Neuro-transcriptomic signatures for mood disorder morbidity and suicide mortality

https://doi.org/10.1016/j.jpsychires.2020.05.013Get rights and content

Abstract

Suicidal behaviors are strongly linked with mood disorders, but the specific neurobiological and functional gene-expression correlates for this linkage remain elusive. We performed neuroimaging-guided RNA-sequencing in two studies to test the hypothesis that imaging-localized gray matter volume (GMV) loss in mood disorders, harbors gene-expression changes associated with disease morbidity and related suicide mortality in an independent postmortem cohort. To do so, first, we conducted study 1 using an anatomical likelihood estimation (ALE) MRI meta-analysis including a total of 47 voxel-based morphometry (VBM) publications (i.e. 26 control versus (vs) major depressive disorder (MDD) studies, and 21 control vs bipolar disorder (BD) studies) in 2387 (living) participants. Study 1 meta-analysis identified a selective anterior insula cortex (AIC) GMV loss in mood disorders. We then used this results to guide study 2 postmortem tissue dissection and RNA-Sequencing of 100 independent donor brain samples with a life-time history of MDD (N = 30), BD (N = 37) and control (N = 33). In study 2, exploratory factor-analysis identified a higher-order factor representing number of Axis-1 diagnoses (e.g. substance use disorders/psychosis/anxiety, etc.), referred to here as morbidity and suicide-completion referred to as mortality. Comparisons of case-vs-control, and factor-analysis defined higher-order-factor contrast variables revealed that the imaging-identified AIC GMV loss sub-region harbors differential gene-expression changes in high morbidity-&-mortality versus low morbidity-&-mortality cohorts in immune, inflammasome, and neurodevelopmental pathways. Weighted gene co-expression network analysis further identified co-activated gene modules for psychiatric morbidity and mortality outcomes. These results provide evidence that AIC anatomical signature for mood disorders are possible correlates for gene-expression abnormalities in mood morbidity and suicide mortality.

Introduction

Major depressive disorder and bipolar disorder ⎯ here together referred to as mood disorders, are the third leading cause of the global disease burden (Collins et al., 2011; Murray et al., 2012). Mood disorders account for the majority of completed suicides (Waern et al., 2002; Marangell et al., 2006) and they were linked to ~48,000 suicides in the United States in 2018 alone (American Foundation for Suicide Prevention, 2019). However, the convergent neurobiological basis for mood symptoms/syndromes and suicide is unknown, limiting advances in developing novel interventions.

Neuroimaging studies have identified reduction in gray matter volume (GMV) in the anterior insular cortex (AIC) and anterior cingulate cortex (ACC) in association with diagnosis of psychiatric disorders in general (Goodkind et al., 2015), and the regional GMV volume reductions in these AIC and ACC network have been especially implicated in mood disorder diagnoses in particular (Goodkind et al., 2015; Wise et al., 2017). Neurobiological integrity of the right AIC is shown to (a) predict mood diagnostic severity (Hatton et al., 2012), (b) modulate subjective responses to distress, pain, and psychosocial adversity (Wager et al., 2013; Eisenberger, 2015), (c) regulate affective interoception (Craig, 2009; Khalsa et al., 2018), (d) associate with stress-related inflammatory markers (Slavich et al., 2010), and (e) predict psycho- and pharmaco-therapeutic efficacy in mood disorders (McGrath et al., 2013). AIC-ACC functional connectivity during affective processing differentiated mood disorder suicide-attempters from non-attempters (Pan et al., 2013). Furthermore, abnormalities in AIC volume and synaptic abnormalities are linked to suicidal-behavior in mood disorder (Wagner et al., 2012; Mathews et al., 2013). AIC response to stress is shown to impact hypothalamic-pituitary-adrenal (HPA) axis-driven inflammatory responses (Khalsa et al., 2013), which may serve to exacerbate mood disorder associated psychiatric morbidity and suicidal-behavior (Oquendo et al., 2014; Wohleb et al., 2016). Although a preponderance of evidence supports abnormal AIC integrity in psychosocial distress (Shneidman, 1998; Mee et al., 2011; Wager et al., 2013) and mood/comorbid psychiatric symptoms, an underlying functional genetic contribution in terms of functional gene-expression changes for these abnormalities remains largely unknown.

