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.

  • Review Article
  • Published:

Maternal immune activation and abnormal brain development across CNS disorders

Key Points

  • The developing brain is particularly sensitive to environmental signals that influence genetically determined developmental processes

  • Infection-induced maternal immune activation (mIA) during pregnancy can have a profound impact on developing neural circuits

  • Strong epidemiological associations exist between exposure to various infections during pregnancy and greater risk of schizophrenia, autism or epilepsy in the progeny

  • Emerging evidence suggests similar links for disorders like cerebral palsy and ageing-associated neurodegenerative diseases, positioning mIA as a factor in the brain's responsiveness to cumulative lifetime exposure to environmental insults

  • Microglia constitute the primary immune mediators of neural functions, and their mIA-induced priming is thought to underlie some of the persistent immunological and/or neurological changes associated with mIA

  • Targeting of immune-related pathways might represent a promising therapeutic strategy for neurodevelopmental, psychiatric and neurological disorders

Abstract

Epidemiological studies have shown a clear association between maternal infection and schizophrenia or autism in the progeny. Animal models have revealed maternal immune activation (mIA) to be a profound risk factor for neurochemical and behavioural abnormalities in the offspring. Microglial priming has been proposed as a major consequence of mIA, and represents a critical link in a causal chain that leads to the wide spectrum of neuronal dysfunctions and behavioural phenotypes observed in the juvenile, adult or aged offspring. Such diversity of phenotypic outcomes in the mIA model are mirrored by recent clinical evidence suggesting that infectious exposure during pregnancy is also associated with epilepsy and, to a lesser extent, cerebral palsy in children. Preclinical research also suggests that mIA might precipitate the development of Alzheimer and Parkinson diseases. Here, we summarize and critically review the emerging evidence that mIA is a shared environmental risk factor across CNS disorders that varies as a function of interactions between genetic and additional environmental factors. We also review ongoing clinical trials targeting immune pathways affected by mIA that may play a part in disease manifestation. In addition, future directions and outstanding questions are discussed, including potential symptomatic, disease-modifying and preventive treatment strategies.

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

Figure 1: The timeline of development.
Figure 2: Proposed causal chain of events.
Figure 3: Developmental priming pathways.

Similar content being viewed by others

References

  1. Roberts, G. W., Royston, M. C. & Götz, M. Pathology of cortical development and neuropsychiatric disorders. Ciba Found. Symp. 193, 296–321 (1995).

    CAS  PubMed  Google Scholar 

  2. Ploeger, A., Raijmakers, M. E., van der Maas, H. L. & Galis, F. The association between autism and errors in early embryogenesis: what is the causal mechanism? Biol. Psychiatry 67, 602–607 (2010).

    Article  PubMed  Google Scholar 

  3. Schmidt-Kastner, R., Van, O. J., Esquivel, G., Steinbusch, H. W. & Rutten, B. P. An environmental analysis of genes associated with schizophrenia: hypoxia and vascular factors as interacting elements in the neurodevelopmental model. Mol. Psychiatry 17, 1194–1205 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Stiles, J. & Jernigan, T. L. The basics of brain development. Neuropsychol. Rev. 20, 327–348 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Atladottir, H. O. et al. Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders. J. Autism Dev. Disord. 40, 1423–1430 (2010).

    Article  PubMed  Google Scholar 

  6. Brown, A. S. Epidemiologic studies of exposure to prenatal infection and risk of schizophrenia and autism. Dev. Neurobiol. 72, 1272–1276 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Fatemi, S. H., Cuadra, A. E., El-Fakahany, E. E., Sidwell, R. W. & Thuras, P. Prenatal viral infection causes alterations in nNOS expression in developing mouse brains. Neuroreport 11, 1493–1496 (2000).

    Article  CAS  PubMed  Google Scholar 

  8. Fatemi, S. H. et al. Prenatal viral infection leads to pyramidal cell atrophy and macrocephaly in adulthood: implications for genesis of autism and schizophrenia. Cell. Mol. Neurobiol. 22, 25–33 (2002).

    Article  PubMed  Google Scholar 

  9. Fatemi, S. H. et al. Defective corticogenesis and reduction in Reelin immunoreactivity in cortex and hippocampus of prenatally infected neonatal mice. Mol. Psychiatry 4, 145–154 (1999).

    Article  CAS  PubMed  Google Scholar 

  10. Fatemi, S. H. et al. Human influenza viral infection in utero alters glial fibrillary acidic protein immunoreactivity in the developing brains of neonatal mice. Mol. Psychiatry 7, 633–640 (2002).

    Article  CAS  PubMed  Google Scholar 

  11. Urakubo, A., Jarskog, L. F., Lieberman, J. A. & Gilmore, J. H. Prenatal exposure to maternal infection alters cytokine expression in the placenta, amniotic fluid, and fetal brain. Schizophr. Res. 47, 27–36 (2001).

    Article  CAS  PubMed  Google Scholar 

  12. Meyer, U., Feldon, J., Schedlowski, M. & Yee, B. K. Towards an immuno-precipitated neurodevelopmental animal model of schizophrenia. Neurosci. Biobehav. Rev. 29, 913–947 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Shi, L., Fatemi, S. H., Sidwell, R. W. & Patterson, P. H. Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J. Neurosci. 23, 297–302 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Zuckerman, L., Rehavi, M., Nachman, R. & Weiner, I. Immune activation during pregnancy in rats leads to a postpubertal emergence of disrupted latent inhibition, dopaminergic hyperfunction, and altered limbic morphology in the offspring: a novel neurodevelopmental model of schizophrenia. Neuropsychopharmacology 28, 1778–1789 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Meyer, U., Nyffeler, M., Yee, B. K., Knuesel, I. & Feldon, J. Adult brain and behavioral pathological markers of prenatal immune challenge during early/middle and late fetal development in mice. Brain Behav. Immun. 22, 469–486 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Brown, A. S. & Derkits, E. J. Prenatal infection and schizophrenia: a review of epidemiologic and translational studies. Am. J. Psychiatry 167, 261–280 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Brown, A. S. et al. Serologic evidence of prenatal influenza in the etiology of schizophrenia. Arch. Gen. Psychiatry 61, 774–780 (2004).

    Article  PubMed  Google Scholar 

  18. Brown, A. S. et al. Elevated maternal interleukin-8 levels and risk of schizophrenia in adult offspring. Am. J. Psychiatry 161, 889–895 (2004).

    Article  PubMed  Google Scholar 

  19. Brown, A. S. et al. Prenatal exposure to maternal infection and executive dysfunction in adult schizophrenia. Am. J. Psychiatry 166, 683–690 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Mednick, S. A., Machon, R. A., Huttunen, M. O. & Bonett, D. Adult schizophrenia following prenatal exposure to an influenza epidemic. Arch. Gen. Psychiatry 45, 189–192 (1988).

    Article  CAS  PubMed  Google Scholar 

  21. Mortensen, P. B. et al. Toxoplasma gondii as a risk factor for early-onset schizophrenia: analysis of filter paper blood samples obtained at birth. Biol. Psychiatry 61, 688–693 (2007).

    Article  PubMed  Google Scholar 

  22. Xiao, J. et al. Serological pattern consistent with infection with type I Toxoplasma gondii in mothers and risk of psychosis among adult offspring. Microbes Infect. 11, 1011–1018 (2009).

    Article  CAS  PubMed  Google Scholar 

  23. Buka, S. L., Cannon, T. D., Torrey, E. F. & Yolken, R. H. Maternal exposure to herpes simplex virus and risk of psychosis among adult offspring. Biol. Psychiatry 63, 809–815 (2008).

    Article  PubMed  Google Scholar 

  24. Mortensen, P. B. et al. A Danish National Birth Cohort study of maternal HSV-2 antibodies as a risk factor for schizophrenia in their offspring. Schizophr. Res. 122, 257–263 (2010).

