Preprint / Version 1

How Does Maternal Immune Activity Affect Fetal Survival and Brain Development?: The critical roles of IL-17A and microglia

##article.authors##

  • Asumi Kubo Laboratory of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba
  • Sara Kamiya Laboratory of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba
  • Koki Higuchi Laboratory of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba
  • Kenyu Nakamura Laboratory of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba
  • Kyoko Kishi Laboratory of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba
  • Tetsuya Sasaki Laboratory of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba https://orcid.org/0000-0002-7723-4417 https://researchmap.jp/tsasak

DOI:

https://doi.org/10.51094/jxiv.861

Keywords:

Autism Spectrum Disorder, Interleukin-17A, Maternal Immune Activation, Microglia, Miscarriage

Abstract

Maternal immune activation (MIA) during pregnancy has been associated with increased risk of fetal loss and neurodevelopmental disorders in offspring. This review summarizes recent findings on the effects of MIA on fetal survival and microglial phenotype. Studies using polyinosinic-polycytidylic acid (poly(I:C))-induced MIA mouse models have revealed a crucial role for interleukin-17A (IL-17A) in mediating these effects. Overexpression of RORγt, a key transcription factor for IL-17A production, enhances poly(I:C)-induced fetal loss, possibly due to increased placental vulnerability. Intraventricular administration of IL-17A in fetal brains activates microglia and alters their localization, particularly in periventricular regions and the medial cortex. These activated microglia may contribute to abnormal synaptic pruning and excessive phagocytosis of neural progenitor cells, potentially leading to long-term neurodevelopmental abnormalities. The insights gained from MIA research have important clinical implications, including the potential for early identification of high-risk pregnancies and the development of novel preventive and therapeutic strategies. Future research should focus on elucidating the roles of other cytokines, determining critical periods of MIA susceptibility, and translating findings to human populations, while carefully considering ethical implications and the need for appropriate risk communication.

Conflicts of Interest Disclosure

There are no conflicts of interest to disclose.

Downloads *Displays the aggregated results up to the previous day.

Download data is not yet available.

Author Biography

Sara Kamiya, Laboratory of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba

Laboratory of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba

References

Patterson PH. Maternal infection and immune involvement in autism. Trends Mol Med. 2011 Jul;17(7):389–94.

Estes ML, McAllister AK. Maternal immune activation: Implications for neuropsychiatric disorders. Science. 2016 Aug 19;353(6301):772–7.

Asumi Kubo A, Sara Kamiya, Tetsuya Sasaki. Effects of Maternal Immune Activation and IL-17A on Abortion. Bio Clinica. 2024 Aug 10;39(1):38–40.

Christensen DL, Braun KVN, Baio J, Bilder D, Charles J, Constantino JN, et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years - Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2012. MMWR Surveill Summ. 2018 Nov 16;65(13):1–23.

Maenner MJ, Shaw KA, Bakian AV, Bilder DA, Durkin MS, Esler A, et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years - Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2018. MMWR Surveill Summ. 2021 Dec 3;70(11):1–16.

佐々木哲也. 胎仔期プログラミングと自閉スペクトラム症. DOHaD研究. 2022 Dec;11:15–6.

Mor G, Cardenas I. The immune system in pregnancy: a unique complexity. Am J Reprod Immunol. 2010 Jun;63(6):425–33.

Reisinger S, Khan D, Kong E, Berger A, Pollak A, Pollak DD. The poly(I:C)-induced maternal immune activation model in preclinical neuropsychiatric drug discovery. Pharmacol Ther. 2015 May;149:213–26.

Smith SEP, Li J, Garbett K, Mirnics K, Patterson PH. Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci. 2007 Oct 3;27(40):10695–702.

Goldenberg RL, Thompson C. The infectious origins of stillbirth. Am J Obstet Gynecol. 2003 Sep;189(3):861–73.

Fell DB, Savitz DA, Kramer MS, Gessner BD, Katz MA, Knight M, et al. Maternal influenza and birth outcomes: systematic review of comparative studies. BJOG. 2017 Jan;124(1):48–59.

Di Mascio D, Khalil A, Saccone G, Rizzo G, Buca D, Liberati M, et al. Outcome of coronavirus spectrum infections (SARS, MERS, COVID-19) during pregnancy: a systematic review and meta-analysis. Am J Obstet Gynecol MFM. 2020 May;2(2):100107.

