Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Neuroimmune transcriptome changes in patient brains of psychiatric and neurological disorders

Abstract

Neuroinflammation has been implicated in multiple brain disorders but the extent and the magnitude of change in immune-related genes (IRGs) across distinct brain disorders has not been directly compared. In this study, 1275 IRGs were curated and their expression changes investigated in 2467 postmortem brains of controls and patients with six major brain disorders, including schizophrenia (SCZ), bipolar disorder (BD), autism spectrum disorder (ASD), major depressive disorder (MDD), Alzheimer’s disease (AD), and Parkinson’s disease (PD). There were 865 IRGs present across all microarray and RNA-seq datasets. More than 60% of the IRGs had significantly altered expression in at least one of the six disorders. The differentially expressed immune-related genes (dIRGs) shared across disorders were mainly related to innate immunity. Moreover, sex, tissue, and putative cell type were systematically evaluated for immune alterations in different neuropsychiatric disorders. Co-expression networks revealed that transcripts of the neuroimmune systems interacted with neuronal-systems, both of which contribute to the pathology of brain disorders. However, only a few genes with expression changes were also identified as containing risk variants in genome-wide association studies. The transcriptome alterations at gene and network levels may clarify the immune-related pathophysiology and help to better define neuropsychiatric and neurological disorders.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Differential expression of immune genes in six disorders.
Fig. 2: Comparison of the effect size of differentially expressed IRGs among neuropsychiatric disease pairs.
Fig. 3: Comparing dIRG-associated function across disorders.
Fig. 4: Shared immune-related co-expression modules.
Fig. 5: Disease-specific co-expression modules.

Similar content being viewed by others

Data availability

All data are available in the main text or the Supplementary Materials.

Code availability

The code of this work can be found at https://github.com/normacyyyyy/IRG-cross-disorder.

References

  1. Parshukova D, Smirnova LP, Ermakov EA, Bokhan NA, Semke AV, Ivanova SA, et al. Autoimmunity and immune system dysregulation in schizophrenia: IgGs from sera of patients hydrolyze myelin basic protein. J Mol Recogn. 2019;32:e2759.

    Article  Google Scholar 

  2. Benedetti F, Aggio V, Pratesi ML, Greco G, Furlan R. Neuroinflammation in Bipolar Depression. Front Psychiatry. 2020;11:71.

    Article  Google Scholar 

  3. Resende R, Fernandes T, Pereira AC, De Pascale J, Marques AP, Oliveira P, et al. Mitochondria, endoplasmic reticulum and innate immune dysfunction in mood disorders: Do Mitochondria-Associated Membranes (MAMs) play a role? Biochim Biophys Acta Mol Basis Dis. 2020;1866:165752.

    Article  CAS  Google Scholar 

  4. Meltzer A, Van de Water J. The Role of the Immune System in Autism Spectrum Disorder. Neuropsychopharmacology. 2017;42:284–98.

    Article  CAS  Google Scholar 

  5. Webers A, Heneka MT, Gleeson PA. The role of innate immune responses and neuroinflammation in amyloid accumulation and progression of Alzheimer’s disease. Immunol Cell Biol. 2020;98:28–41.

    Article  Google Scholar 

  6. De Virgilio A, Greco A, Fabbrini G, Inghilleri M, Rizzo MI, Gallo A, et al. Parkinson’s disease: Autoimmunity and neuroinflammation. Autoimmun Rev. 2016;15:1005–11.

    Article  Google Scholar 

  7. Lu Y, Li K, Hu Y, Wang X. Expression of Immune Related Genes and Possible Regulatory Mechanisms in Alzheimer’s Disease. Front Immunol. 2021;12:768966.

    Article  CAS  Google Scholar 

  8. Zhang X, Shao Z, Xu S, Liu Q, Liu C, Luo Y, et al. Immune Profiling of Parkinson’s Disease Revealed Its Association With a Subset of Infiltrating Cells and Signature Genes. Front Aging Neurosci. 2021;13:605970.

    Article  CAS  Google Scholar 

  9. Horiuchi F, Yoshino Y, Kumon H, Hosokawa R, Nakachi K, Kawabe K, et al. Identification of aberrant innate and adaptive immunity based on changes in global gene expression in the blood of adults with autism spectrum disorder. J Neuroinflammation. 2021;18:102.

    Article  CAS  Google Scholar 

  10. Fillman SG, Sinclair D, Fung SJ, Webster MJ, Shannon, Weickert C. Markers of inflammation and stress distinguish subsets of individuals with schizophrenia and bipolar disorder. Transl Psychiatry. 2014;4:e365.