    Article  PubMed  Google Scholar 

  25. Torrey, E. F., Bartko, J. J., Lun, Z. R. & Yolken, R. H. Antibodies to Toxoplasma gondii in patients with schizophrenia: a meta-analysis. Schizophr. Bull. 33, 729–736 (2007).

    Article  PubMed  Google Scholar 

  26. Torrey, E. F., Bartko, J. J. & Yolken, R. H. Toxoplasma gondii and other risk factors for schizophrenia: an update. Schizophr. Bull. 38, 642–647 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Clarke, M. C., Tanskanen, A., Huttunen, M., Whittaker, J. C. & Cannon, M. Evidence for an interaction between familial liability and prenatal exposure to infection in the causation of schizophrenia. Am. J. Psychiatry 166, 1025–1030 (2009).

    Article  PubMed  Google Scholar 

  28. Nielsen, P. R., Laursen, T. M. & Mortensen, P. B. Association between parental hospital-treated infection and the risk of schizophrenia in adolescence and early adulthood. Schizophr. Bull. 39, 230–237 (2013).

    Article  PubMed  Google Scholar 

  29. Sorensen, H. J., Mortensen, E. L., Reinisch, J. M. & Mednick, S. A. Association between prenatal exposure to bacterial infection and risk of schizophrenia. Schizophr. Bull. 35, 631–637 (2009).

    Article  PubMed  Google Scholar 

  30. Brown, A. S. et al. Elevated maternal C-reactive protein and autism in a national birth cohort. Mol. Psychiatry 19, 259–264 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Buka, S. L. et al. Maternal cytokine levels during pregnancy and adult psychosis. Brain Behav. Immun. 15, 411–420 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Canetta, S. et al. Elevated maternal C-reactive protein and increased risk of schizophrenia in a national birth cohort. Am. J. Psychiatry 171, 960–968 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Brown, A. S. et al. Prenatal infection and cavum septum pellucidum in adult schizophrenia. Schizophr. Res. 108, 285–287 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Dogan, Y. et al. Intracranial ultrasound abnormalities and fetal cytomegalovirus infection: report of 8 cases and review of the literature. Fetal Diagn. Ther. 30, 141–149 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Ellman, L. M. et al. Structural brain alterations in schizophrenia following fetal exposure to the inflammatory cytokine interleukin-8. Schizophr. Res. 121, 46–54 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Bayer, T. A., Buslei, R., Havas, L. & Falkai, P. Evidence for activation of microglia in patients with psychiatric illnesses. Neurosci. Lett. 271, 126–128 (1999).

    Article  CAS  PubMed  Google Scholar 

  37. Radewicz, K., Garey, L. J., Gentleman, S. M. & Reynolds, R. Increase in HLA-DR immunoreactive microglia in frontal and temporal cortex of chronic schizophrenics. J. Neuropathol. Exp. Neurol. 59, 137–150 (2000).

    Article  CAS  PubMed  Google Scholar 

  38. Upthegrove, R., Manzanares-Teson, N. & Barnes, N. M. Cytokine function in medication-naive first episode psychosis: a systematic review and meta-analysis. Schizophr. Res. 155, 101–108 (2014).

    Article  PubMed  Google Scholar 

  39. Yamashita, Y., Fujimoto, C., Nakajima, E., Isagai, T. & Matsuishi, T. Possible association between congenital cytomegalovirus infection and autistic disorder. J. Autism Dev. Disord. 33, 455–459 (2003).

    Article  PubMed  Google Scholar 

  40. Abdallah, M. W. et al. Amniotic fluid chemokines and autism spectrum disorders: an exploratory study utilizing a Danish Historic Birth Cohort. Brain Behav. Immun. 26, 170–176 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Abdallah, M. W. et al. Amniotic fluid inflammatory cytokines: potential markers of immunologic dysfunction in autism spectrum disorders. World J. Biol. Psychiatry 14, 528–538 (2013).

    Article  PubMed  Google Scholar 

  42. Braunschweig, D. et al. Autism: maternally derived antibodies specific for fetal brain proteins. Neurotoxicology 29, 226–231 (2008).

    CAS  PubMed  Google Scholar 

  43. Dalton, P. et al. Maternal neuronal antibodies associated with autism and a language disorder. Ann. Neurol. 53, 533–537 (2003).

    Article  PubMed  Google Scholar 

  44. Singer, H. S. et al. Antibodies against fetal brain in sera of mothers with autistic children. J. Neuroimmunol. 194, 165–172 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Zerbo, O. et al. Is maternal influenza or fever during pregnancy associated with autism or developmental delays? Results from the CHARGE (CHildhood Autism Risks from Genetics and Environment) study. J. Autism Dev. Disord. 43, 25–33 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Zerbo, O., Iosif, A. M., Delwiche, L., Walker, C. & Hertz-Picciotto, I. Month of conception and risk of autism. Epidemiology 22, 469–475 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Morgan, J. T. et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol. Psychiatry 68, 368–376 (2010).

    Article  PubMed  Google Scholar 

  48. Suzuki, K. et al. Microglial activation in young adults with autism spectrum disorder. JAMA Psychiatry 70, 49–58 (2013).

    Article  PubMed  Google Scholar 

  49. Vargas, D. L., Nascimbene, C., Krishnan, C., Zimmerman, A. W. & Pardo, C. A. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann. Neurol. 57, 67–81 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Chez, M. G., Dowling, T., Patel, P. B., Khanna, P. & Kominsky, M. Elevation of tumor necrosis factor-alpha in cerebrospinal fluid of autistic children. Pediatr. Neurol. 36, 361–365 (2007).

    Article  PubMed  Google Scholar 

  51. Pardo, C. A., Vargas, D. L. & Zimmerman, A. W. Immunity, neuroglia and neuroinflammation in autism. Int. Rev. Psychiatry 17, 485–495 (2005).

    Article  PubMed  Google Scholar 

  52. Parker-Athill, E. C. & Tan, J. Maternal immune activation and autism spectrum disorder: interleukin-6 signaling as a key mechanistic pathway. Neurosignals 18, 113–128 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Goudriaan, A. et al. Specific glial functions contribute to schizophrenia susceptibility. Schizophr. Bull. 40, 925–935 (2014).

    Article  PubMed  Google Scholar 

  54. Stefansson, H. et al. Common variants conferring risk of schizophrenia. Nature 460, 744–747 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014).

  56. Voineagu, I. et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature 474, 380–384 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Drexhage, R. C. et al. The mononuclear phagocyte system and its cytokine inflammatory networks in schizophrenia and bipolar disorder. Expert Rev. Neurother. 10, 59–76 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Jyonouchi, H., Geng, L., Streck, D. L. & Toruner, G. A. Children with autism spectrum disorders (ASD) who exhibit chronic gastrointestinal (GI) symptoms and marked fluctuation of behavioral symptoms exhibit distinct innate immune abnormalities and transcriptional profiles of peripheral blood (PB) monocytes. J. Neuroimmunol. 238, 73–80 (2011).

    Article  CAS  PubMed  Google Scholar 

  59. Cardno, A. G. & Gottesman, I. I. Twin studies of schizophrenia: from bow-and-arrow concordances to star wars Mx and functional genomics. Am. J. Med. Genet. 97, 12–17 (2000).

    Article  CAS  PubMed  Google Scholar 

  60. Ben-David, E. & Shifman, S. Networks of neuronal genes affected by common and rare variants in autism spectrum disorders. PLoS Genet. 8, e1002556 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Smoller, J. W. et al. Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet 381, 1371–1379 (2013).

