of MDD: the GlaxoSmithKline-High-Throughput Disease-specific target Identification Program [GSK-HiTDiP] study (113 patients and 57 healthy control subjects) and the Janssen-Brain Resource Company study (94 patients and 100 control subjects). Genome-wide differential gene expression analysis (18,863 probes) resulted in a p value for each gene in each study. A Bayesian method identified the largest p-value threshold (q = .025) associated with twice the number of genes differentially expressed in both studies compared with the number of coincidental case-control differences expected by chance.

Results: A total of 165 genes were differentially expressed in both studies with concordant direction of fold change. The 90 genes overexpressed (or UP genes) in MDD were significantly enriched for immune response to infection, were concentrated in a module of the gene coexpression network associated with innate immunity, and included clusters of genes with correlated expression in monocytes, monocyte-derived dendritic cells, and neutrophils. In contrast, the 75 genes underexpressed (or DOWN genes) in MDD were associated with the adaptive immune response and included clusters of genes with correlated expression in T cells, natural killer cells, and erythroblasts. Consistently, the MDD patients with overexpression of UP genes also had underexpression of DOWN genes (correlation > .70 in both studies).

Conclusions: MDD was replicably associated with proinflammatory activation of the peripheral innate immune system, coupled with relative inactivation of the adaptive immune system, indicating the potential of transcriptional biomarkers for immunological stratification of patients with depression.

Keywords: Affymetrix; Bayesian; Biomarker; Inflammation; Systems; Transcriptome.

Copyright © 2017 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
Protein–protein interaction network for proteins coded by the set of 165 replicably concordant genes differentially expressed in both case-control studies of major depressive disorder. The network is represented by an undirected graph where links correspond to known protein–protein interactions and weights are proportional to the Search Tool for the Retrieval of Interacting Genes/Proteins confidence score . Only high-confidence (>0.7) links are retained, and disconnected genes are not shown. Red (blue) nodes correspond to genes over- (under-) expressed in major depressive disorder in both the GlaxoSmithKline–High-Throughput Disease-specific target Identification Program and Janssen–Brain Resource Company datasets. Smaller inner circles highlight proteins that are coded by genes also differentially expressed and with the same sign of fold change in a third large independent case-control study of major depressive disorder (Netherlands Study of Depression and Anxiety [NESDA]) thresholded to control false discovery rate (FDR) at 20% (white circles), 10% (gray circles), and 5% (black circles).
Figure 2
Figure 2
Major depressive disorder (MDD)-related genes in the context of the normative whole-genome transcriptome. Overexpressed genes (or UP genes) in patients with MDD are concentrated in a module of the normative gene coexpression network specialized for innate immune response, whereas underexpressed genes (or DOWN genes) are concentrated in a module partially specialized for adaptive immune response. (A) The modules of the normative transcriptome are highlighted in different colors. (B) The MDD-related genes are colored according to their normative module affiliation, and representative genes are text-labeled. (C) The MDD-related genes are colored green for overexpressed (or UP) genes and are colored red for underexpressed (or DOWN) genes. The text labels highlight the functions of the corresponding modules of the normative transcriptome. rRNA, ribosomal RNA; SRP, signal recognition particle.
Figure 3
Figure 3
Cell class–specific expression patterns for the set of 165 replicably concordant genes. We estimated the correlation between each possible pair of the 165 major depressive disorder (MDD)-related genes and identified clusters of genes with similar expression patterns in an independent microarray dataset on specific cell types. The set of 165 replicably concordant genes formed six clusters, with each cluster comprising a subset of genes that had strong mutual coexpression across a range of eight distinct cell classes: erythroblasts, monocytes, monocyte-derived dendritic cells (MDDCs), neutrophils, B cells, CD4+ T cells, CD8+ T cells, and natural killer (NK) cells. (A) The six clusters of genes with strongly correlated expression profiles. Clusters 1 to 4 (left) comprised genes that were overexpressed in MDD (or UP genes), and clusters 5 and 6 (right) comprised genes that were underexpressed in MDD (or DOWN genes). (B) Histograms of clustered gene expression across cell types for each of the four clusters of UP genes overexpressed in MDD (from top to bottom: clusters 1–4). The x-axis color legend codes for different cell types. (C) Histograms of clustered gene expression across cell types for each of the two clusters of DOWN genes underexpressed in MDD (from top to bottom: clusters 5 and 6). The x-axis color legend codes for different cell types.
Figure 4
Figure 4
Opposing and coordinated expression of innate and adaptive immune transcripts in patients with major depressive disorder (MDD). (A, B) Scatter plots for the mean expression across the 75 underexpressed (DOWN) genes plotted against the mean expression of 90 overexpressed (UP) genes in the Janssen–Brain Resource Company (Janssen–BRC) (A) and GlaxoSmithKline–High-Throughput Disease-specific target Identification Program (GSK–HiTDiP) (B) datasets. Each point corresponds to a patient. Blue indicates case, and gray indicates control (CTL). Regression lines are shown in blue and gray, respectively. In (A), an individual patient’s data point (red outline) is projected onto the regression line (red circle). The distance from the origin to the point on the regression line is the bioscalar value for that patient. Inset illustrates the projection (black) of all individual patient data points (blue) onto the sample regression line. (C, D) Left panels: Box plot of MDD-165 bioscalar values in controls and cases. Green line indicates the threshold identifying the top third (tertile) of the MDD-165 bioscalar distribution as a subgroup of inflamed patients with MDD (designated immune-MDD). Right panels: Receiver operating characteristic curves were calculated for MDD vs. CTL and immune-MDD vs. CTL classifications. (E) Correlation of each depressed patient’s MDD-165 bioscalar with body mass index (BMI). (F, G) Box plots of MDD-165 bioscalar for subgroups defined by a comorbid diagnosis of anxiety disorder. AUC, area under the curve.