The lack of a well-defined relationship between aberrant brain structure and function with underlying molecular changes within these abnormal brain regions is an impediment to understanding pathophysiology. Moreover, evidence for shared genetic mechanisms underlying psychiatric diagnoses (Brainstorm consortium, Anttila et al., 2018) is not well-integrated with brain imaging correlates of psychiatric disease-morbidity and specific behaviors, in this case, suicide. In the present study, MRI meta-analysis was used to test the hypothesis that reduced AIC volume will be the most prominent neuroanatomical signature for mood disorder diagnoses. We confirmed this hypothesis with our meta-analytic findings in study 1 and then used this anatomical hallmark to guide dissection of postmortem brain tissue for analyses of molecular/gene-expression signatures that could pave the way for precision profiling of gene functions underlying mood symptoms across diagnoses in clinically-relevant brain sub-regions in study 2. This approach enabled us to further test the hypothesis that the voxel-based morphometry (VBM) imaging meta-analysis defined in study 1 will harbor postmortem gene-expression signatures for psychiatric disease morbidity and related suicide-mortality in postmortem mood disorder brains, thereby providing a neurobiological framework for characterizing convergent neural-and-gene expression signatures for behavioral brain diseases.

Section snippets

Participants & Samples

The imaging meta-analysis provided a consolidation of the current mood disorder VBM work by quantitatively integrating published results of volumetric comparisons of interest between controls vs mood disorder participants, or correlations of volumetric measures with mood disorder symptom-specific measures. In total, 47 previous VBM publications consisting of volumetric comparisons or experimental contrasts assessing GMV reductions in healthy controls vs major depression from 26 independent

VBM meta-analysis

Convergence across the findings reported in previous VBM studies was assessed using ALE, which in brief consists of first modelling the spatial uncertainty associated with each reported location for significant between-group differences (Eickhoff et al., 2009; Turkeltaub et al., 2012). The ALE method therefore computes the convergence across experiments by the union of the ensuing probabilistic model relative to a null-distribution reflecting a random spatial association between the findings of

Identification of a mood disorder neuroanatomical signature in living brains

The study 1 VBM meta-analysis (N = 2387) revealed reduced GMV in the right AIC in mood disorders (p < 0.0001 corrected) (Fig. 1A) consistent across both major depressive disorder and bipolar disorder, since major depressive disorder and bipolar disorder groups did not differ significantly (Fig. 1A). The localized reduced AIC neuroanatomical-signature for mood disorders was manually segmented in ITKSNAP (http://www.itksnap.org/pmwiki/pmwiki.php) (Fig. 1B) and the segmented volume guided

Discussion

Using a neuroimaging meta-analysis to refine a structurally reduced AIC brain region of interest in major depression and bipolar disorder, we identified a postmortem across-diagnostic mood disorder linked psychiatric morbidity-&-mortality associated gene-expression signature within this neuroimaging meta-analysis identified reduced AIC gray matter signature. Given this AIC sub-region's documented role in regulating affective and physical pain/distress, general bodily homeostatic and

CRediT authorship contribution statement

Mbemba Jabbi conceived and designed the studies, and acquired postmortem material from the NIMH HBCC. Mbemba Jabbi, Dhivya Arasappan, Simon B. Eickhoff, and Hans A. Hofmann performed the experiments and analyzed the data and results. Mbemba Jabbi drafted the manuscript and Dhivya Arasappan, Simon B. Eickhoff, Stephen M. Strakowski, Charles B. Nemeroff, and Hans A. Hofmann contributed critically to the original drafting, interpretation of the findings, and writing of the paper.

Declaration of competing interest

Mbemba Jabbi, none.

Dhivya Arasappan, none.

Simon Eickhoff none.

Hans Hofmann, none.

Stephen Strakowski: chairs DSMBs for Sunovion as a Consultant, and has research grants with Janssen pharmaceuticals and Alkermes.

Charles Nemeroff: 1) Consulting (last three years) for Xhale, Takeda, Taisho Pharmaceutical Inc., Bracket (Clintara), Fortress Biotech, Sunovion Pharmaceuticals Inc., Sumitomo Dainippon Pharma, Janssen Research & Development LLC, Magstim, Inc., Navitor Pharmaceuticals, Inc., TC MSO, Inc.,

Acknowledgements

The NIMH Human Brain Collection Core provided RNA-samples for all 100 postmortem data and we thank the NIMH and Drs. Barbara Lipska, Stefano Marenco, Pavan Auluck and HBCC colleagues for providing the studied samples. We thank Wade Weber of Dell Medical School Psychiatry Department, UT Austin for assistance in preparing the manuscript, Dr. Mark Bond of Dell Medical School Psychiatry Department, UT Austin for statistical reviews, Nicole Elmer of the UT Austin Biomedical research support for help

References (63)

  • S.M. Smith et al.

    Threshold-free cluster enhancement: addressing problems of smoothing, threshold dependence and localisation in cluster inference

    Neuroimage

    (2009)
  • W.J. Su

    NLRP3 gene knockout blocks NF-kappaB and MAPK signaling pathway in CUMS-induced depression mouse model

    Behav. Brain Res.

    (2017)
  • M.D. Turner et al.