    Article  CAS  Google Scholar 

  62. Talkowski, M. E. et al. Sequencing chromosomal abnormalities reveals neurodevelopmental loci that confer risk across diagnostic boundaries. Cell 149, 525–537 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Fromer, M. et al. De novo mutations in schizophrenia implicate synaptic networks. Nature 506, 179–184 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Gulsuner, S. et al. Spatial and temporal mapping of de novo mutations in schizophrenia to a fetal prefrontal cortical network. Cell 154, 518–529 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Torrey, E. F., Miller, J., Rawlings, R. & Yolken, R. H. Seasonal birth patterns of neurological disorders. Neuroepidemiology 19, 177–185 (2000).

    Article  CAS  PubMed  Google Scholar 

  66. Sun, Y., Vestergaard, M., Christensen, J., Nahmias, A. J. & Olsen, J. Prenatal exposure to maternal infections and epilepsy in childhood: a population-based cohort study. Pediatrics 121, e1100–e1107 (2008).

    Article  PubMed  Google Scholar 

  67. Sun, Y., Vestergaard, M., Christensen, J. & Olsen, J. Prenatal exposure to elevated maternal body temperature and risk of epilepsy in childhood: a population-based pregnancy cohort study. Paediatr. Perinat. Epidemiol. 25, 53–59 (2011).

    Article  PubMed  Google Scholar 

  68. Whitehead, E. et al. Relation of pregnancy and neonatal factors to subsequent development of childhood epilepsy: a population-based cohort study. Pediatrics 117, 1298–1306 (2006).

    Article  PubMed  Google Scholar 

  69. Hirvonen, J. et al. Increased in vivo expression of an inflammatory marker in temporal lobe epilepsy. J. Nucl. Med. 53, 234–240 (2012).

    Article  CAS  PubMed  Google Scholar 

  70. During, M. J. & Spencer, D. D. Extracellular hippocampal glutamate and spontaneous seizure in the conscious human brain. Lancet 341, 1607–1610 (1993).

    Article  CAS  PubMed  Google Scholar 

  71. Kraguljac, N. V., Reid, M. A., White, D. M., den Hollander, J. & Lahti, A. C. Regional decoupling of N-acetyl-aspartate and glutamate in schizophrenia. Neuropsychopharmacology 37, 2635–2642 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Small, S. A., Schobel, S. A., Buxton, R. B., Witter, M. P. & Barnes, C. A. A pathophysiological framework of hippocampal dysfunction in ageing and disease. Nat. Rev. Neurosci. 12, 585–601 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Fatemi, S. H. et al. Human influenza viral infection in utero increases nNOS expression in hippocampi of neonatal mice. Synapse 29, 84–88 (1998).

    Article  CAS  PubMed  Google Scholar 

  74. Smith, S. E., Li, J., Garbett, K., Mirnics, K. & Patterson, P. H. Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. 27, 10695–10702 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Meyer, U. Developmental neuroinflammation and schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 42, 20–34 (2013).

    Article  CAS  PubMed  Google Scholar 

  76. Harvey, L. & Boksa, P. Prenatal and postnatal animal models of immune activation: relevance to a range of neurodevelopmental disorders. Dev. Neurobiol. 72, 1335–1348 (2012).

    Article  CAS  PubMed  Google Scholar 

  77. Kneeland, R. E. & Fatemi, S. H. Viral infection, inflammation and schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 42, 35–48 (2013).

    Article  CAS  PubMed  Google Scholar 

  78. Meyer, U. Prenatal poly(i:C) exposure and other developmental immune activation models in rodent systems. Biol. Psychiatry 75, 307–315 (2014).

    Article  CAS  PubMed  Google Scholar 

  79. Meyer, U. & Feldon, J. To poly(I:C) or not to poly(I:C): advancing preclinical schizophrenia research through the use of prenatal immune activation models. Neuropharmacology 62, 1308–1321 (2012).

    Article  CAS  PubMed  Google Scholar 

  80. Patterson, P. H. Maternal infection and immune involvement in autism. Trends Mol. Med. 17, 389–394 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Malkova, N. V., Yu, C. Z., Hsiao, E. Y., Moore, M. J. & Patterson, P. H. Maternal immune activation yields offspring displaying mouse versions of the three core symptoms of autism. Brain Behav. Immun. 26, 607–616 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Bauman, M. D. et al. Activation of the maternal immune system during pregnancy alters behavioral development of rhesus monkey offspring. Biol. Psychiatry 75, 332–341 (2014).

    Article  CAS  PubMed  Google Scholar 

  83. Martin, L. A. et al. Stereotypies and hyperactivity in rhesus monkeys exposed to IgG from mothers of children with autism. Brain Behav. Immun. 22, 806–816 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Bauman, M. D. et al. Maternal antibodies from mothers of children with autism alter brain growth and social behavior development in the rhesus monkey. Transl. Psychiatry 3, e278 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Qulu, L., Daniels, W. M. & Mabandla, M. V. Exposure to prenatal stress enhances the development of seizures in young rats. Metab. Brain Dis. 27, 399–404 (2012).

    Article  CAS  PubMed  Google Scholar 

  86. Pineda, E. et al. Maternal immune activation promotes hippocampal kindling epileptogenesis in mice. Ann. Neurol. 74, 11–19 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Hawkes, C. H., Del, T. K. & Braak, H. A timeline for Parkinson's disease. Parkinsonism Relat. Disord. 16, 79–84 (2010).

    Article  PubMed  Google Scholar 

  88. Musiek, E. S. & Holtzman, D. M. Origins of Alzheimer's disease: reconciling cerebrospinal fluid biomarker and neuropathology data regarding the temporal sequence of amyloid-beta and tau involvement. Curr. Opin. Neurol. 25, 715–720 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Jacobsson, B. & Hagberg, G. Antenatal risk factors for cerebral palsy. Best Pract. Res. Clin. Obstet. Gynaecol. 18, 425–436 (2004).

    Article  PubMed  Google Scholar 

  90. Ribiani, E. et al. Perinatal infections and cerebral palsy. Minerva Ginecol. 59, 151–157 (2007).

    CAS  PubMed  Google Scholar 

  91. Leviton, A. & Dammann, O. Coagulation, inflammation, and the risk of neonatal white matter damage. Pediatr. Res. 55, 541–545 (2004).

    Article  PubMed  Google Scholar 

  92. Nelson, K. B. & Lynch, J. K. Stroke in newborn infants. Lancet Neurol. 3, 150–158 (2004).

    Article  PubMed  Google Scholar 

  93. Wu, Y. W. et al. Perinatal stroke in children with motor impairment: a population-based study. Pediatrics 114, 612–619 (2004).

    Article  PubMed  Google Scholar 

  94. Savman, K., Heyes, M. P., Svedin, P. & Karlsson, A. Microglia/macrophage-derived inflammatory mediators galectin-3 and quinolinic acid are elevated in cerebrospinal fluid from newborn infants after birth asphyxia. Transl. Stroke Res. 4, 228–235 (2013).

    Article  CAS  PubMed  Google Scholar 

  95. Dammann, O. & Leviton, A. Maternal intrauterine infection, cytokines, and brain damage in the preterm newborn. Pediatr. Res. 42, 1–8 (1997).

    Article  CAS  PubMed  Google Scholar 

  96. Grether, J. K. & Nelson, K. B. Maternal infection and cerebral palsy in infants of normal birth weight. JAMA 278, 207–211 (1997).

    Article  CAS  PubMed  Google Scholar 

  97. Hagberg, H., Mallard, C. & Jacobsson, B. Role of cytokines in preterm labour and brain injury. BJOG 112 (Suppl. 1), 16–18 (2005).

    Article  CAS  PubMed  Google Scholar 

  98. Fleiss, B. & Gressens, P. Tertiary mechanisms of brain damage: a new hope for treatment of cerebral palsy? Lancet Neurol. 11, 556–566 (2012).