References

    1. Wium-Andersen M.K., Ørsted D.D., Nielsen S.F., Nordestgaard B.G. Elevated C-reactive protein levels, psychological distress, and depression in 73,131 individuals. JAMA Psychiatry. 2013;70:176–184.
    1. Valkanova V., Ebmeier K.P., Allan C.L. CRP, IL-6 and depression: A systematic review and meta-analysis of longitudinal studies. J Affect Disord. 2013;150:736–744.
    1. Haapakoski R., Mathieu J., Ebmeier K.P., Alenius H., Kivimaki M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behav Immun. 2015;49:206–215.
    1. Matcham F., Rayner L., Steer S., Hotopf M. The prevalence of depression in rheumatoid arthritis: A systematic review and meta-analysis. Rheumatology. 2013;52:2136–2148.
    1. Khandaker G.M., Pearson R.M., Zammit S., Lewis G., Jones P.B. Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: A population-based longitudinal study. JAMA Psychiatry. 2014;71:1121–1128.
    1. Bull S.J., Huezo-Diaz P., Binder E.B., Cubells J.F., Ranjith G., Maddock C. Functional polymorphisms in the interleukin-6 and serotonin transporter genes, and depression and fatigue induced by interferon-α and ribavirin treatment. Mol Psychiatry. 2009;14:1095–1104.
    1. Hepgul N., Cattaneo A., Agarwal K., Baraldi S., Borsini A., Bufalino C. Transcriptomics in interferon-α-treated patients identifies inflammation-, neuroplasticity- and oxidative stress-related signatures as predictors and correlates of depression. Neuropsychopharmacology. 2016;41:2502–2511.
    1. Dantzer R., O’Connor J., Freund G., Johnson R., Kelley K. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat Rev Neurosci. 2008;9:46–56.
    1. Banks W.A., Kastin A.J., Gutierrez E.G. Penetration of interleukin-6 across the murine blood-brain barrier. Neurosci Lett. 1994;179:53–56.
    1. Perry V.H., Cunningham C., Holmes C. Systemic infections and inflammation affect chronic neurodegeneration. Nat Rev Immunol. 2007;7:161–167.
    1. Miller A.H., Raison C.L. Are anti-inflammatory therapies viable treatments for psychiatric disorders? Where the rubber meets the road. JAMA Psychiatry. 2015;72:527–528.
    1. Raison C., Capuron L., Miller A. Cytokines sing the blues: Inflammation and the pathogenesis of depression. Trends Immunol. 2006;27:24–31.
    1. Lin E., Tsai S.J. Genome-wide microarray analysis of gene expression profiling in major depression and antidepressant therapy. Prog Neuropsychopharmacol Biol Psychiatry. 2016;64:334–340.
    1. Jansen R., Penninx B.W.J.H., Madar V., Xia K., Milaneschi Y., Hottenga J.J. Gene expression in major depressive disorder. Mol Psychiatry. 2016;21:444.
    1. Garbett K.A., Vereczkei A., Kálmán S., Brown J.A., Taylor W.D., Faludi G. Coordinated messenger RNA/microRNA changes in fibroblasts of patients with major depression. Biol Psychiatry. 2015;77:256–265.
    1. Mostafavi S., Battle A., Zhu X., Potash J.B., Weissman M.M., Shi J. Type I interferon signaling genes in recurrent major depression: Increased expression detected by whole-blood RNA sequencing. Mol Psychiatry. 2014;19:1267–1274.
    1. Glahn D.C., Curran J.E., Winkler A.M., Carless M.A., Kent J.W., Charlesworth J.C. High dimensional endophenotype ranking in the search for major depression risk genes. Biol Psychiatry. 2012;71:6–14.
