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:

A common epigenetic mechanism across different cellular origins underlies systemic immune dysregulation in an idiopathic autism mouse model

Abstract

Immune dysregulation plays a key role in the pathogenesis of autism. Changes occurring at the systemic level, from brain inflammation to disturbed innate/adaptive immune in the periphery, are frequently observed in patients with autism; however, the intrinsic mechanisms behind them remain elusive. We hypothesize a common etiology may lie in progenitors of different types underlying widespread immune dysregulation. By single-cell RNA sequencing (sc-RNA seq), we trace the developmental origins of immune dysregulation in a mouse model of idiopathic autism. It is found that both in aorta-gonad-mesonephros (AGM) and yolk sac (YS) progenitors, the dysregulation of HDAC1-mediated epigenetic machinery alters definitive hematopoiesis during embryogenesis and downregulates the expression of the AP-1 complex for microglia development. Subsequently, these changes result in the dysregulation of the immune system, leading to gut dysbiosis and hyperactive microglia in the brain. We further confirm that dysregulated immune profiles are associated with specific microbiota composition, which may serve as a biomarker to identify autism of immune-dysregulated subtypes. Our findings elucidate a shared mechanism for the origin of immune dysregulation from the brain to the gut in autism and provide new insight to dissecting the heterogeneity of autism, as well as the therapeutic potential of targeting immune-dysregulated autism subtypes.

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: Single-cell RNA-seq analysis of AGM progenitor cells identified alteration of HDAC1-mediated transcriptional repression in delaying the process of endothelial-to-hematopoietic transition (EHT).
Fig. 2: Single-cell RNA-seq analysis of YS progenitors also identified HDAC1 as the pathogenic factor.
Fig. 3: A shared mechanism underlying the pathologic progenitors can be modulated at specific developmental windows to restore immune dysregulation.
Fig. 4: Pathologic progenitor causes immune dysregulation and dysbiosis in the gut microbiome.

Similar content being viewed by others

Data availability

All the sequencing data that support the findings of this study have been deposited in the Gene Expression Omnibus with the accession code GSE197618.

References

  1. Bourgeron T. From the genetic architecture to synaptic plasticity in autism spectrum disorder. Nat Rev Neurosci. 2015;16:551–63.

    Article  CAS  PubMed  Google Scholar 

  2. Takumi T, Tamada K, Hatanaka F, Nakai N, Bolton PF. Behavioral neuroscience of autism. Neurosci Biobehav Rev. 2020;110:60–76.

    Article  PubMed  Google Scholar 

  3. de la Torre-Ubieta L, Won H, Stein JL, Geschwind DH. Advancing the understanding of autism disease mechanisms through genetics. Nat Med. 2016;22:345–61.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nat Rev Genet. 2008;9:341–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wisniowiecka-Kowalnik B, Nowakowska BA. Genetics and epigenetics of autism spectrum disorder-current evidence in the field. J Appl Genet. 2019;60:37–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Meltzer A, Van de Water J. The role of the immune system in autism spectrum disorder. Neuropsychopharmacology. 2017;42:284–98.

    Article  CAS  PubMed  Google Scholar 

  7. Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2005;57:67–81.

    Article  CAS  PubMed  Google Scholar 

  8. Morgan JT, Chana G, Pardo CA, Achim C, Semendeferi K, Buckwalter J, et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol psychiatry. 2010;68:368–76.

    Article  PubMed  Google Scholar 

  9. Li X, Chauhan A, Sheikh AM, Patil S, Chauhan V, Li XM, et al. Elevated immune response in the brain of autistic patients. J Neuroimmunol. 2009;207:111–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah I, Van de Water J. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun. 2011;25:40–5.

    Article  CAS  PubMed  Google Scholar 

  11. Vuong HE, Hsiao EY. Emerging roles for the gut microbiome in autism spectrum disorder. Biol Psychiatry. 2017;81:411–23.

    Article  PubMed  Google Scholar 

  12. Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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  PubMed  PubMed Central  Google Scholar 

  14. Gupta S, Ellis SE, Ashar FN, Moes A, Bader JS, Zhan J, et al. Transcriptome analysis reveals dysregulation of innate immune response genes and neuronal activity-dependent genes in autism. Nat Commun. 2014;5:5748.