    Article  PubMed  Google Scholar 

  99. Ahlin, K. et al. Cerebral palsy and perinatal infection in children born at term. Obstet. Gynecol. 122, 41–49 (2013).

    Article  PubMed  Google Scholar 

  100. Hermansen, M. C. & Hermansen, M. G. Perinatal infections and cerebral palsy. Clin. Perinatol. 33, 315–333 (2006).

    Article  PubMed  Google Scholar 

  101. Andrews, W. W. et al. Early preterm birth: association between in utero exposure to acute inflammation and severe neurodevelopmental disability at 6 years of age. Am. J. Obstet. Gynecol. 198, 466–466 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  102. O'Callaghan, M. E., MacLennan, A. H., Haan, E. A. & Dekker, G. The genomic basis of cerebral palsy: a HuGE systematic literature review. Hum. Genet. 126, 149–172 (2009).

    Article  CAS  PubMed  Google Scholar 

  103. Clouchoux, C. & Limperopoulos, C. Novel applications of quantitative MRI for the fetal brain. Pediatr. Radiol. 42 (Suppl. 1), S24–S32 (2012).

    Article  PubMed  Google Scholar 

  104. Girard, N. J. & Chaumoitre, K. The brain in the belly: what and how of fetal neuroimaging? J. Magn. Reson. Imaging 36, 788–804 (2012).

    Article  PubMed  Google Scholar 

  105. Gousias, I. S. et al. Magnetic resonance imaging of the newborn brain: automatic segmentation of brain images into 50 anatomical regions. PLoS ONE 8, e59990 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Shrivastava, K. et al. Temporal expression of cytokines and signal transducer and activator of transcription factor 3 activation after neonatal hypoxia/ischemia in mice. Dev. Neurosci. 35, 212–225 (2013).

    Article  CAS  PubMed  Google Scholar 

  107. Cai, Z., Pan, Z. L., Pang, Y., Evans, O. B. & Rhodes, P. G. Cytokine induction in fetal rat brains and brain injury in neonatal rats after maternal lipopolysaccharide administration. Pediatr. Res. 47, 64–72 (2000).

    Article  CAS  PubMed  Google Scholar 

  108. Larouche, A. et al. Neuronal injuries induced by perinatal hypoxic–ischemic insults are potentiated by prenatal exposure to lipopolysaccharide: animal model for perinatally acquired encephalopathy. Dev. Neurosci. 27, 134–142 (2005).

    Article  CAS  PubMed  Google Scholar 

  109. Paintlia, M. K., Paintlia, A. S., Barbosa, E., Singh, I. & Singh, A. K. N-acetylcysteine prevents endotoxin-induced degeneration of oligodendrocyte progenitors and hypomyelination in developing rat brain. J. Neurosci. Res. 78, 347–361 (2004).

    Article  CAS  PubMed  Google Scholar 

  110. Rousset, C. I. et al. Maternal exposure to LPS induces hypomyelination in the internal capsule and programmed cell death in the deep gray matter in newborn rats. Pediatr. Res. 59, 428–433 (2006).

    Article  CAS  PubMed  Google Scholar 

  111. Rousset, C. I. et al. Antenatal bacterial endotoxin sensitizes the immature rat brain to postnatal excitotoxic injury. J. Neuropathol. Exp. Neurol. 67, 994–1000 (2008).

    Article  PubMed  Google Scholar 

  112. Bell, M. J. & Hallenbeck, J. M. Effects of intrauterine inflammation on developing rat brain. J. Neurosci. Res. 70, 570–579 (2002).

    Article  CAS  PubMed  Google Scholar 

  113. Saadani-Makki, F. et al. Intrauterine administration of endotoxin leads to motor deficits in a rabbit model: a link between prenatal infection and cerebral palsy. Am. J. Obstet. Gynecol. 199, 651–657 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Stridh, L. et al. Toll-like receptor-3 activation increases the vulnerability of the neonatal brain to hypoxia-ischemia. J. Neurosci. 33, 12041–12051 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Dean, D. C. 3rd et al. Brain differences in infants at differential genetic risk for late-onset Alzheimer disease: a cross-sectional imaging study. JAMA Neurol. 71, 11–22 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Knickmeyer, R. C. et al. Common variants in psychiatric risk genes predict brain structure at birth. Cereb. Cortex 24, 1230–1246 (2014).

    Article  PubMed  Google Scholar 

  117. Braak, H. & Del Tredici, K. The pathological process underlying Alzheimer's disease in individuals under thirty. Acta Neuropathol. 121, 171–181 (2011).

    Article  PubMed  Google Scholar 

  118. Lambert, J. C. et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease. Nat. Genet. 41, 1094–1099 (2009).

    Article  CAS  PubMed  Google Scholar 

  119. Chen, L. H. et al. Polymorphisms of CR1, CLU and PICALM confer susceptibility of Alzheimer's disease in a southern Chinese population. Neurobiol. Aging 33, 210.e1–210.e7 (2012).

    Article  CAS  Google Scholar 

  120. Jonsson, T. et al. Variant of TREM2 associated with the risk of Alzheimer's disease. N. Engl. J. Med. 368, 107–116 (2013).

    Article  CAS  PubMed  Google Scholar 

  121. Guerreiro, R. & Hardy, J. TREM2 and neurodegenerative disease. N. Engl. J. Med. 369, 1569–1570 (2013).

    CAS  PubMed  Google Scholar 

  122. Ray, S. et al. Classification and prediction of clinical Alzheimer's diagnosis based on plasma signaling proteins. Nat. Med. 13, 1369–1362 (2007).

    Article  CAS  Google Scholar 

  123. Britschgi, M. et al. Modeling of pathological traits in Alzheimer's disease based on systemic extracellular signaling proteome. Mol. Cell. Proteomics 10, M111.008862 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Hu, W. T. et al. Novel CSF biomarkers for Alzheimer's disease and mild cognitive impairment. Acta Neuropathol. 119, 669–678 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Bertram, L., McQueen, M. B., Mullin, K., Blacker, D. & Tanzi, R. E. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat. Genet. 39, 17–23 (2007).

    Article  CAS  PubMed  Google Scholar 

  126. Takahashi, K., Prinz, M., Stagi, M., Chechneva, O. & Neumann, H. TREM2-transduced myeloid precursors mediate nervous tissue debris clearance and facilitate recovery in an animal model of multiple sclerosis. PLoS Med. 4, e124 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Glass, C. K., Saijo, K., Winner, B., Marchetto, M. C. & Gage, F. H. Mechanisms underlying inflammation in neurodegeneration. Cell 140, 918–934 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Krstic, D. et al. Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice. J. Neuroinflammation 9, 151 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Hao, L. Y., Hao, X. Q., Li, S. H. & Li, X. H. Prenatal exposure to lipopolysaccharide results in cognitive deficits in age-increasing offspring rats. Neuroscience 166, 763–770 (2010).

    Article  CAS  PubMed  Google Scholar 

  130. Bilbo, S. D. Early-life infection is a vulnerability factor for aging-related glial alterations and cognitive decline. Neurobiol. Learn. Mem. 94, 57–64 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  131. Bitanihirwe, B. K., 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 35, 2462–2478 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Knuesel, I. et al. Age-related accumulation of Reelin in amyloid-like deposits. Neurobiol. Aging 30, 697–716 (2009).

    Article  CAS  PubMed  Google Scholar 

  133. Richetto, J., Calabrese, F., Riva, M. A. & Meyer, U. Prenatal immune activation induces maturation-dependent alterations in the prefrontal GABAergic transcriptome. Schizophr. Bull. 40, 351–361 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  134. Doehner, J., Genoud, C., Imhof, C., Krstic, D. & Knuesel, I. Extrusion of misfolded and aggregated protein—a protective strategy of aging neurons? Eur. J. Neurosci. 35, 1938–1950 (2012).