    Article  CAS  Google Scholar 

  11. Saetre P, Emilsson L, Axelsson E, Kreuger J, Lindholm E, Jazin E. Inflammation-related genes up-regulated in schizophrenia brains. BMC Psychiatry. 2007;7:46.

    Article  Google Scholar 

  12. Cai HQ, Catts VS, Webster MJ, Galletly C, Liu D, O’Donnell M, et al. Increased macrophages and changed brain endothelial cell gene expression in the frontal cortex of people with schizophrenia displaying inflammation. Mol Psychiatry. 2020;25:761–75.

    Article  CAS  Google Scholar 

  13. Zhang Y, Catts VS, Sheedy D, McCrossin T, Kril JJ, Shannon Weickert C. Cortical grey matter volume reduction in people with schizophrenia is associated with neuro-inflammation. Transl Psychiatry. 2016;6:e982.

    Article  CAS  Google Scholar 

  14. Volk DW. Role of microglia disturbances and immune-related marker abnormalities in cortical circuitry dysfunction in schizophrenia. Neurobiol Dis. 2017;99:58–65.

    Article  CAS  Google Scholar 

  15. Gandal MJ, Haney JR, Parikshak NN, Leppa V, Ramaswami G, Hartl C, et al. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science. 2018;359:693–7.

    Article  CAS  Google Scholar 

  16. Gandal MJ, Zhang P, Hadjimichael E, Walker RL, Chen C, Liu S, et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science. 2018;362:6420–15.

  17. Yuan N, Chen Y, Xia Y, Dai J, Liu C. Inflammation-related biomarkers in major psychiatric disorders: a cross-disorder assessment of reproducibility and specificity in 43 meta-analyses. Transl Psychiatry. 2019;9:233–13.

  18. Hart BAT, den Dunnen WF. Commentary on special issue: CNS diseases and the immune system. J Neuroimmune Pharm. 2013;8:757–9.

    Article  Google Scholar 

  19. Gendelman HE. Neural immunity: Friend or foe? J Neurovirol. 2002;8:474–9.

    Article  CAS  Google Scholar 

  20. Mitra S, Chakrabarti N, Dutta SS, Ray S, Bhattacharya P, Sinha P, et al. Gender-specific brain regional variation of neurons, endogenous estrogen, neuroinflammation and glial cells during rotenone-induced mouse model of Parkinson’s disease. Neuroscience. 2015;292:46–70.

    Article  CAS  Google Scholar 

  21. Sparkman NL, Johnson RW. Neuroinflammation associated with aging sensitizes the brain to the effects of infection or stress. Neuroimmunomodulation. 2008;15:323–30.

    Article  CAS  Google Scholar 

  22. Owens T, Wekerle H, Antel J. Genetic models for CNS inflammation. Nat Med. 2001;7:161–6.

    Article  CAS  Google Scholar 

  23. Dantzer R. Neuroimmune Interactions: From the Brain to the Immune System and Vice Versa. Physiol Rev. 2018;98:477–504.

    Article  CAS  Google Scholar 

  24. Nasr IW, Chun Y, Kannan S. Neuroimmune responses in the developing brain following traumatic brain injury. Exp Neurol. 2019;320:112957.

    Article  CAS  Google Scholar 

  25. D’Mello C, Swain MG. Immune-to-Brain Communication Pathways in Inflammation-Associated Sickness and Depression. Curr Top Behav Neurosci. 2017;31:73–94.

    Article  Google Scholar 

  26. Kelley KW, Bluthe RM, Dantzer R, Zhou JH, Shen WH, Johnson RW. et al. Cytokine-induced sickness behavior. Brain Behav Immun. 2003;17 Suppl 1:S112–8.

    Article  CAS  Google Scholar 

  27. Lyman M, Lloyd DG, Ji X, Vizcaychipi MP, Ma D. Neuroinflammation: the role and consequences. Neurosci Res. 2014;79:1–12.

    Article  CAS  Google Scholar 

  28. Ozawa K, Hashimoto K, Kishimoto T, Shimizu E, Ishikura H, Iyo M. Immune activation during pregnancy in mice leads to dopaminergic hyperfunction and cognitive impairment in the offspring: a neurodevelopmental animal model of schizophrenia. Biol Psychiatry. 2006;59:546–54.

    Article  CAS  Google Scholar 

  29. Vuillermot S, Weber L, Feldon J, Meyer U. A longitudinal examination of the neurodevelopmental impact of prenatal immune activation in mice reveals primary defects in dopaminergic development relevant to schizophrenia. J Neurosci. 2010;30:1270–87.