    Article  CAS  PubMed  Google Scholar 

  15. Geschwind DH. Genetics of autism spectrum disorders. Trends Cogn Sci. 2011;15:409–16.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Hsiao EY, McBride SW, Chow J, Mazmanian SK, Patterson PH. Modeling an autism risk factor in mice leads to permanent immune dysregulation. Proc Natl Acad Sci USA. 2012;109:12776–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Heo Y, Zhang Y, Gao D, Miller VM, Lawrence DA. Aberrant immune responses in a mouse with behavioral disorders. PloS One. 2011;6:e20912.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Golubeva AV, Joyce SA, Moloney G, Burokas A, Sherwin E, Arboleya S, et al. Microbiota-related changes in bile acid & tryptophan metabolism are associated with gastrointestinal dysfunction in a mouse model of autism. EBioMedicine. 2017;24:166–78.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Li Z, Lan Y, He W, Chen D, Wang J, Zhou F, et al. Mouse embryonic head as a site for hematopoietic stem cell development. cell stem cell. 2012;11:663–75.

    Article  CAS  PubMed  Google Scholar 

  20. Morgan K, Kharas M, Dzierzak E, Gilliland DG. Isolation of early hematopoietic stem cells from murine yolk sac and AGM. J Vis Exp. 2008:789. https://doi.org/10.3791/789.e789.

  21. Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36:411–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019;20:296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014;32:381–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic acids Res. 2009;37:W305–311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Onore CE, Careaga M, Babineau BA, Schwartzer JJ, Berman RF, Ashwood P. Inflammatory macrophage phenotype in BTBR T+tf/J mice. Front Neurosci. 2013;7:158.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Schwartzer JJ, Onore CE, Rose D, Ashwood P. C57BL/6J bone marrow transplant increases sociability in BTBR T(+) Itpr3(tf)/J mice. Brain Behav Immun. 2017;59:55–61.

    Article  CAS  PubMed  Google Scholar 

  28. Gao X, Xu C, Asada N, Frenette PS. The hematopoietic stem cell niche: from embryo to adult. Development. 2018;145:dev139691.

  29. Rybtsov S, Ivanovs A, Zhao S, Medvinsky A. Concealed expansion of immature precursors underpins acute burst of adult HSC activity in foetal liver. Development. 2016;143:1284–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhou F, Li X, Wang W, Zhu P, Zhou J, He W, et al. Tracing haematopoietic stem cell formation at single-cell resolution. Nature. 2016;533:487–92.

    Article  CAS  PubMed  Google Scholar 

  31. Lancrin C, Sroczynska P, Stephenson C, Allen T, Kouskoff V, Lacaud G. The haemangioblast generates haematopoietic cells through a haemogenic endothelium stage. Nature. 2009;457:892–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Oatley M, Bolukbasi OV, Svensson V, Shvartsman M, Ganter K, Zirngibl K, et al. Single-cell transcriptomics identifies CD44 as a marker and regulator of endothelial to haematopoietic transition. Nat Commun. 2020;11:586.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Baron CS, Kester L, Klaus A, Boisset JC, Thambyrajah R, Yvernogeau L, et al. Single-cell transcriptomics reveal the dynamic of haematopoietic stem cell production in the aorta. Nat Commun. 2018;9:2517.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Chen MJ, Yokomizo T, Zeigler BM, Dzierzak E, Speck NA. Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter. Nature. 2009;457:887–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Swiers G, Baumann C, O’Rourke J, Giannoulatou E, Taylor S, Joshi A, et al. Early dynamic fate changes in haemogenic endothelium characterized at the single-cell level. Nat Commun. 2013;4:2924.

    Article  PubMed  Google Scholar 

  36. Huang HT, Kathrein KL, Barton A, Gitlin Z, Huang YH, Ward TP, et al. A network of epigenetic regulators guides developmental haematopoiesis in vivo. Nat cell Biol. 2013;15:1516–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang J, Kalkum M, Yamamura S, Chait BT, Roeder RG. E protein silencing by the leukemogenic AML1-ETO fusion protein. Science. 2004;305:1286–9.

    Article  CAS  PubMed  Google Scholar 

  38. Nagarajan S, Rao SV, Sutton J, Cheeseman D, Dunn S, Papachristou EK, et al. ARID1A influences HDAC1/BRD4 activity, intrinsic proliferative capacity and breast cancer treatment response. Nat Genet. 2020;52:187–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Thambyrajah R, Fadlullah MZH, Proffitt M, Patel R, Cowley SM, Kouskoff V, et al. HDAC1 and HDAC2 modulate TGF-beta signaling during endothelial-to-hematopoietic transition. Stem Cell Rep. 2018;10:1369–83.

    Article  CAS  Google Scholar 

  40. Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 2015;518:547–51.

    Article  PubMed  Google Scholar 

  41. Sasagawa Y, Danno H, Takada H, Ebisawa M, Tanaka K, Hayashi T, et al. Quartz-Seq2: a high-throughput single-cell RNA-sequencing method that effectively uses limited sequence reads. Genome Biol. 2018;19:29.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Gosselin D, Link VM, Romanoski CE, Fonseca GJ, Eichenfield DZ, Spann NJ, et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell. 2014;159:1327–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gosselin D, Skola D, Coufal NG, Holtman IR, Schlachetzki JCM, Sajti E et al. An environment-dependent transcriptional network specifies human microglia identity. Science. 2017;356:eaal3222.

  44. Aibar S, Gonzalez-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulselmans G, et al. SCENIC: single-cell regulatory network inference and clustering. Nat Methods. 2017;14:1083–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rabouw HH, Visser LJ, Passchier TC, Langereis MA, Liu F, Giansanti P, et al. Inhibition of the integrated stress response by viral proteins that block p-eIF2-eIF2B association. Nat Microbiol. 2020;5:1361–73.

    Article  CAS  PubMed  Google Scholar 

  46. Eckschlager T, Plch J, Stiborova M, Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 2017;18:1414.

  47. Kratsman N, Getselter D, Elliott E. Sodium butyrate attenuates social behavior deficits and modifies the transcription of inhibitory/excitatory genes in the frontal cortex of an autism model. Neuropharmacology. 2016;102:136–45.

    Article  CAS  PubMed  Google Scholar 

  48. Qin L, Ma K, Wang ZJ, Hu Z, Matas E, Wei J, et al. Social deficits in Shank3-deficient mouse models of autism are rescued by histone deacetylase (HDAC) inhibition. Nat Neurosci. 2018;21:564–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Davie JR. Inhibition of histone deacetylase activity by butyrate. J Nutr. 2003;133:2485S–93S.

    Article  CAS  PubMed  Google Scholar 

  50. VanderMolen KM, McCulloch W, Pearce CJ, Oberlies NH. Romidepsin (Istodax, NSC 630176, FR901228, FK228, depsipeptide): a natural product recently approved for cutaneous T-cell lymphoma. J Antibiot. 2011;64:525–31.

    Article  CAS  Google Scholar 

  51. Baronio D, Bauer-Negrini G, Castro K, Della-Flora Nunes G, Riesgo R, Mendes-da-Cruz DA, et al. Reduced CD4 T lymphocytes in lymph nodes of the mouse model of autism induced by valproic acid. Neuroimmunomodulation. 2018;25:280–4.

    Article  CAS  PubMed  Google Scholar 

  52. Nicolini C, Fahnestock M. The valproic acid-induced rodent model of autism. Exp Neurol. 2018;299(Pt A):217–27.

    Article  CAS  PubMed  Google Scholar 

  53. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–41.

    Article  CAS  PubMed  Google Scholar 

  54. Coretti L, Cristiano C, Florio E, Scala G, Lama A, Keller S, et al. Sex-related alterations of gut microbiota composition in the BTBR mouse model of autism spectrum disorder. Sci Rep. 2017;7:45356.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Needham BD, Tang W, Wu WL. Searching for the gut microbial contributing factors to social behavior in rodent models of autism spectrum disorder. Dev Neurobiol. 2018;78:474–99.

    Article  PubMed  Google Scholar 

  56. Cedar H, Bergman Y. Epigenetics of haematopoietic cell development. Nat Rev Immunol. 2011;11:478–88.

    Article  CAS  PubMed  Google Scholar 

  57. Gerstner T, Bell N, Longin E, Konig SA. Oral rapid loading of valproic acid-an alternative to the usual saturation scheme? Seizure. 2006;15:630–2.

    Article  PubMed  Google Scholar 

  58. Lee J, Sayed N, Hunter A, Au KF, Wong WH, Mocarski ES, et al. Activation of innate immunity is required for efficient nuclear reprogramming. Cell. 2012;151:547–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhao M, Tan Y, Peng Q, Huang C, Guo Y, Liang G, et al. IL-6/STAT3 pathway induced deficiency of RFX1 contributes to Th17-dependent autoimmune diseases via epigenetic regulation. Nat Commun. 2018;9:583.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah IN, Van de Water J. Altered T cell responses in children with autism. Brain, Behav, Immun. 2011;25:840–9.