    Article  PubMed  Google Scholar 

  135. Krstic, D. & Knuesel, I. Deciphering the mechanism underlying late-onset Alzheimer disease. Nat. Rev. Neurol. 9, 25–34 (2013).

    Article  CAS  PubMed  Google Scholar 

  136. Mattock, C., Marmot, M. & Stern, G. Could Parkinson's disease follow intra-uterine influenza? A speculative hypothesis. J. Neurol. Neurosurg. Psychiatry 51, 753–756 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Ebmeier, K. P. et al. Does idiopathic parkinsonism in Aberdeen follow intrauterine influenza? J. Neurol. Neurosurg. Psychiatry 52, 911–913 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Toovey, S., Jick, S. S. & Meier, C. R. Parkinson's disease or Parkinson symptoms following seasonal influenza. Influenza Other Respir. Viruses 5, 328–333 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  139. Hardy, J., Lewis, P., Revesz, T., Lees, A. & Paisan-Ruiz, C. The genetics of Parkinson's syndromes: a critical review. Curr. Opin. Genet. Dev. 19, 254–265 (2009).

    Article  CAS  PubMed  Google Scholar 

  140. Lema Tome, C. M. et al. Inflammation and alpha-synuclein's prion-like behavior in Parkinson's disease—is there a link? Mol. Neurobiol. 47, 561–574 (2013).

    Article  CAS  PubMed  Google Scholar 

  141. Snyder-Keller, A. & Stark, P. F. Prenatal inflammatory effects on nigrostriatal development in organotypic cultures. Brain Res. 1233, 160–167 (2008).

    Article  CAS  PubMed  Google Scholar 

  142. Carvey, P. M., Chang, Q., Lipton, J. W. & Ling, Z. Prenatal exposure to the bacteriotoxin lipopolysaccharide leads to long-term losses of dopamine neurons in offspring: a potential, new model of Parkinson's disease. Front. Biosci. 8, s826–s837 (2003).

    Article  CAS  PubMed  Google Scholar 

  143. Vuillermot, S. et al. Prenatal immune activation interacts with genetic Nurr1 deficiency in the development of attentional impairments. J. Neurosci. 32, 436–451 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Saucedo-Cardenas, O. et al. Nurr1 is essential for the induction of the dopaminergic phenotype and the survival of ventral mesencephalic late dopaminergic precursor neurons. Proc. Natl Acad. Sci. USA 95, 4013–4018 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Saijo, K. et al. A Nurr1/CoREST pathway in microglia and astrocytes protects dopaminergic neurons from inflammation-induced death. Cell 137, 47–59 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Garbett, K. A., Hsiao, E. Y., Kalman, S., Patterson, P. H. & Mirnics, K. Effects of maternal immune activation on gene expression patterns in the fetal brain. Transl. Psychiatry 2, e98 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. van Noort, J. M. et al. αβ-Crystallin is a target for adaptive immune responses and a trigger of innate responses in preactive multiple sclerosis lesions. J. Neuropathol. Exp. Neurol. 69, 694–703 (2010).

    Article  CAS  PubMed  Google Scholar 

  148. Braak, H., Del Tredici, K., Sandmann-Kiel, D., Rub, U. & Schultz, C. Nerve cells expressing heat-shock proteins in Parkinson's disease. Acta Neuropathol. 102, 449–454 (2001).

    CAS  PubMed  Google Scholar 

  149. Jang, H. et al. Highly pathogenic H5N1 influenza virus can enter the central nervous system and induce neuroinflammation and neurodegeneration. Proc. Natl Acad. Sci. USA 106, 14063–14068 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Ginhoux, F. et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Alliot, F., Godin, I. & Pessac, B. Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Brain Res. Dev. Brain Res. 117, 145–152 (1999).

    Article  CAS  PubMed  Google Scholar 

  152. Hutchins, K. D., Dickson, D. W., Rashbaum, W. K. & Lyman, W. D. Localization of morphologically distinct microglial populations in the developing human fetal brain: implications for ontogeny. Brain Res. Dev. Brain Res. 55, 95–102 (1990).

    Article  CAS  PubMed  Google Scholar 

  153. Bhat, R. & Steinman, L. Innate and adaptive autoimmunity directed to the central nervous system. Neuron 64, 123–132 (2009).

    Article  CAS  PubMed  Google Scholar 

  154. Hagberg, H., Gressens, P. & Mallard, C. Inflammation during fetal and neonatal life: implications for neurologic and neuropsychiatric disease in children and adults. Ann. Neurol. 71, 444–457 (2012).

    Article  PubMed  Google Scholar 

  155. Aguzzi, A., Barres, B. A. & Bennett, M. L. Microglia: scapegoat, saboteur, or something else? Science 339, 156–161 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Boulanger, L. M. & Shatz, C. J. Immune signalling in neural development, synaptic plasticity and disease. Nat. Rev. Neurosci. 5, 521–531 (2004).

    Article  CAS  PubMed  Google Scholar 

  157. Marin, I. & Kipnis, J. Learning and memory ... and the immune system. Learn. Mem. 20, 601–606 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Schafer, D. P. et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Stevens, B. et al. The classical complement cascade mediates CNS synapse elimination. Cell 131, 1164–1178 (2007).

    Article  CAS  PubMed  Google Scholar 

  160. Ueno, M. & Yamashita, T. Bidirectional tuning of microglia in the developing brain: from neurogenesis to neural circuit formation. Curr. Opin. Neurobiol. 27C, 8–15 (2014).

    Article  CAS  Google Scholar 

  161. Xu, D. X. et al. Tumor necrosis factor alpha partially contributes to lipopolysaccharide-induced intra-uterine fetal growth restriction and skeletal development retardation in mice. Toxicol. Lett. 163, 20–29 (2006).

    Article  CAS  PubMed  Google Scholar 

  162. Meyer, U. et al. Adult behavioral and pharmacological dysfunctions following disruption of the fetal brain balance between pro-inflammatory and IL-10-mediated anti-inflammatory signaling. Mol. Psychiatry 13, 208–221 (2008).

    Article  CAS  PubMed  Google Scholar 

  163. Hsiao, E. Y., McBride, S. W., Chow, J., Mazmanian, S. K. & Patterson, P. H. Modeling an autism risk factor in mice leads to permanent immune dysregulation. Proc. Natl Acad. Sci. USA 109, 12776–12781 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Zager, A., Pinheiro, M. L., Ferraz-de-Paula, V., Ribeiro, A. & Palermo-Neto, J. Increased cell-mediated immunity in male mice offspring exposed to maternal immune activation during late gestation. Int. Immunopharmacol. 17, 633–637 (2013).

    Article  CAS  PubMed  Google Scholar 

  165. Stolp, H. B. & Dziegielewska, K. M. Review: role of developmental inflammation and blood-brain barrier dysfunction in neurodevelopmental and neurodegenerative diseases. Neuropathol. Appl. Neurobiol. 35, 132–146 (2009).

    Article  CAS  PubMed  Google Scholar 

  166. Theoharides, T. C., Kempuraj, D. & Redwood, L. Autism: an emerging 'neuroimmune disorder' in search of therapy. Expert Opin. Pharmacother. 10, 2127–2143 (2009).

    Article  CAS  PubMed  Google Scholar 

  167. Makinodan, M. et al. Maternal immune activation in mice delays myelination and axonal development in the hippocampus of the offspring. J. Neurosci. Res. 86, 2190–2200 (2008).

    Article  CAS  PubMed  Google Scholar 

  168. Li, Q. et al. Voxel-based analysis of postnatal white matter microstructure in mice exposed to immune challenge in early or late pregnancy. Neuroimage 52, 1–8 (2010).