    Article  CAS  Google Scholar 

  30. Lammert CR, Lukens JR. Modeling Autism-Related Disorders in Mice with Maternal Immune Activation (MIA). Methods Mol Biol. 2019;1960:227–36.

    Article  CAS  Google Scholar 

  31. Volk DW, Chitrapu A, Edelson JR, Roman KM, Moroco AE, Lewis DA. Molecular mechanisms and timing of cortical immune activation in schizophrenia. Am J Psychiatry 2015;172:1112–21.

  32. Lee JW, Lee YK, Yuk DY, Choi DY, Ban SB, Oh KW, et al. Neuro-inflammation induced by lipopolysaccharide causes cognitive impairment through enhancement of beta-amyloid generation. J Neuroinflammation. 2008;5:37.

    Article  Google Scholar 

  33. Purves-Tyson TD, Robinson K, Brown AM, Boerrigter D, Cai HQ, Weissleder C, et al. Increased Macrophages and C1qA, C3, C4 Transcripts in the Midbrain of People With Schizophrenia. Front Immunol. 2020;11:2002.

    Article  CAS  Google Scholar 

  34. Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4:147ra111.

    Article  Google Scholar 

  35. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523:337–41.

    Article  CAS  Google Scholar 

  36. Morey JN, Boggero IA, Scott AB, Segerstrom SC. Current Directions in Stress and Human Immune Function. Curr Opin Psychol. 2015;5:13–7.

    Article  Google Scholar 

  37. Kim SY, Buckwalter M, Soreq H, Vezzani A, Kaufer D. Blood-brain barrier dysfunction-induced inflammatory signaling in brain pathology and epileptogenesis. Epilepsia. 2012;53 Suppl 6:37–44.

    Article  CAS  Google Scholar 

  38. Mukherjee S. Immune gene network of neurological diseases: Multiple sclerosis (MS), Alzheimer’s disease (AD), Parkinson’s disease (PD) and Huntington’s disease (HD). Heliyon. 2021;7:e08518.

    Article  CAS  Google Scholar 

  39. Liang WS, Dunckley T, Beach TG, Grover A, Mastroeni D, Walker DG, et al. Gene expression profiles in anatomically and functionally distinct regions of the normal aged human brain. Physiol Genomics. 2007;28:311–22.

    Article  CAS  Google Scholar 

  40. Blalock EM, Buechel HM, Popovic J, Geddes JW, Landfield PW. Microarray analyses of laser-captured hippocampus reveal distinct gray and white matter signatures associated with incipient Alzheimer’s disease. J Chem Neuroanat. 2011;42:118–26.

    Article  CAS  Google Scholar 

  41. Zhang B, Gaiteri C, Bodea LG, Wang Z, McElwee J, Podtelezhnikov AA, et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell. 2013;153:707–20.

    Article  CAS  Google Scholar 

  42. Psych EC, Akbarian S, Liu C, Knowles JA, Vaccarino FM, Farnham PJ, et al. The PsychENCODE project. Nat Neurosci. 2015;18:1707–12.

    Article  Google Scholar 

  43. Voineagu I, Wang X, Johnston P, Lowe JK, Tian Y, Horvath S, et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature. 2011;474:380–4.

    Article  CAS  Google Scholar 

  44. Chow ML, Li HR, Winn ME, April C, Barnes CC, Wynshaw-Boris A, et al. Genome-wide expression assay comparison across frozen and fixed postmortem brain tissue samples. BMC Genom. 2011;12:449.

    Article  CAS  Google Scholar 

  45. Garbett K, Ebert PJ, Mitchell A, Lintas C, Manzi B, Mirnics K, et al. Immune transcriptome alterations in the temporal cortex of subjects with autism. Neurobiol Dis. 2008;30:303–11.

    Article  CAS  Google Scholar 

  46. Hoffman GE, Bendl J, Voloudakis G, Montgomery KS, Sloofman L, Wang YC, et al. CommonMind Consortium provides transcriptomic and epigenomic data for Schizophrenia and Bipolar Disorder. Sci Data. 2019;6:180.

    Article  Google Scholar 

  47. Chen C, Meng Q, Xia Y, Ding C, Wang L, Dai R, et al. The transcription factor POU3F2 regulates a gene coexpression network in brain tissue from patients with psychiatric disorders. Sci Transl Med. 2018;10.

  48. Lanz TA, Reinhart V, Sheehan MJ, Rizzo SJS, Bove SE, James LC, et al. Postmortem transcriptional profiling reveals widespread increase in inflammation in schizophrenia: a comparison of prefrontal cortex, striatum, and hippocampus among matched tetrads of controls with subjects diagnosed with schizophrenia, bipolar or major depressive disorder. Transl Psychiatry. 2019;9:151.

    Article  Google Scholar 

  49. Iwamoto K, Bundo M, Kato T. Altered expression of mitochondria-related genes in postmortem brains of patients with bipolar disorder or schizophrenia, as revealed by large-scale DNA microarray analysis. Hum Mol Genet. 2005;14:241–53.

    Article  CAS  Google Scholar 

  50. Chang LC, Jamain S, Lin CW, Rujescu D, Tseng GC, Sibille E. A conserved BDNF, glutamate- and GABA-enriched gene module related to human depression identified by coexpression meta-analysis and DNA variant genome-wide association studies. PLoS ONE. 2014;9:e90980.

    Article  Google Scholar 

  51. Dumitriu A, Golji J, Labadorf AT, Gao B, Beach TG, Myers RH, et al. Integrative analyses of proteomics and RNA transcriptomics implicate mitochondrial processes, protein folding pathways and GWAS loci in Parkinson disease. BMC Med Genom. 2016;9:5.

    Article  Google Scholar 

  52. Zhang Y, James M, Middleton FA, Davis RL. Transcriptional analysis of multiple brain regions in Parkinson’s disease supports the involvement of specific protein processing, energy metabolism, and signaling pathways, and suggests novel disease mechanisms. Am J Med Genet B Neuropsychiatr Genet. 2005;137B:5–16.

    Article  Google Scholar 

  53. Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, et al. PGC-1alpha, a potential therapeutic target for early intervention in Parkinson’s disease. Sci Transl Med. 2010;2:52ra73.

    Article  Google Scholar 

  54. Riley BE, Gardai SJ, Emig-Agius D, Bessarabova M, Ivliev AE, Schule B, et al. Systems-based analyses of brain regions functionally impacted in Parkinson’s disease reveals underlying causal mechanisms. PLoS ONE. 2014;9:e102909.

    Article  Google Scholar 

  55. Maycox PR, Kelly F, Taylor A, Bates S, Reid J, Logendra R, et al. Analysis of gene expression in two large schizophrenia cohorts identifies multiple changes associated with nerve terminal function. Mol Psychiatry. 2009;14:1083–94.

    Article  CAS  Google Scholar 

  56. Narayan S, Tang B, Head SR, Gilmartin TJ, Sutcliffe JG, Dean B, et al. Molecular profiles of schizophrenia in the CNS at different stages of illness. Brain Res. 2008;1239:235–48.

    Article  CAS  Google Scholar 

  57. Davis AP, Grondin CJ, Johnson RJ, Sciaky D, McMorran R, Wiegers J, et al. The Comparative Toxicogenomics Database: update 2019. Nucleic Acids Res. 2019;47:D948–54.

    Article  CAS  Google Scholar 

  58. Bhattacharya S, Dunn P, Thomas CG, Smith B, Schaefer H, Chen J, et al. ImmPort, toward repurposing of open access immunological assay data for translational and clinical research. Sci Data. 2018;5:180015.

    Article  CAS  Google Scholar 

  59. Diaz-Ramos MC, Engel P, Bastos R. Towards a comprehensive human cell-surface immunome database. Immunol Lett. 2011;134:183–7.

    Article  CAS  Google Scholar 

  60. Breuer K, Foroushani AK, Laird MR, Chen C, Sribnaia A, Lo R, et al. InnateDB: systems biology of innate immunity and beyond—recent updates and continuing curation. Nucleic Acids Res. 2013;41:D1228–33.

    Article  CAS  Google Scholar 

  61. Godec J, Tan Y, Liberzon A, Tamayo P, Bhattacharya S, Butte AJ, et al. Compendium of Immune Signatures Identifies Conserved and Species-Specific Biology in Response to Inflammation. Immunity. 2016;44:194–206.

    Article  CAS  Google Scholar 

  62. The Gene Ontology C. The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Res. 2019;47:D330–8.

    Article  Google Scholar 

  63. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28:27–30.

    Article  CAS  Google Scholar 

  64. Birnbaum R, Jaffe AE, Chen Q, Shin JH, BrainSeq C, Kleinman JE, et al. Investigating the neuroimmunogenic architecture of schizophrenia. Mol Psychiatry. 2018;23:1251–60.

    Article  CAS  Google Scholar 

  65. Yang H, Zhao K, Kang H, Wang M, Wu A. Exploring immune-related genes with prognostic value in microenvironment of breast cancer from TCGA database. Medicine. 2020;99:e19561.

    Article  CAS  Google Scholar 

  66. Zhang M, Wang X, Chen X, Zhang Q, Hong J. Novel Immune-Related Gene Signature for Risk Stratification and Prognosis of Survival in Lower-Grade Glioma. Front Genet. 2020;11:363.

    Article  CAS  Google Scholar 

  67. Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882–3.

    Article  CAS  Google Scholar 

  68. “Picard Toolkit.” Broad Institute, GitHub Repository. Broad Institute. 2018. http://broadinstitute.github.io/picard/

  69. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinform. 2008;9:559.

    Article  Google Scholar 

  70. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–7.

    Article  CAS  Google Scholar 

  71. Zambon AC, Gaj S, Ho I, Hanspers K, Vranizan K, Evelo CT, et al. GO-Elite: a flexible solution for pathway and ontology over-representation. Bioinformatics. 2012;28:2209–10.

    Article  CAS  Google Scholar 

  72. Raudvere U, Kolberg L, Kuzmin I, Arak T, Adler P, Peterson H, et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 2019;47:W191–8.

    Article  CAS  Google Scholar 

  73. 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;38:4218–30.

    Article  CAS  Google Scholar 

  74. Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA, Blumenthal PD, et al. Purification and Characterization of Progenitor and Mature Human Astrocytes Reveals Transcriptional and Functional Differences with Mouse. Neuron. 2016;89:37–53.

    Article  CAS  Google Scholar 

  75. de Leeuw CA, Mooij JM, Heskes T, Posthuma D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput Biol. 2015;11:e1004219.

    Article  Google Scholar 

  76. Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011;27:1739–40.

    Article  CAS  Google Scholar 

  77. Wu X, Shukla R, Alganem K, Zhang X, Eby HM, Devine EA, et al. Transcriptional profile of pyramidal neurons in chronic schizophrenia reveals lamina-specific dysfunction of neuronal immunity. Mol Psychiatry. 2021;26:7699–708.

    Article  CAS  Google Scholar 

  78. Alganem K, Shukla R, Eby H, Abel M, Zhang X, McIntyre WB, et al. Kaleidoscope: A New Bioinformatics Pipeline Web Application for In Silico Hypothesis Exploration of Omics Signatures. bioRxiv 2020. https://doi.org/10.1101/2020.05.01.070805.

  79. Spitsin S, Stevens KE, Douglas SD. Expression of substance P, neurokinin-1 receptor and immune markers in the brains of individuals with HIV-associated neuropathology. J Neurol Sci. 2013;334:18–23.

    Article  CAS  Google Scholar 

  80. Brainstorm C, Anttila V, Bulik-Sullivan B, Finucane HK, Walters RK, Bras J, et al. Analysis of shared heritability in common disorders of the brain. Science. 2018;360:40.

  81. Lotz M, Ebert S, Esselmann H, Iliev AI, Prinz M, Wiazewicz N, et al. Amyloid beta peptide 1-40 enhances the action of Toll-like receptor-2 and -4 agonists but antagonizes Toll-like receptor-9-induced inflammation in primary mouse microglial cell cultures. J Neurochem. 2005;94:289–98.

    Article  CAS  Google Scholar 

  82. Morimoto K, Nakajima K. Role of the Immune System in the Development of the Central Nervous System. Front Neurosci. 2019;13:916.

    Article  Google Scholar 

  83. Nistico R, Salter E, Nicolas C, Feligioni M, Mango D, Bortolotto ZA, et al. Synaptoimmunology - roles in health and disease. Mol Brain. 2017;10:26.

    Article  Google Scholar 

  84. Novellino F, Sacca V, Donato A, Zaffino P, Spadea MF, Vismara M, et al. Innate Immunity: A Common Denominator between Neurodegenerative and Neuropsychiatric Diseases. Int J Mol Sci. 2020; 21:1115–38.

  85. Lenz KM, Nelson LH. Microglia and Beyond: Innate Immune Cells As Regulators of Brain Development and Behavioral Function. Front Immunol. 2018;9:698.

    Article  Google Scholar 

  86. Parab S, Quick RE, Matsuoka RL Endothelial cell-type-specific molecular requirements for angiogenesis drive fenestrated vessel development in the brain. Elife. 2021;10:68.

  87. Goines P, Van de Water J. The immune system’s role in the biology of autism. Curr Opin Neurol. 2010;23:111–7.

    Article  Google Scholar 

  88. Bitanihirwe BK, Peleg-Raibstein D, Mouttet F, Feldon J, Meyer U. Late prenatal immune activation in mice leads to behavioral and neurochemical abnormalities relevant to the negative symptoms of schizophrenia. Neuropsychopharmacology. 2010;35:2462–78.

    Article  CAS  Google Scholar 

  89. Parboosing R, Bao Y, Shen L, Schaefer CA, Brown AS. Gestational influenza and bipolar disorder in adult offspring. JAMA Psychiatry. 2013;70:677–85.

    Article  Google Scholar 

  90. Simanek AM, Meier HC. Association Between Prenatal Exposure to Maternal Infection and Offspring Mood Disorders: A Review of the Literature. Curr Probl Pediatr Adolesc Health Care. 2015;45:325–64.

    Article  Google Scholar 

  91. Caccamo A, De Pinto V, Messina A, Branca C, Oddo S. Genetic reduction of mammalian target of rapamycin ameliorates Alzheimer’s disease-like cognitive and pathological deficits by restoring hippocampal gene expression signature. J Neurosci. 2014;34:7988–98.

    Article  CAS  Google Scholar 

  92. Tanabe S, Yamashita T. The role of immune cells in brain development and neurodevelopmental diseases. Int Immunol. 2018;30:437–44.

    Article  CAS  Google Scholar 

  93. Pais TF, Penha-Goncalves C. Brain Endothelium: The “Innate Immunity Response Hypothesis” in Cerebral Malaria Pathogenesis. Front Immunol. 2018;9:3100.

    Article  CAS  Google Scholar 

  94. Reddy PH, Mani G, Park BS, Jacques J, Murdoch G, Whetsell W Jr, et al. Differential loss of synaptic proteins in Alzheimer’s disease: implications for synaptic dysfunction. J Alzheimers Dis. 2005;7:103–17.

    Article  CAS  Google Scholar 

  95. Russo AJ. Increased Epidermal Growth Factor Receptor (EGFR) Associated with Hepatocyte Growth Factor (HGF) and Symptom Severity in Children with Autism Spectrum Disorders (ASDs). J Cent Nerv Syst Dis. 2014;6:79–83.

    Article  CAS  Google Scholar 

  96. Durieux AM, Fernandes C, Murphy D, Labouesse MA, Giovanoli S, Meyer U, et al. Targeting Glia with N-Acetylcysteine Modulates Brain Glutamate and Behaviors Relevant to Neurodevelopmental Disorders in C57BL/6J Mice. Front Behav Neurosci. 2015;9:343.

    Article  Google Scholar 

  97. Li F, Tian X, Zhou Y, Zhu L, Wang B, Ding M, et al. Dysregulated expression of secretogranin III is involved in neurotoxin-induced dopaminergic neuron apoptosis. J Neurosci Res. 2012;90:2237–46.

    Article  CAS  Google Scholar 

  98. Harris N, Fetter RD, Brasier DJ, Tong A, Davis GW. Molecular Interface of Neuronal Innate Immunity, Synaptic Vesicle Stabilization, and Presynaptic Homeostatic Plasticity. Neuron. 2018;100:1163–79.e4.

    Article  CAS  Google Scholar 

  99. Sasada T, Azuma K, Ohtake J, Fujimoto Y. Immune Responses to Epidermal Growth Factor Receptor (EGFR) and Their Application for Cancer Treatment. Front Pharm. 2016;7:405.

    Article  Google Scholar 

  100. Network, Pathway Analysis Subgroup of Psychiatric Genomics C. Psychiatric genome-wide association study analyses implicate neuronal, immune and histone pathways. Nat Neurosci. 2015;18:199–209.

    Article  Google Scholar 

  101. Tylee DS, Sun J, Hess JL, Tahir MA, Sharma E, Malik R, et al. Genetic correlations among psychiatric and immune-related phenotypes based on genome-wide association data. Am J Med Genet B Neuropsychiatr Genet. 2018;177:641–57.

    Article  Google Scholar 

  102. Pape K, Tamouza R, Leboyer M, Zipp F. Immunoneuropsychiatry—novel perspectives on brain disorders. Nat Rev Neurol. 2019;15:317–28.

    Article  Google Scholar 

  103. Costa-Pinto FA, Palermo-Neto J. Neuroimmune interactions in stress. Neuroimmunomodulation. 2010;17:196–9.

    Article  CAS  Google Scholar 

  104. Haddick PC, Larson JL, Rathore N, Bhangale TR, Phung QT, Srinivasan K, et al. A Common Variant of IL-6R is Associated with Elevated IL-6 Pathway Activity in Alzheimer’s Disease Brains. J Alzheimers Dis. 2017;56:1037–54.

    Article  CAS  Google Scholar 

  105. Jansen IE, Savage JE, Watanabe K, Bryois J, Williams DM, Steinberg S, et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat Genet. 2019;51:404–13.

    Article  CAS  Google Scholar 

  106. Pan PY, Tammimies K, Bolte S. The Association Between Somatic Health, Autism Spectrum Disorder, and Autistic Traits. Behav Genet. 2020;50:233–46.

    Article  Google Scholar 

  107. Xiong J, Chen S, Pang N, Deng X, Yang L, He F, et al. Neurological Diseases With Autism Spectrum Disorder: Role of ASD Risk Genes. Front Neurosci. 2019;13:349.

    Article  Google Scholar 

  108. Khan SA, Khan SA, Narendra AR, Mushtaq G, Zahran SA, Khan S, et al. Alzheimer’s Disease and Autistic Spectrum Disorder: Is there any Association? CNS Neurol Disord Drug Targets. 2016;15:390–402.

    Article  CAS  Google Scholar 

  109. Starkstein S, Gellar S, Parlier M, Payne L, Piven J. High rates of parkinsonism in adults with autism. J Neurodev Disord. 2015;7:29.

    Article  Google Scholar 

  110. Asadi S, Theoharides TC. Corticotropin-releasing hormone and extracellular mitochondria augment IgE-stimulated human mast-cell vascular endothelial growth factor release, which is inhibited by luteolin. J Neuroinflammation. 2012;9:85.

    Article  CAS  Google Scholar 

  111. Mashaghi A, Marmalidou A, Tehrani M, Grace PM, Pothoulakis C, Dana R. Neuropeptide substance P and the immune response. Cell Mol Life Sci. 2016;73:4249–64.

    Article  CAS  Google Scholar 

  112. He ZX, Yin YY, Xi K, Xing ZK, Cao JB, Liu TY, et al. Nucleus Accumbens Tac1-Expressing Neurons Mediate Stress-Induced Anhedonia-like Behavior in Mice. Cell Rep. 2020;33:108343.

    Article  CAS  Google Scholar 

  113. Claes SJ. Corticotropin-releasing hormone (CRH) in psychiatry: from stress to psychopathology. Ann Med. 2004;36:50–61.

    Article  CAS  Google Scholar 

  114. Tsagarakis S, Grossman A. Corticotropin-releasing hormone: interactions with the immune system. Neuroimmunomodulation. 1994;1:329–34.

    Article  CAS  Google Scholar 

  115. O’Kane M, Murphy EP, Kirby B. The role of corticotropin-releasing hormone in immune-mediated cutaneous inflammatory disease. Exp Dermatol. 2006;15:143–53.

    Article  Google Scholar 

  116. Connors EJ, Shaik AN, Migliore MM, Kentner AC. Environmental enrichment mitigates the sex-specific effects of gestational inflammation on social engagement and the hypothalamic pituitary adrenal axis-feedback system. Brain Behav Immun. 2014;42:178–90.

    Article  CAS  Google Scholar 

  117. Sinclair D, Fillman SG, Webster MJ, Weickert CS. Dysregulation of glucocorticoid receptor co-factors FKBP5, BAG1 and PTGES3 in prefrontal cortex in psychotic illness. Sci Rep. 2013;3:3539.

    Article  Google Scholar 

  118. Sinclair D, Webster MJ, Fullerton JM, Weickert CS. Glucocorticoid receptor mRNA and protein isoform alterations in the orbitofrontal cortex in schizophrenia and bipolar disorder. BMC Psychiatry. 2012;12:84.

    Article  CAS  Google Scholar 

  119. Sinclair D, Fullerton JM, Webster MJ, Shannon Weickert C. Glucocorticoid receptor 1B and 1C mRNA transcript alterations in schizophrenia and bipolar disorder, and their possible regulation by GR gene variants. PLoS ONE. 2012;7:e31720.

    Article  CAS  Google Scholar 

  120. Sinclair D, Tsai SY, Woon HG, Weickert CS. Abnormal glucocorticoid receptor mRNA and protein isoform expression in the prefrontal cortex in psychiatric illness. Neuropsychopharmacology. 2011;36:2698–709.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Published microarray datasets analyzed in this study are available on Gene Expression Omnibus (accession No. GSE28521, GSE28475, GSE35978, GSE53987, GSE17612, GSE12649, GSE21138, GSE54567, GSE54568, GSE54571, GSE54572, GSE29555, GSE5281, GSE28146, GSE20168, GSE20295, GSE54282, GSE68719 and GSE11223), ArrayExpress (accession no. E-MTAB-184), or directly from the study authors. RNA-seq data (available on Synapse with accession numbers syn4590909 and syn4587609, with access governed by NIMH Repository and Genomics Resource) were generated as part of the PsychENCODE Consortium, supported by grants U01MH103339, U01MH103365, U01MH103392, U01MH103340, U01MH103346, R01MH105472, R01MH094714, R01MH105898, R21MH102791, R21MH105881, R21MH103877, and P50MH106934 awarded to Schahram Akbarian (Icahn School of Medicine at Mount Sinai), Gregory Crawford (Duke), Stella Dracheva (Icahn School of Medicine at Mount Sinai), Peggy Farnham (USC), Mark Gerstein (Yale), Daniel Geschwind (UCLA), Thomas M. Hyde (LIBD), Andrew Jaffe (LIBD), James A. Knowles (USC), Chunyu Liu (SUNY), Dalila Pinto (Icahn School of Medicine at Mount Sinai), Nenad Sestan (Yale), Pamela Sklar (Icahn School of Medicine at Mount Sinai), Matthew State (UCSF), Patrick Sullivan (UNC), Flora Vaccarino (Yale), Sherman Weissman (Yale), Kevin White (UChicago), and Peter Zandi (JHU). RNA-seq data from the CommonMind Consortium used in this study (Synapse accession no. syn2759792) was supported by funding from Takeda Pharmaceuticals Company, F. Hoffman-La Roche and NIH grants R01MH085542, R01MH093725, P50MH066392, P50MH080405, R01MH097276, RO1-MH-075916, P50M096891, P50MH084053S1, R37MH057881, R37MH057881S1, HHSN271201300031C, AG02219, AG05138, and MH06692. RNA-seq data from the ROSMAP were provided by the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago. Data collection was supported through funding by NIA grants P30AG10161 (ROS), R01AG15819 (ROSMAP; genomics and RNA-seq), R01AG17917 (MAP), R01AG30146, R01AG36042 (5hC methylation, ATACseq), RC2AG036547 (H3K9Ac), R01AG36836 (RNA-seq), R01AG48015 (monocyte RNA-seq) RF1AG57473 (single nucleus RNA-seq), U01AG32984 (genomic and whole exome sequencing), U01AG46152 (ROSMAP AMP-AD, targeted proteomics), U01AG46161(TMT proteomics), U01AG61356 (whole genome sequencing, targeted proteomics, ROSMAP AMP-AD), the Illinois Department of Public Health (ROSMAP), and the Translational Genomics Research Institute (genomic). Brain tissue for the study was obtained from the following brain bank collections: the Mount Sinai NIH Brain and Tissue Repository, the University of Pennsylvania Alzheimer’s Disease Core Center, the University of Pittsburgh NeuroBioBank and Brain and Tissue Repositories, the Harvard Brain Bank as part of the Autism Tissue Project (ATP), the Stanley Medical Research Institute, and the NIMH Human Brain Collection Core. We thank Mingrui Yu, and Yan Xia for reviewing the paper.

Funding

National Natural Science Foundation of China Nos. 82022024. National Natural Science Foundation of China Nos. 31970572. National Natural Science Foundation of China Nos. 31871276. the National Key R&D Project of China Grants No. 2016YFC1306000. the National Key R&D Project of China Grants No. 2017YFC0908701. Innovation-driven Project of Central South University Grant Nos. 2020CX003. NIH grants U01MH122591. NIH grants U01MH116489. NIH grants R01MH110920.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: YC, CL. Methodology: YC, JD, LT. Investigation: YC, JD, QL, QH, ML, JZ. Visualization: YC. Funding acquisition: CL, CC. Project administration: CL. Supervision: CL, CC. Writing—original draft: YC, JD, CL, Writing—review ānd editing: YC, CL, JD, T.M., RK, CC,CW.

Corresponding authors

Correspondence to Chao Chen or Chunyu Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Y., Dai, J., Tang, L. et al. Neuroimmune transcriptome changes in patient brains of psychiatric and neurological disorders. Mol Psychiatry 28, 710–721 (2023). https://doi.org/10.1038/s41380-022-01854-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-022-01854-7

This article is cited by

Search

Quick links