Eaton WW, Mortensen PB, Thomsen PH, Frydenberg M. Obstetric complications and risk for severe psychopathology in childhood. J Autism Dev Disord. 2001 Jun;31(3):279–85.

Larsson HJ, Eaton WW, Madsen KM, Vestergaard M, Olesen AV, Agerbo E, et al. Risk factors for autism: perinatal factors, parental psychiatric history, and socioeconomic status. Am J Epidemiol. 2005 May 15;161(10):916–25; discussion 926-8.

Walker CK, Krakowiak P, Baker A, Hansen RL, Ozonoff S, Hertz-Picciotto I. Preeclampsia, placental insufficiency, and autism spectrum disorder or developmental delay. JAMA Pediatr. 2015 Feb;169(2):154–62.

Gaffen SL. Structure and signalling in the IL-17 receptor family. Nat Rev Immunol. 2009 Jul 3;9(8):556–67.

Dong C. TH17 cells in development: an updated view of their molecular identity and genetic programming. Nat Rev Immunol. 2008 May;8(5):337–48.

Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, et al. The Orphan Nuclear Receptor RORγt Directs the Differentiation Program of Proinflammatory IL-17+ T Helper Cells. Cell. 2006 Sep 22;126(6):1121–33.

Huber M, Heink S, Grothe H, Guralnik A, Reinhard K, Elflein K, et al. A Th17-like developmental process leads to CD8(+) Tc17 cells with reduced cytotoxic activity. Eur J Immunol. 2009 Jul;39(7):1716–25.

Wang W-J, Hao C-F, Yi-Lin, Yin G-J, Bao S-H, Qiu L-H, et al. Increased prevalence of T helper 17 (Th17) cells in peripheral blood and decidua in unexplained recurrent spontaneous abortion patients. J Reprod Immunol. 2010 Mar;84(2):164–70.

Fu B, Tian Z, Wei H. TH17 cells in human recurrent pregnancy loss and pre-eclampsia. Cell Mol Immunol. 2014 Nov;11(6):564–70.

Liu Y-S, Wu L, Tong X-H, Wu L-M, He G-P, Zhou G-X, et al. Study on the relationship between Th17 cells and unexplained recurrent spontaneous abortion. Am J Reprod Immunol. 2011 May;65(5):503–11.

Tome S, Sasaki T, Takahashi S, Takei Y. Elevated maternal retinoic acid-related orphan receptor-γt enhances the effect of polyinosinic-polycytidylic acid in inducing fetal loss. Exp Anim. 2019 Nov 6;68(4):491–7.

Sasaki T, Nagata R, Takahashi S, Takei Y. Effects of RORγt overexpression on the murine central nervous system. Neuropsychopharmacol Rep. 2021 Mar;41(1):102–10.

Yoh K, Morito N, Ojima M, Shibuya K, Yamashita Y, Morishima Y, et al. Overexpression of RORγt under control of the CD2 promoter induces polyclonal plasmacytosis and autoantibody production in transgenic mice. Eur J Immunol. 2012 Aug;42(8):1999–2009.

Al-Ayadhi LY, Mostafa GA. Elevated serum levels of interleukin-17A in children with autism. J Neuroinflammation. 2012 Jul 2;9:158.

Li H, Dang Y, Yan Y. Serum interleukin-17 A and homocysteine levels in children with autism. BMC Neurosci. 2024 Mar 12;25(1):17.

Kim S, Kim H, Yim YS, Ha S, Atarashi K, Tan TG, et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature. 2017 Sep 28;549(7673):528–32.

Choi GB, Yim YS, Wong H, Kim S, Kim H, Kim SV, et al. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science. 2016 Feb 26;351(6276):933–9.

Yousef H, Czupalla CJ, Lee D, Chen MB, Burke AN, Zera KA, et al. Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1. Nat Med. 2019 Jun;25(6):988–1000.

Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med. 2017 Sep 8;23(9):1018–27.

Ostrem BEL, Domínguez-Iturza N, Stogsdill JA, Faits T, Kim K, Levin JZ, et al. Fetal brain response to maternal inflammation requires microglia. Development [Internet]. 2024 May 15;151(10). Available from: http://dx.doi.org/10.1242/dev.202252

Ozaki K, Kato D, Ikegami A, Hashimoto A, Sugio S, Guo Z, et al. Maternal immune activation induces sustained changes in fetal microglia motility. Sci Rep. 2020 Dec 7;10(1):21378.

Das Sarma J, Ciric B, Marek R, Sadhukhan S, Caruso ML, Shafagh J, et al. Functional interleukin-17 receptor A is expressed in central nervous system glia and upregulated in experimental autoimmune encephalomyelitis. J Neuroinflammation. 2009 Apr 28;6:14.

Kang Z, Wang C, Zepp J, Wu L, Sun K, Zhao J, et al. Act1 mediates IL-17–induced EAE pathogenesis selectively in NG2+ glial cells. Nat Neurosci. 2013 Sep 1;16(10):1401–8.

Sasaki T, Tome S, Takei Y. Intraventricular IL-17A administration activates microglia and alters their localization in the mouse embryo cerebral cortex. Mol Brain. 2020 Jun 16;13(1):93.

哲也佐々木. 免疫系分子が大脳皮質形成に果たす役割と精神疾患におけるその異常. DOHaD研究. 2023;11(2):126–34.

Cunningham CL, Martínez-Cerdeño V, Noctor SC. Microglia regulate the number of neural precursor cells in the developing cerebral cortex. J Neurosci. 2013 Mar 6;33(10):4216–33.

佐田文宏. DOHaDとの出会いと10年間の歩み. DOHaD研究. 2023;11(1):17–9.

佐田文宏/福岡秀輿, editor. DOHaD 先制医療への展開. 金原出版株式会社; 2023年5月10日.

Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the immune system. Front Behav Neurosci. 2009 Aug 24;3:14.

Mattei D, Ivanov A, Ferrai C, Jordan P, Guneykaya D, Buonfiglioli A, et al. Maternal immune activation results in complex microglial transcriptome signature in the adult offspring that is reversed by minocycline treatment. Transl Psychiatry. 2017 May 9;7(5):e1120.

Dalmau I, Finsen B, Zimmer J, González B, Castellano B. Development of microglia in the postnatal rat hippocampus. Hippocampus. 1998;8(5):458–74.

Fox CJ, Russell KI, Wang YT, Christie BR. Contribution of NR2A and NR2B NMDA subunits to bidirectional synaptic plasticity in the hippocampus in vivo. Hippocampus. 2006;16(11):907–15.

Argaw AT, Asp L, Zhang J, Navrazhina K, Pham T, Mariani JN, et al. Astrocyte-derived VEGF-A drives blood-brain barrier disruption in CNS inflammatory disease. J Clin Invest. 2012 Jul;122(7):2454–68.

Chen X, Oppenheim JJ. Th17 cells and Tregs: unlikely allies. J Leukoc Biol. 2014 May;95(5):723–31.

Okubo Y, Mera T, Wang L, Faustman DL. Homogeneous expansion of human T-regulatory cells via tumor necrosis factor receptor 2. Sci Rep. 2013 Nov 6;3:3153.

Nedoszytko B, Lange M, Sokołowska-Wojdyło M, Renke J, Trzonkowski P, Sobjanek M, et al. The role of regulatory T cells and genes involved in their differentiation in pathogenesis of selected inflammatory and neoplastic skin diseases. Part I: Treg properties and functions. Postepy Dermatol Alergol. 2017 Aug;34(4):285–94.

Prajeeth CK, Kronisch J, Khorooshi R, Knier B, Toft-Hansen H, Gudi V, et al. Effectors of Th1 and Th17 cells act on astrocytes and augment their neuroinflammatory properties. J Neuroinflammation. 2017 Oct 16;14(1):204.

Prajeeth CK, Löhr K, Floess S, Zimmermann J, Ulrich R, Gudi V, et al. Effector molecules released by Th1 but not Th17 cells drive an M1 response in microglia. Brain Behav Immun. 2014 Mar;37:248–59.

Engelhardt B, Ransohoff RM. Capture, crawl, cross: the T cell code to breach the blood–brain barriers. Trends Immunol. 2012 Dec 1;33(12):579–89.

Cipollini V, Anrather J, Orzi F, Iadecola C. Th17 and Cognitive Impairment: Possible Mechanisms of Action. Front Neuroanat. 2019 Nov 19;13:95.

Rostami A, Ciric B. Role of Th17 cells in the pathogenesis of CNS inflammatory demyelination. J Neurol Sci. 2013 Oct 15;333(1–2):76–87.

Posted


Submitted: 2024-08-27 20:36:10 UTC

Published: 2024-08-30 00:40:14 UTC
Section
Biology, Life Sciences & Basic Medicine