    Article  CAS  PubMed  Google Scholar 

  169. Corriveau, R. A., Huh, G. S. & Shatz, C. J. Regulation of class I MHC gene expression in the developing and mature CNS by neural activity. Neuron 21, 505–520 (1998).

    Article  CAS  PubMed  Google Scholar 

  170. Glynn, M. W. et al. MHCI negatively regulates synapse density during the establishment of cortical connections. Nat. Neurosci. 14, 442–451 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Huh, G. S. et al. Functional requirement for class I MHC in CNS development and plasticity. Science 290, 2155–2159 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Stephan, A. H. et al. A dramatic increase of C1q protein in the CNS during normal aging. J. Neurosci. 33, 13460–13474 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Chu, Y. et al. Enhanced synaptic connectivity and epilepsy in C1q knockout mice. Proc. Natl Acad. Sci. USA 107, 7975–7980 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Lee, H. et al. Synapse elimination and learning rules co-regulated by MHC class I H2-Db. Nature 509, 195–200 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Elmer, B. M., Estes, M. L., Barrow, S. L. & McAllister, A. K. MHCI requires MEF2 transcription factors to negatively regulate synapse density during development and in disease. J. Neurosci. 33, 13791–13804 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Ozawa, K. et al. Immune activation during pregnancy in mice leads to dopaminergic hyperfunction and cognitive impairment in the offspring: a neurodevelopmental animal model of schizophrenia. Biol. Psychiatry 59, 546–554 (2006).

    Article  CAS  PubMed  Google Scholar 

  177. Romero, E., Guaza, C., Castellano, B. & Borrell, J. Ontogeny of sensorimotor gating and immune impairment induced by prenatal immune challenge in rats: implications for the etiopathology of schizophrenia. Mol. Psychiatry 15, 372–383 (2010).

    Article  CAS  PubMed  Google Scholar 

  178. Dalton, V. S., Verdurand, M., Walker, A., Hodgson, D. M. & Zavitsanou, K. Synergistic effect between maternal infection and adolescent cannabinoid exposure on serotonin 5HT1A receptor binding in the hippocampus: testing the “two hit” hypothesis for the development of schizophrenia. ISRN Psychiatry 2012, 451865 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Lin, Y. L., Lin, S. Y. & Wang, S. Prenatal lipopolysaccharide exposure increases anxiety-like behaviors and enhances stress-induced corticosterone responses in adult rats. Brain Behav. Immun. 26, 459–468 (2012).

    Article  CAS  PubMed  Google Scholar 

  180. Nouel, D., Burt, M., Zhang, Y., Harvey, L. & Boksa, P. Prenatal exposure to bacterial endotoxin reduces the number of GAD67- and reelin-immunoreactive neurons in the hippocampus of rat offspring. Eur. Neuropsychopharmacol. 22, 300–307 (2012).

    Article  CAS  PubMed  Google Scholar 

  181. Nyffeler, M., Meyer, U., Yee, B. K., Feldon, J. & Knuesel, I. Maternal immune activation during pregnancy increases limbic GABAA receptor immunoreactivity in the adult offspring: implications for schizophrenia. Neuroscience 143, 51–62 (2006).

    Article  CAS  PubMed  Google Scholar 

  182. Roumier, A. et al. Prenatal activation of microglia induces delayed impairment of glutamatergic synaptic function. PLoS ONE 3, e2595 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Leviton, A. & Gressens, P. Neuronal damage accompanies perinatal white-matter damage. Trends Neurosci. 30, 473–478 (2007).

    Article  CAS  PubMed  Google Scholar 

  184. Meyer, U. et al. The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology. J. Neurosci. 26, 4752–4762 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Oskvig, D. B., Elkahloun, A. G., Johnson, K. R., Phillips, T. M. & Herkenham, M. Maternal immune activation by LPS selectively alters specific gene expression profiles of interneuron migration and oxidative stress in the fetus without triggering a fetal immune response. Brain Behav. Immun. 26, 623–634 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Folsom, T. D. & Fatemi, S. H. The involvement of Reelin in neurodevelopmental disorders. Neuropharmacology 68, 122–135 (2013).

    Article  CAS  PubMed  Google Scholar 

  187. Knuesel, I. Reelin-mediated signaling in neuropsychiatric and neurodegenerative diseases. Prog. Neurobiol. 91, 257–274 (2010).

    Article  CAS  PubMed  Google Scholar 

  188. Fatemi, S. H., Stary, J. M. & Egan, E. A. Reduced blood levels of reelin as a vulnerability factor in pathophysiology of autistic disorder. Cell. Mol. Neurobiol. 22, 139–152 (2002).

    Article  CAS  PubMed  Google Scholar 

  189. Haas, C. A. & Frotscher, M. Reelin deficiency causes granule cell dispersion in epilepsy. Exp. Brain Res. 200, 141–149 (2010).

    Article  PubMed  Google Scholar 

  190. Chin, J. et al. Reelin depletion in the entorhinal cortex of human amyloid precursor protein transgenic mice and humans with Alzheimer's disease. J. Neurosci. 27, 2727–2733 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Fatemi, S. H., Pearce, D. A., Brooks, A. I. & Sidwell, R. W. Prenatal viral infection in mouse causes differential expression of genes in brains of mouse progeny: a potential animal model for schizophrenia and autism. Synapse 57, 91–99 (2005).

    Article  CAS  PubMed  Google Scholar 

  192. Connor, C. M. et al. Maternal immune activation alters behavior in adult offspring, with subtle changes in the cortical transcriptome and epigenome. Schizophr. Res. 140, 175–184 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  193. Tang, B., Jia, H., Kast, R. J. & Thomas, E. A. Epigenetic changes at gene promoters in response to immune activation in utero. Brain Behav. Immun. 30, 168–175 (2013).

    Article  CAS  PubMed  Google Scholar 

  194. Pickett, J. Current investigations in autism brain tissue research. J. Autism Dev. Disord. 31, 521–527 (2001).

    Article  CAS  PubMed  Google Scholar 

  195. Martins-de-Souza, D. et al. Proteome analysis of the thalamus and cerebrospinal fluid reveals glycolysis dysfunction and potential biomarkers candidates for schizophrenia. J. Psychiatr. Res. 44, 1176–1189 (2010).

    Article  PubMed  Google Scholar 

  196. Renkawek, K., Voorter, C. E., Bosman, G. J., van Workum, F. P. & de Jong, W. W. Expression of alpha B-crystallin in Alzheimer's disease. Acta Neuropathol. 87, 155–160 (1994).

    Article  CAS  PubMed  Google Scholar 

  197. Perry, V. H. & Holmes, C. Microglial priming in neurodegenerative disease. Nat. Rev. Neurol. 10, 217–224 (2014).

    Article  CAS  PubMed  Google Scholar 

  198. Zhang, Z., Trautmann, K. & Schluesener, H. J. Microglia activation in rat spinal cord by systemic injection of TLR3 and TLR7/8 agonists. J. Neuroimmunol. 164, 154–160 (2005).

    Article  CAS  PubMed  Google Scholar 

  199. Giovanoli, S. et al. Stress in puberty unmasks latent neuropathological consequences of prenatal immune activation in mice. Science 339, 1095–1099 (2013).

    Article  CAS  PubMed  Google Scholar 

  200. Juckel, G. et al. Microglial activation in a neuroinflammational animal model of schizophrenia—a pilot study. Schizophr. Res. 131, 96–100 (2011).