    Article  CAS  PubMed  Google Scholar 

  61. Coretti L, Paparo L, Riccio MP, Amato F, Cuomo M, Natale A, et al. Gut microbiota features in young children with autism spectrum disorders. Front Microbiol. 2018;9:3146.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Ivanovs A, Rybtsov S, Ng ES, Stanley EG, Elefanty AG, Medvinsky A. Human haematopoietic stem cell development: from the embryo to the dish. Development. 2017;144:2323–37.

    Article  CAS  PubMed  Google Scholar 

  63. Bezawada N, Phang TH, Hold GL, Hansen R. Autism spectrum disorder and the gut microbiota in children: a systematic review. Ann Nutr Metab. 2020;76:16–29.

    Article  CAS  PubMed  Google Scholar 

  64. Sgritta M, Dooling SW, Buffington SA, Momin EN, Francis MB, Britton RA, et al. Mechanisms underlying microbial-mediated changes in social behavior in mouse models of autism spectrum disorder. Neuron. 2019;101:246–259 e246.

    Article  CAS  PubMed  Google Scholar 

  65. Silverman JL, Yang M, Lord C, Crawley JN. Behavioural phenotyping assays for mouse models of autism. Nat Rev Neurosci. 2010;11:490–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Buffington SA, Di Prisco GV, Auchtung TA, Ajami NJ, Petrosino JF, Costa-Mattioli M. Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring. Cell. 2016;165:1762–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci USA. 2010;107:12204–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all technical staff of Takumi laboratory for their assistance, with special thanks to K. Yanaka for the assistance with FACS analysis, Y. Nomura, and Y. Kusakari for the assistance with 10x genomics library preparation. We also thank S. Chikuma (Keio University) for his instruction and discussion on BMT experiments and M. Izumi (RIKEN Nishina Center for Accelerator-Based Science) for the guidance on usage and maintenance of X-ray generator. We are grateful to the Support Unit for Animal Resources Development and the Support Unit for Bio-Material Analysis, RIKEN CBS Research Resources Division, with special thanks to T. Arai for mouse embryo manipulation and K. Ohtawa for the technical support with FACS analysis and single-cell sorting for quartez-seq2. We thank D. Polygalov (Laboratory for Circuit and Behavioral Physiology, RIKEN CBS) for his assistance on SCENIC analysis. For Quartz-seq2, we thank A. Matsushima and M. Ishii (RIKEN BDR) for assistance with the infrastructure for the data analysis; H. Danno (Knowledge Palette, Inc.) for the development of data analysis software for single-cell transcriptomes.

Funding

C-WL was supported by fellowships from the Japan Society for the Promotion of Science (JSPS) and the Tokyo Biochemical Research Foundation (TBRF). This work was in part supported by KAKENHI (15F15105, 16H06316, 16H06463, 19K16529, 21H00202, 21H04813, 21K19351) from JSPS and the Ministry of Education, Culture, Sports, Science, and Technology, Japan Agency for Medical Research and Development (AMED) under Grant Number JP21wm0425011, Intramural Research Grant (30-9) for Neurological and Psychiatric Disorders of NCNP, the Takeda Science Foundation, Smoking Research Foundation, SENSHIN Medical Research Foundation, TBRF, Hyogo Science and Technology Association, Kawano Masanori Memorial Public Interest Incorporated Foundation for Promotion of Pediatrics, Taiju Life Social Welfare Foundation, Naito Foundation, JST CREST (JPMJCR16G3), RIKEN Epigenetics Program, RIKEN Epigenome Control Program, and the Projects for Technological Development, and Research Center Network for Realization of Regenerative Medicine from AMED.

Author information

Authors and Affiliations

Authors

Contributions

C-WL and TT conceived the study. C-WL performed or was involved in all experiments. MK, KA, KTakeshita, and KH performed 16S rRNA metagenomic sequencing. DES performed 10x genomics single-cell RNA-seq and the FACS analysis. JN helped singe-cell RNA-seq analysis. YS, KTanaka, and IN performed Quarz-seq2. C-WL wrote the paper, with editing provided by KA, KTamada, H-WC, TJM, and TT.

Corresponding author

Correspondence to Toru Takumi.

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, CW., Septyaningtrias, D.E., Chao, HW. et al. A common epigenetic mechanism across different cellular origins underlies systemic immune dysregulation in an idiopathic autism mouse model. Mol Psychiatry 27, 3343–3354 (2022). https://doi.org/10.1038/s41380-022-01566-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-022-01566-y

This article is cited by

Search

Quick links