    Cytokines and chemokines: at the crossroads of cell signaling and inflammatory disease

    Biochem Biophys Acta

    (2014)
  • G. Wagner et al.

    Prefrontal cortical thickness in depressed patients with high-risk for suicidal behavior

    J. Psychiatr. Res.

    (2012)
  • S. Anders et al.

    Differential expression analysis for sequence count data

    Genome Biol.

    (2010)
  • S. Andrews

    FastQC: a Quality Control Tool for High Throughput Sequence Data

    (2010)
  • V. Anttila

    Analysis of shared heritability in common disorders of the brain

    Science

    (2018)
  • F.C. Bennett et al.

    The immune system and psychiatric disease: a basic science perspective

    Clin. Exp. Immunol.

    (2019)
  • N.L. Bray et al.

    Near-optimal probabilistic RNA-seq quantification

    Nat. Biotechnol.

    (2016)
  • O. Butovsky et al.

    Microglial signatures and their role in health and disease

    Nat. Rev. Neurosci.

    (2018)
  • A. Caviedes et al.

    BDNF/NF-κB signaling in the neurobiology of depression

    Curr. Pharmaceut. Des.

    (2017)
  • E.Y. Chen et al.

    Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool

    BMC Bioinf.

    (2013)
  • P.Y. Collins

    Grand challenges in global mental health

    Nature

    (2011)
  • A.B. Costello et al.

    Best practices in exploratory factor analysis: four recommendations for getting the most from your analysis

    Practical Assess. Res. Eval.

    (2005)
  • S.B. Eickhoff et al.

    Coordinate-based activation likelihood estimation meta-analysis of neuroimaging data: a random-effects approach based on empirical estimates of spatial uncertainty

    Hum. Brain Mapp.

    (2009)
  • AD. Craig

    How do you feel—now 2009? The anterior insula and human awareness

    Nat. Rev. Neurosci.

    (2009)
  • N.I. Eisenberger

    Social pain and the brain: controversies, questions, and where to go from here

    Annu. Rev. Psychol.

    (2015)
  • M.J. Gandal

    Common Mind Consortium; PsychENCODE Consortium; iPSYCH-BROAD Working Group, Horvath S, Geschwind DH. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap

    Science

    (2018)
  • M. Goodkind

    Identification of a common neurobiological substrate for mental illness

    JAMA Psychiatry

    (2015)
  • S.N. Hatton et al.

    Correlating anterior insula gray matter volume changes in young people with clinical and neurocognitive outcomes: an MRI study

    BMC Psychiatr.

    (2012)
  • M. Jabbi et al.

    The Williams syndrome chromosome 7q11.23 hemideletion confers hypersocial, anxious personality coupled with altered insula structure and function

    Proc of the Nat Acad of Sci USA

    (2012)
  • Cited by (17)

    • The Role of Inflammation in the Pathophysiology of Depression and Suicidal Behavior: Implications for Treatment

      2023, Medical Clinics of North America
      Citation Excerpt :

      As such, extensive efforts have been put forward to comprehensively characterize the presence of inflammatory markers in various types of specimens from depressed subjects. Among different neurobiological hypotheses, genome-wide association studies have been carried out to identify candidate genes that are linked to depression vulnerability.30,31 Notably, various genetic networks of inflammation have been identified, with variants in cytokines and inflammatory mediators as the most reproducible findings.32

    • Integrating biobehavioral information to predict mood disorder suicide risk

      2022, Brain, Behavior, and Immunity - Health
      Citation Excerpt :

      In contrast to these chemokine findings, another study found increased chemokine gene expression in the dorsal anterior cingulate of depressed suicides (Torres-Platas et al., 2014). However, CCL4 was downregulated in mood disorders when comparing high vs. low psychiatric morbidity and suicide mortality (Jabbi et al., 2020b). Jabbi et al. (2020b) replicated the Pantazatos finding (Pantazatos et al., 2017) of CCL4 downregulion in MDD and suicide.

    • Hippocampal neuropathology in suicide: Gaps in our knowledge and opportunities for a breakthrough

      2022, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      Evidently, mapping the cellular and molecular complexity of the events that underlie the conversion from suicide ideation to suicide completion will be key to identifying novel and putatively druggable regulators and pathways. Only a handful of studies assaying the human ‘suicide’ brain in an unbiased, genome-wide manner, and in relation to MDs, or rather independently of them, have been published to date (Glavan et al., 2021; Jabbi et al., 2020; Pantazatos et al., 2017; Punzi et al., 2019; Zhou et al., 2018). Strikingly, a common theme emerging from all these reports is the involvement of immune and inflammatory responses in the transcriptomic signatures related to suicide.

    View all citing articles on Scopus
    View full text