    Article  PubMed  Google Scholar 

  201. Abazyan, B. et al. Prenatal interaction of mutant DISC1 and immune activation produces adult psychopathology. Biol. Psychiatry 68, 1172–1181 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Lipina, T. V., Zai, C., Hlousek, D., Roder, J. C. & Wong, A. H. Maternal immune activation during gestation interacts with Disc1 point mutation to exacerbate schizophrenia-related behaviors in mice. J. Neurosci. 33, 7654–7666 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Ehninger, D. et al. Gestational immune activation and Tsc2 haploinsufficiency cooperate to disrupt fetal survival and may perturb social behavior in adult mice. Mol. Psychiatry 17, 62–70 (2012).

    Article  CAS  PubMed  Google Scholar 

  204. Burt, M. A., Tse, Y. C., Boksa, P. & Wong, T. P. Prenatal immune activation interacts with stress and corticosterone exposure later in life to modulate N-methyl-D-aspartate receptor synaptic function and plasticity. Int. J. Neuropsychopharmacol. 16, 1835–1848 (2013).

    Article  CAS  PubMed  Google Scholar 

  205. Deslauriers, J., Larouche, A., Sarret, P. & Grignon, S. Combination of prenatal immune challenge and restraint stress affects prepulse inhibition and dopaminergic/GABAergic markers. Prog. Neuropsychopharmacol. Biol. Psychiatry 45, 156–164 (2013).

    Article  CAS  PubMed  Google Scholar 

  206. Suvisaari, J. M., Haukka, J. K., Tanskanen, A. J. & Lonnqvist, J. K. Decline in the incidence of schizophrenia in Finnish cohorts born from 1954 to 1965. Arch. Gen. Psychiatry 56, 733–740 (1999).

    Article  CAS  PubMed  Google Scholar 

  207. Rook, G. A., Raison, C. L. & Lowry, C. A. Microbiota, immunoregulatory old friends and psychiatric disorders. Adv. Exp. Med. Biol. 817, 319–356 (2014).

    Article  CAS  PubMed  Google Scholar 

  208. Cono, J., Cragan, J. D., Jamieson, D. J. & Rasmussen, S. A. Prophylaxis and treatment of pregnant women for emerging infections and bioterrorism emergencies. Emerg. Infect. Dis. 12, 1631–1637 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  209. O'Grady, K. A. et al. FluMum: a prospective cohort study of mother-infant pairs assessing the effectiveness of maternal influenza vaccination in prevention of influenza in early infancy. BMJ Open 4, e005676 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  210. Trotta, F. et al. Evaluation of safety of A/H1N1 pandemic vaccination during pregnancy: cohort study. BMJ 348, g3361 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Cleary, B. J., Rice, U., Eogan, M., Metwally, N. & McAuliffe, F. 2009 A/H1N1 influenza vaccination in pregnancy: uptake and pregnancy outcomes—a historical cohort study. Eur. J. Obstet. Gynecol. Reprod. Biol. 178, 163–168 (2014).

    Article  PubMed  Google Scholar 

  212. Nahmias, A. J., Nahmias, S. B. & Danielsson, D. The possible role of transplacentally-acquired antibodies to infectious agents, with molecular mimicry to nervous system sialic acid epitopes, as causes of neuromental disorders: prevention and vaccine implications. Clin. Dev. Immunol. 13, 167–183 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Brown, A. S. & Patterson, P. H. Maternal infection and schizophrenia: implications for prevention. Schizophr. Bull. 37, 284–290 (2011).

    Article  PubMed  Google Scholar 

  214. Cervantes-Gonzalez, M. & Launay, O. Pandemic influenza A (H1N1) in pregnant women: impact of early diagnosis and antiviral treatment. Expert Rev. Anti Infect.Ther. 8, 981–984 (2010).

    Article  PubMed  Google Scholar 

  215. Petersen, I., Gilbert, R., Evans, S., Ridolfi, A. & Nazareth, I. Oral antibiotic prescribing during pregnancy in primary care: UK population-based study. J. Antimicrob. Chemother. 65, 2238–2246 (2010).

    Article  CAS  PubMed  Google Scholar 

  216. Wollenhaupt, M., Chandrasekaran, A. & Tomianovic, D. The safety of oseltamivir in pregnancy: an updated review of post-marketing data. Pharmacoepidemiol. Drug Saf. http://dx.doi.org/10.1002/pds.3673.

  217. Beau, A. B. et al. Safety of oseltamivir during pregnancy: a comparative study using the EFEMERIS database. BJOG 121, 895–900 (2014).

    Article  CAS  PubMed  Google Scholar 

  218. Jamieson, D. J. et al. H1N1 2009 influenza virus infection during pregnancy in the USA. Lancet 374, 451–458 (2009).

    Article  PubMed  Google Scholar 

  219. Treatment of influenza during pregnancy. US Food and Drug Administration [online], (2009).

  220. Parker-Athill, E. et al. Flavonoids, a prenatal prophylaxis via targeting JAK2/STAT3 signaling to oppose IL-6/MIA associated autism. J. Neuroimmunol. 217, 20–27 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Chicha, L., Smith, T. & Guzman, R. Stem cells for brain repair in neonatal hypoxia–ischemia. Childs Nerv. Syst. 30, 37–46 (2014).

    Article  CAS  PubMed  Google Scholar 

  222. Miller, B. J., Buckley, P., Seabolt, W., Mellor, A. & Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol. Psychiatry 70, 663–671 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Aberg, K. A. et al. Methylome-wide association study of schizophrenia: identifying blood biomarker signatures of environmental insults. JAMA. Psychiatry 71, 255–264 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Llano, D. A., Devanarayan, V. & Simon, A. J. Evaluation of plasma proteomic data for Alzheimer disease state classification and for the prediction of progression from mild cognitive impairment to Alzheimer disease. Alzheimer Dis. Assoc. Disord. 27, 233–243 (2013).

    Article  CAS  PubMed  Google Scholar 

  225. Kim, S. et al. Influence of genetic variation on plasma protein levels in older adults using a multi-analyte panel. PLoS ONE 8, e70269 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Busse, S. et al. Different distribution patterns of lymphocytes and microglia in the hippocampus of patients with residual versus paranoid schizophrenia: further evidence for disease course-related immune alterations? Brain Behav. Immun. 26, 1273–1279 (2012).

    Article  CAS  PubMed  Google Scholar 

  227. Doorduin, J. et al. Neuroinflammation in schizophrenia-related psychosis: a PET study. J. Nucl. Med. 50, 1801–1807 (2009).

    Article  PubMed  Google Scholar 

  228. van Berckel, B. N. et al. Microglia activation in recent-onset schizophrenia: a quantitative (R)-[11C]PK11195 positron emission tomography study. Biol. Psychiatry 64, 820–822 (2008).

    Article  PubMed  Google Scholar 

  229. Holmes, S. E. et al. Evidence for neuroinflammation in major depressive disorder and in schizophrenia—a positron emission tomography (PET) study using 11C-(R)-PK11195. J. Psychopharm. 28 (Suppl.), A47 (2014).

    Google Scholar 

  230. Takano, A. et al. Peripheral benzodiazepine receptors in patients with chronic schizophrenia: a PET study with [11C]DAA1106. Int. J. Neuropsychopharmacol. 13, 943–950 (2010).

    Article  CAS  PubMed  Google Scholar 

  231. Sommer, I. E. et al. Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophr. Bull. 40, 181–191 (2014).

    Article  PubMed  Google Scholar 

  232. Zavitsanou, K. et al. Effect of maternal immune activation on the kynurenine pathway in preadolescent rat offspring and on MK801-induced hyperlocomotion in adulthood: amelioration by COX-2 inhibition. Brain Behav. Immun. http://dx.doi.org/10.1016/j.bbi.2014.05.011.

  233. Mattei, D. et al. Minocycline rescues decrease in neurogenesis, increase in microglia cytokines and deficits in sensorimotor gating in an animal model of schizophrenia. Brain Behav. Immun. http://dx.doi.org/10.1016/j.bbi.2014.01.019.