    1. Shelton R.C., Claiborne J., Sidoryk-Wegrzynowicz M., Reddy R., Aschner M., Lewis D.A. Altered expression of genes involved in inflammation and apoptosis in frontal cortex in major depression. Mol Psychiatry. 2011;16:751–762.
    1. Blume J., Douglas S.D., Evans D.L. Immune suppression and immune activation in depression. Brain Behav Immun. 2011;25:221–229.
    1. Miller A., Maletic V., Raison C. Inflammation and its discontents: The role of cytokines in the pathophysiology of major depression. Biol Psychiatry. 2009;65:732–741.
    1. Grosse L., Hoogenboezem T., Ambrée O., Bellingrath S., Jörgens S., de Wit H.J. Deficiencies of the T and natural killer cell system in major depressive disorder: T regulatory cell defects are associated with inflammatory monocyte activation. Brain Behav Immun. 2016;54:38–44.
    1. Snijders G., Schiweck C., Mesman E., Grosse L., de Wit H.J., Nolen W.A. A dynamic course of T cell defects in individuals at risk for mood disorders. Brain Behav Immun. 2016;58:11–17.
    1. Grosse L., Carvalho L.A., Wijkhuijs A.M., Bellingrath S., Ruland T., Ambree O. Clinical characteristics of inflammation-associated depression: Monocyte gene expression is age-related in major depressive disorder. Brain Behav Immun. 2015;44:48–56.
    1. Domenici E., Willé D.R., Tozzi F., Prokopenko I., Miller S., McKeown A. Plasma protein biomarkers for depression and schizophrenia by multi analyte profiling of case-control collections. PLoS One. 2010;5:e9166.
    1. Muglia P., Tozzi F., Galwey N.W., Francks C., Upmanyu R., Kong X.Q. Genome-wide association study of recurrent major depressive disorder in two European case-control cohorts. Mol Psychiatry. 2010;15:589–601.
    1. Tilahun A., Lin D., Shkedy Z., Geys H., Alonso A., Peeters P. Genomic biomarkers for depression: Feature-specific and joint biomarkers. Stat Biopharm Res. 2010;2:419–434.
    1. Zoon H., Veth C., Arns M., Drinkenburg W., Talloen W., Peeters P. EEG alpha power as an intermediate measure between brain-derived neurotrophic factor Val66Met and depression severity in patients with major depressive disorder. J Clin Neurophys. 2013;30:261–267.
    1. Liu Y., Yieh L., Yang T., Drinkenburg W., Peeters P., Steckler T. Metabolomic biosignature differentiates melancholic depressive patients from healthy controls. BMC Genomics. 2016;17:669.
    1. Sheehan D.V., Lecrubier Y., Sheehan K.H., Amorim P., Janavs J., Weiller E. The Mini-International Neuropsychiatric Interview (MINI): The development and validation of a structured diagnostic psychiatric interview for DSM-IV and ICD-10. J Clin Psychiatry. 1998;59:22–33.
    1. Ritchie M.E., Phipson B., Wu D., Hu Y., Law C.W., Shi W. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.
    1. Smyth G.K. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004;3:3.
    1. Blangiardo M., Richardson S. Statistical tools for synthesizing lists of differentially expressed features in related experiments. Genome Biol. 2007;8:R54.
    1. Blangiardo M., Cassese A., Richardson S. sdef: An R package to synthesize lists of significant features in related experiments. BMC Bioinformatics. 2010;11:270.
    1. Zhang B., Horvath S. A general framework for weighted gene co-expression network analysis. Stat Appl Genet Mol Biol. 2005;4:17.
    1. Alexa A, Rahnenfuhrer J (2010): topGO: Enrichment analysis for gene ontology. R package, version 2.0.
    1. Ashburner M., Ball C.A., Blake J.A., Botstein D., Butler H., Cherry J.M. Gene Ontology: Tool for the unification of biology. Nat Genet. 2000;25:25–29.
    1. Szklarczyk D., Franceschini A., Wyder S., Forslund K., Heller D., Huerta-Cepas J. STRING v10: Protein–protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43:D447–D452.
    1. Franceschini A., Szklarczyk D., Frankild S., Kuhn M., Simonovic M., Roth A. STRING v9: 1. Protein–protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 2013;41:D808–D815.
    1. Mabbott N.A., Baillie J.K., Brown H., Freeman T.C., Hume D.A. An expression atlas of human primary cells: Inference of gene function from coexpression networks. BMC Genomics. 2013;14:632.
    1. Freeman T.C., Goldovsky L., Brosch M., Van Dongen S., Mazière P., Grocock R.J. Construction, visualisation, and clustering of transcription networks from microarray expression data. PLoS Comput Biol. 2007;3:e206.
    1. Theocharidis A., van Dongen S., Enright A., Freeman T. Network visualization and analysis of gene expression data using BioLayout Express3D. Nat Protoc. 2009;4:1535–1550.
    1. Langfelder P., Horvath S. WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559.
    1. Inamdar A., Merlo-Pich E., Gee M., Makumi C., Mistry P., Robertson J. Evaluation of antidepressant properties of the p38 MAP kinase inhibitor losmapimod (GW856553) in major depressive disorder: Results from two randomised, placebo-controlled, double-blind, multicentre studies using a Bayesian approach. J Psychopharmacol. 2014;28:570–581.
    1. Zorrilla E.P., Luborsky L., McKay J.R., Rosenthal R., Houldin A., Tax A. The relationship of depression and stressors to immunological assays: A meta-analytic review. Brain Behav Immun. 2001;15:199–226.
    1. Carvalho L.A., Bergink V., Sumaski L., Wijkhuijs J., Hoogendijk W.J., Birkenhager T.K. Inflammatory activation is associated with a reduced glucocorticoid receptor alpha/beta expression ratio in monocytes of inpatients with melancholic major depressive disorder. Transl Psychiatry. 2014;4:e344.
    1. Weigelt K., Carvalho L.A., Drexhage R.C., Wijkhuijs A., de Wit H., van Beveren N.J. TREM-1 and DAP12 expression in monocytes of patients with severe psychiatric disorders: EGR3, ATF3 and PU.1 as important transcription factors. Brain Behav Immun. 2011;25:1162–1169.
    1. Maes M., Lambrechts J., Bosmans E., Jacobs J., Suy E., Vandervorst C. Evidence for a systemic immune activation during depression: Results of leukocyte enumeration by flow cytometry in conjunction with monoclonal antibody staining. Psychol Med. 1992;22:45–53.
    1. Wong M.L., 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;13:800–812.
    1. Major Depressive Disorder Working Group of the Psychiatric GWAS Consortium. Ripke S., Wray N.R., Lewis C.M., Hamilton S.P., Weissman M.M. A mega-analysis of genome-wide association studies for major depressive disorder. Mol Psychiatry. 2013;18:497–511.
    1. Danese A., Pariante C.M., Caspi A., Taylor A., Poulton R. Childhood maltreatment predicts adult inflammation in a life-course study. Proc Natl Acad Sci U S A. 2007;104:1319–1324.
    1. Rohleder N. Stimulation of systemic low-grade inflammation by psychosocial stress. Psychosom Med. 2014;76:181–189.

Source: PubMed

3
S'abonner