  234. Ozkara, C. & Vigevano, F. Immuno- and antiinflammatory therapies in epileptic disorders. Epilepsia 52 (Suppl. 3), 45–51 (2011).

    Article  PubMed  Google Scholar 

  235. Vlad, S. C., Miller, D. R., Kowall, N. W. & Felson, D. T. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology 70, 1672–1677 (2008).

    Article  CAS  PubMed  Google Scholar 

  236. Chen, H. et al. Nonsteroidal antiinflammatory drug use and the risk for Parkinson's disease. Ann. Neurol. 58, 963–967 (2005).

    Article  CAS  PubMed  Google Scholar 

  237. McGeer, P. L., Schulzer, M. & McGeer, E. G. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer's disease: a review of 17 epidemiologic studies. Neurology 47, 425–432 (1996).

    Article  CAS  PubMed  Google Scholar 

  238. Hoozemans, J. J. & O'Banion, M. K. The role of COX-1 and COX-2 in Alzheimer's disease pathology and the therapeutic potentials of non-steroidal anti-inflammatory drugs. Curr. Drug Targets CNS Neurol. Disord. 4, 307–315 (2005).

    Article  CAS  PubMed  Google Scholar 

  239. US National Library of Medicine. ClinicalTrials.gov [online], (2014).

  240. Butovsky, O. et al. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat. Neurosci. 17, 131–143 (2014).

    Article  CAS  PubMed  Google Scholar 

  241. Figuera-Losada, M., Rojas, C. & Slusher, B. S. Inhibition of microglia activation as a phenotypic assay in early drug discovery. J. Biomol. Screen. 19, 17–31 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Tabas, I. & Glass, C. K. Anti-inflammatory therapy in chronic disease: challenges and opportunities. Science 339, 166–172 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Andersen, S. L. Trajectories of brain development: point of vulnerability or window of opportunity? Neurosci. Biobehav. Rev. 27, 3–18 (2003).

    Article  PubMed  Google Scholar 

  244. Bhugra, D. The global prevalence of schizophrenia. PLoS Med. 2, e151 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  245. Tandon, R. et al. Definition and description of schizophrenia in the DSM-5. Schizophr. Res. 150, 3–10 (2013).

    Article  PubMed  Google Scholar 

  246. Lauritsen, M. B. Autism spectrum disorders. Eur. Child. Adolesc. Psychiatry 22 (Suppl. 1), S37–S42 (2013).

    Article  PubMed  Google Scholar 

  247. Ronald, A. & Hoekstra, R. A. Autism spectrum disorders and autistic traits: a decade of new twin studies. Am. J. Med. Genet. B Neuropsychiatr. Genet. 156B, 255–274 (2011).

    Article  PubMed  Google Scholar 

  248. Righini, A. et al. Diffusion tensor imaging of early changes in corpus callosum after acute cerebral hemisphere lesions in newborns. Neuroradiology 52, 1025–1035 (2010).

    Article  PubMed  Google Scholar 

  249. Hauser, W. A. The prevalence and incidence of convulsive disorders in children. Epilepsia 35 (Suppl. 2), S1–S6 (1994).

    Article  PubMed  Google Scholar 

  250. Weinstein, S. E. & Gaillard, W. D. in Children with Disabilities 6th edn (eds Batshaw, M. L. et al.) 439–460 (Brookes Publishing Co., 2007).

    Google Scholar 

  251. Kurtz, Z., Tookey, P. & Ross, E. Epilepsy in young people: 23 year follow up of the British national child development study. BMJ 316, 339–342 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Theoharides, T. C. & Zhang, B. Neuro-inflammation, blood-brain barrier, seizures and autism. J. Neuroinflammation 8, 168 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  253. Hankins, G. D. & Speer, M. Defining the pathogenesis and pathophysiology of neonatal encephalopathy and cerebral palsy. Obstet. Gynecol. 102, 628–636 (2003).

    PubMed  Google Scholar 

  254. Nelson, K. B. The epidemiology of cerebral palsy in term infants. Ment. Retard. Dev. Disabil. Res. Rev. 8, 146–150 (2002).

    Article  PubMed  Google Scholar 

  255. Leviton, A. et al. The relationship between early concentrations of 25 blood proteins and cerebral white matter injury in preterm newborns: the ELGAN study. J. Pediatr. 158, 897–903 (2011).

    Article  CAS  PubMed  Google Scholar 

  256. Mann, J. R., McDermott, S., Bao, H. & Bersabe, A. Maternal genitourinary infection and risk of cerebral palsy. Dev. Med. Child Neurol. 51, 282–288 (2009).

    Article  PubMed  Google Scholar 

  257. Patrick, L. A. & Smith, G. N. Proinflammatory cytokines: a link between chorioamnionitis and fetal brain injury. J. Obstet. Gynaecol. Can. 24, 705–709 (2002).

    Article  PubMed  Google Scholar 

  258. Nagae, L. M. et al. Diffusion tensor imaging in children with periventricular leukomalacia: variability of injuries to white matter tracts. AJNR Am. J. Neuroradiol. 28, 1213–1222 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Ferri, C. P. et al. Global prevalence of dementia: a Delphi consensus study. Lancet 366, 2112–2117 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  260. Castellani, R. J., Rolston, R. K. & Smith, M. A. Alzheimer disease. Dis. Mon. 56, 484–546 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  261. Serrano-Pozo, A., Frosch, M. P., Masliah, E. & Hyman, B. T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 1, a006189 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Berg, D. et al. Changing the research criteria for the diagnosis of Parkinson's disease: obstacles and opportunities. Lancet Neurol. 12, 514–524 (2013).

    Article  PubMed  Google Scholar 

  263. Dickson, D. W. et al. Neuropathological assessment of Parkinson's disease: refining the diagnostic criteria. Lancet Neurol. 8, 1150–1157 (2009).

    Article  CAS  PubMed  Google Scholar 

  264. Goedert, M., Spillantini, M. G., Del, T. K. & Braak, H. 100 years of Lewy pathology. Nat. Rev. Neurol. 9, 13–24 (2013).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Support for third-party editorial assistance for this manuscript was provided by Roche. The authors would like to thank Drs Anirvan Ghosh, Omar Khwaja, Christoph Meier, Joseph Wettstein and Urs Meyer for inspiring discussions as well as the late Dr Paul H. Patterson whose work has been a starting point and continuous inspiration for the current Review. We also thank Ruth Habermacher Britschgi for her support in conceptualizing Figure 2. J.A.H. receives funding from the Autism Speaks Autism Treatment Network and the National Institutes of Mental Health, USA.

Author information

Authors and Affiliations

Authors

Contributions

I.K. and L.C. (equal contribution), together with M. Britschgi, S.A.S. and E.P.P., did the primary searches of the literature and wrote the article. M. Bodmer, J.A.H. and S.T. also contributed to researching data and assisted with writing the article. All authors made substantial contributions to the discussion of content, and helped to revise and/or edit the manuscript before submission.

Corresponding author

Correspondence to Eric P. Prinssen.

Ethics declarations

Competing interests

I.K., M. Britschgi, S.A.S. and E.P.P are full-time employees at Roche. J.A.H. is an investigator for studies funded by the Autism Speaks Autism Treatment Network, Forest Laboratories, Shire, Sunovion, and the National Institues of Mental Health, USA, and has been an advisor to Abbott Laboratories and a consultant to Ferring Pharmaceuticals. S.T. is a consultant paid by Roche. The other authors declare no competing interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Knuesel, I., Chicha, L., Britschgi, M. et al. Maternal immune activation and abnormal brain development across CNS disorders. Nat Rev Neurol 10, 643–660 (2014). https://doi.org/10.1038/nrneurol.2014.187

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneurol.2014.187

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing