Elsevier

Neuropharmacology

Volume 62, Issue 3, March 2012, Pages 1290-1298
Neuropharmacology

The viral theory of schizophrenia revisited: Abnormal placental gene expression and structural changes with lack of evidence for H1N1 viral presence in placentae of infected mice or brains of exposed offspring

https://doi.org/10.1016/j.neuropharm.2011.01.011Get rights and content

Abstract

Researchers have long noted an excess of patients with schizophrenia were born during the months of January and March. This winter birth effect has been hypothesized to result either from various causes such as vitamin D deficiency (McGrath, 1999, McGrath et al., 2010), or from maternal infection during pregnancy. Infection with a number of viruses during pregnancy including influenza, and rubella are known to increase the risk of schizophrenia in the offspring (Brown, 2006). Animal models using influenza virus or Poly I:C, a viral mimic, have been able to replicate many of the brain morphological, genetic, and behavioral deficits of schizophrenia (Meyer et al., 2006, Meyer et al., 2008a, Meyer et al., 2009, Bitanihirwe et al., 2010, Meyer and Feldon, 2010, Short et al., 2010). Using a murine model of prenatal viral infection, our laboratory has shown that viral infection on embryonic days 9, 16, and 18 leads to abnormal expression of brain genes and brain structural abnormalities in the exposed offspring (Fatemi et al., 2005, Fatemi et al., 2008a, Fatemi et al., 2008b, Fatemi et al., 2009a, Fatemi et al., 2009b). The purpose of the current study was to examine gene expression and morphological changes in the placenta, hippocampus, and prefrontal cortex as a result of viral infection on embryonic day 7 of pregnancy. Pregnant mice were either infected with influenza virus [A/WSN/33 strain (H1N1)] or sham-infected with vehicle solution. At E16, placentas were harvested and prepared for either microarray analysis or for light microscopy. We observed significant, upregulation of 77 genes and significant downregulation of 93 genes in placentas. In brains of exposed offspring following E7 infection, there were changes in gene expression in prefrontal cortex (6 upregulated and 24 downregulated at P0; 5 upregulated and 14 downregulated at P56) and hippocampus (4 upregulated and 6 downregulated at P0; 6 upregulated and 13 downregulated at P56). QRT-PCR verified the direction and magnitude of change for a number of genes associated with hypoxia, inflammation, schizophrenia, and autism. Placentas from infected mice showed a number of morphological abnormalities including presence of thrombi and increased presence of immune cells. Additionally, we searched for presence of H1N1 viral-specific genes for M1/M2, NA, and NS1 in placentas of infected mice and brains of exposed offspring and found none. Our results demonstrate that prenatal viral infection disrupts structure and gene expression of the placenta, hippocampus, and prefrontal cortex potentially explaining deleterious effects in the exposed offspring without evidence for presence of viral RNAs in the target tissues.

This article is part of a Special Issue entitled ‘Schizophrenia’.

Highlights

► Infection at E7 altered gene expression in the placentae of infected dams. ► Infection at E7 altered gene expression in PFC and hippocampus of exposed offspring. ► mRNA for viral genes was not found in placentae, PFC, or hippocampus. ► Infection at E7 caused structural changes in the placenta. ► Infection at E7 had little effect on brain structure of exposed offspring.

Introduction

Increased rates of schizophrenia have been associated with higher latitude and colder climate (Kinney et al., 2009). Different hypotheses have been proposed for this increase including vitamin D deficiency (McGrath, 1999, McGrath et al., 2010) and prenatal viral infection (Lewis, 2001, Fatemi, 2005, Brown, 2006). First proposed by McGrath (1999), prenatal vitamin D deficiency as a risk factor for schizophrenia could potentially explain several epidemiological findings including season of birth, latitude, differences in rates of schizophrenia between urban vs. rural populations, and the increased risk in 2nd generation migrants to northern countries (McGrath, 1999, Kinney et al., 2009, McGrath et al., 2010). Animal models of developmental vitamin D (DVD) deficiency have shown changes in brain development (Eyles et al., 2009), synaptic plasticity (Grecksch et al., 2009), and behavior (Grecksch et al., 2009, Fernandes de Abreu et al., 2010, Kesby et al., 2010) consistent with schizophrenia.

Over the past thirty years, numerous reports have established that viral infections during pregnancy increase the risk for the development of schizophrenia in the offspring (Lewis, 2001, Fatemi, 2005, Brown, 2006). An excess of schizophrenic patients are born during late winter and spring indicating the potential of influenza infections for these cases. Indeed, studies suggest that 5–15% excess schizophrenic births in the Northern Hemisphere occur during the months of January and March (Hare et al., 1972; Machon et al., 1983, Susser et al., 1999, Boyd et al., 1986, Pallast et al., 1994). Currently, there is debate whether direct infection of the offspring leads to changes ultimately resulting in schizophrenia or whether production of maternal cytokines in response to infection is responsible (Aronsson et al., 2001, Aronsson et al., 2002, Shi et al., 2009).

The current prevailing opinion is in favor of maternal cytokines as the causative agent of pathology in the developing offspring. A series of studies in different animal models for schizophrenia (Carpenter and Koenig, 2008) shows that maternal infection with human influenza or poly I:C can cause abnormalities in behavioral indices such as PPI and latent inhibition (LI) (Shi et al., 2003, Smith et al., 2007, Zuckerman and Weiner, 2003), both of which are similarly disrupted in subjects with schizophrenia. Recently, it has been demonstrated that injection of pregnant mice with IL-6, but not IFNγ, at E12.5 resulted in deficits in PPI and LI in the adult offspring but not juveniles (Smith et al., 2007), similar to what is observed in schizophrenia. Smith et al. (2007) also found that poly I:C-induced behavioral deficits in PPI, LI, and open field tests could be reversed if the poly I:C was co-administered with anti-IL-6 antibodies (Smith et al., 2007). Using IL-6 knockout mice, injection of poly I:C had no effect on PPI, open field test, and social interaction test (Smith et al., 2007). Inhibition of IL-6-mediated JAK2/STAT3 signaling using luteolin and its structural analog diosmin, has been shown to reduce behavioral deficits in social interaction in the offspring of mice born to dams injected with IL-6 on E12.5 of pregnancy (Parker-Athill et al., 2009). Interestingly, overexpression of anti-inflammatory cytokine IL-10 modulates poly I:C-induced behaviors such as PPI and LI (Meyer et al., 2008b). Taken together, these results suggest that pro-inflammatory cytokines, in particular IL-6, are key to mediating the effects of poly I:C infection on behavioral deficits in rodents that mimic similar deficits in schizophrenia and autism (Smith et al., 2007).

There is also evidence that the virus itself crosses the placenta to directly infect the fetus. Using a mouse animal model Aronsson et al., 2001, Aronsson et al., 2002 have shown that viral RNA for influenza A/WSN/33 strain (H1N1) persisted in the brains of offspring of virally infected dams. Recently a human postmortem study (Gu et al., 2007) demonstrated that the avian influenza virus (H5N1) can be transmitted across the placenta from mother to fetus. Taken together, these data show that both strains, at the expected doses contracted, can infect and be transmitted transplacentally to the fetus. These studies further provide clear evidence that one does not need viremia to induce placental passage of the virus.

An important aspect neglected in this debate is that the site of pathology may be the placenta. Whether the virus, traveling from the lungs, crosses the placenta; or whether in response to infection, maternal cytokines are produced which cross the placenta remains unknown. In the current study we examined the placenta as a site of pathology following prenatal viral infection on E7, in addition to prefrontal cortex and hippocampus in the exposed offspring. We hypothesized that viral infection would lead to gene expression and anatomical abnormalities in the exposed mice.

Section snippets

Viral infection

All experimental protocols used in this study were approved by the Institute for Animal Care and Use and Institutional Biosafety Committees at the University of Minnesota. A sublethal dose of influenza A/NWS/33 (H1N1) (diluted to 10−4.5) was administered intranasally (i.n.) on the seventh day of pregnancy (E7) to pregnant C57BL6J mice (N = 41) (Charles River, Wilmington, MA) as previously described (Fatemi et al., 2008a, Fatemi et al., 2008b, Fatemi et al., 2009a, Fatemi et al., 2009b). A

Gene expression changes in placenta following infection at E7

Microarray analysis of placental tissue found a significant (fold change of at least 1.5, p < 0.05) upregulation of 77 genes and downregulation of 93 genes (Appendix A). Among the important downregulated genes were: Glutamate receptor, ionotropic, AMPA1 (alpha 1) (Gria1), catenin (cadherin associated protein), delta 1 (Ctnn1), forkhead box O3 (fox03), and dysferlin (Dysf) (Table 1). Important upregulated genes included phosphodiesterase 10A (Pde10a), fyn proto-oncogene (Fyn), programmed cell

Discussion

In the current study we found that prenatal viral infection at E7 (approximately middle first trimester) resulted in a number of changes to the placentae of infected dams and brains of exposed offspring. We identified gene expression changes in the placentae of infected dams and in the hippocampi and frontal cortices of exposed offspring. Importantly, we did not detect mRNA for three H1N1 influenza viral genes in any of the tissues examined. Placentae of infected dams displayed a number of

Conclusions

Prenatal viral infection at E7 led to gene expression and morphological changes in placenta at E16. Many of the altered placental genes were associated with hypoxia, inflammation, immune system response, and apoptosis, suggesting that immune challenge had deleterious effects on the placental environment. Moreover, infection at E7 led to gene expression changes in the hippocampus and PFC of exposed offspring at birth and these changes persisted into young adulthood. Infection at E7 had minimal

Disclosure/conflict of interest

None of the authors have any conflict of interest with the information published in this article.

Acknowledgments

Grant support by the National Institute of Child Health and Human Development (#5R01HD046589-04 and #5R01HD046589-04S1) to SHF is gratefully acknowledged. Grant support from NIH (#R01EB003543) to SM is gratefully acknowledged. Technical assistance from DF Smee is gratefully acknowledged.

References (87)

  • M. Gannagé et al.

    Matrix protein 2 of influenza A virus blocks autophagosome fusion with lysosomes

    Cell Host Microbe

    (2009)
  • G. Grecksch et al.

    Transient prenatal vitamin D deficiency is associated with changes of synaptic plasticity in the dentate gyrus of adult rats

    Psychoneuroendocrinology

    (2009)
  • J. Gu et al.

    H5N1 infection of the respiratory tract and beyond: a molecular pathology study

    Lancet

    (2007)
  • K.J. Livak et al.

    Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method

    Methods

    (2001)
  • C. Ma et al.

    Differential protein expression in mouse splenic mononuclear cells treated with polysaccharides from spores of Ganoderma lucidum

    Phytomedicine

    (2008)
  • J. McGrath

    Hypothesis: is low prenatal vitamin D a risk-modifying factor for schizophrenia?

    Schizophr. Res.

    (1999)
  • U. Meyer et al.

    Adult brain and behavioral pathological markers of prenatal immune challenge during early/middle and late fetal development in mice

    Brain Behav. Immun.

    (2008)
  • U. Meyer et al.

    In-vivo rodent models for the experimental investigation of prenatal immune activation effects in neurodevelopmental brain disorders

    Neurosci. Biobehav. Rev.

    (2009)
  • U. Meyer et al.

    Epidemiology-driven neurodevelopmental animal models of schizophrenia

    Prog. Neurobiol.

    (2010)
  • J.A. O'Connor et al.

    Elevated Gria expression in Layer II/III and V pyramidal cells in DLPFC in schizophrenia

    Schizophr. Res.

    (2007)
  • T. Ohnuma et al.

    Expression of Fyn, a non-receptor tyrosine kinase in prefrontal cortex from patients with schizophrenia and its correlation with clinical onset

    Brain Res. Mol. Brain Res.

    (2003)
  • K. Okuda et al.

    Protective immunity against influenza A virus induced by immunization with DNA plasmid containing influenza M gene

    Vaccine

    (2001)
  • E. Parker-Athill et al.

    Flavonoids, a prenatal prophalaxis targeting JAK2/STAT3 signaling to oppose IL-6/MIA associated autism

    J. Neuroimmunol.

    (2009)
  • A.E. Rehn et al.

    An animal model of chronic placental insufficiency: relevance to neurodevelopmental disorders including schizophrenia

    Neuroscience

    (2004)
  • B. Schaubach et al.

    Regulation of murine ADA gene expression in the placenta by transcription factor RUNX1

    Placenta

    (2006)
  • L. Shi et al.

    Maternal influenza infection is likely to alter fetal brain development indirectly: the virus is not detected in fetus

    Int. J. Dev. Neurosci.

    (2005)
  • L. Shi et al.

    Activation of maternal immune system alters cerebellar development in the offspring

    Brain Behav. Immun.

    (2009)
  • S.J. Short et al.

    Maternal influenza infection during pregnancy impacts postnatal brain development in the rhesus monkey

    Biol. Psychiatry

    (2010)
  • A. Anitha et al.

    Genetic analyses of roundabout (ROBO) axon guidance receptors in autism

    Am. J. Med. Genet. B Neuropsychiatr. Genet.

    (2008)
  • F. Aronsson et al.

    Persistence of the influenza A/WSN/33 virus RNA at midbrain levels of immunodefective mice

    J. Neurovirol.

    (2001)
  • F. Aronsson et al.

    Persistence of viral RNA in the brain of offspring to mice infected with influenza A/WSN/33 during pregnancy

    J. Neurovirol.

    (2002)
  • M. Bacher et al.

    An essential regulatory role for macrophage migration inhibitory factor in T-cell activation

    Proc. Natl. Acad. Sci. USA

    (1996)
  • F. Benigni et al.

    The proinflammatory mediator macrophage migration inhibitory factor induces glucose catabolism in muscle

    J. Clin. Invest.

    (2000)
  • B.K. Bitanihirwe et al.

    Late prenatal immune activation in mice leads to behavioral and neurochemical abnormalities relevant to the negative symptoms of schizophrenia

    Neuropsychopharmacology

    (2010)
  • J.H. Boyd et al.

    Season of birth: schizophrenia and bipolar disorder

    Schizophr. Bull.

    (1986)
  • A.S. Brown

    Prenatal infection as a risk factor for schizophrenia

    Schizophr. Bull.

    (2006)
  • R.C. Bunn et al.

    Early developmental changes in IGF-1, IGF-II, IGF binding protein-1 and IGF binding protein-3 concentration in the cerebrospinal fluid of children

    Pediatr. Res.

    (2005)
  • T. Calandra et al.

    The macrophage is an important and previously unrecognized source of macrophage-migration inhibitory factor

    J. Exp. Med.

    (1994)
  • W.T. Carpenter et al.

    The evolution of drug development in schizophrenia: past issues and future opportunities

    Neuropsychopharmacology

    (2008)
  • C.J. Carter

    Schizophrenia susceptibility genes directly implicated in the life cycles of pathogens: cytomegalovirus, influenza, herpes simplex, rubella, and Toxoplasma gondii

    Schizophr. Bull.

    (2009)
  • M.J. Chen et al.

    Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter

    Nature

    (2009)
  • S.H. Fatemi

    Prenatal viral infection, brain development and schizophrenia

  • S.H. Fatemi et al.

    Defective corticogenesis and reduction in Reelin immunoreactivity in cortex and hippocampus of prenatally infected neonatal mice

    Mol. Psychiatry

    (1999)
  • Cited by (58)

    • Biological hypotheses, risk factors, and biomarkers of schizophrenia

      2023, Progress in Neuro-Psychopharmacology and Biological Psychiatry
      Citation Excerpt :

      However, evidence for maternal influenza during pregnancy as a risk factor for schizophrenia is insufficient (Selten and Termorshuizen, 2017). Epidemiological data indicate that environmental risk factors for schizophrenia include several prenatal and perinatal complications (Costas-Carrera et al., 2020; Davies et al., 2020), the use of cannabis (tetrahydrocannabinol can cause psychosis and schizophrenia in at-risk populations) (Patel et al., 2020), childhood trauma (Setien-Suero et al., 2020), social stressors (Susser and Patel, 2014), malnutrition leading to maternal vitamin D deficiency (Cui et al., 2021; Lisi et al., 2020) or to low folate and high homocysteine (Picker and Coyle, 2005), infection with influenza virus (Fatemi et al., 2012) and human endogenous retroviruses (Balestrieri et al., 2019), lower premorbid intelligence quotient (Schulz et al., 2014), and drug abuse (Ham et al., 2017). Only a history of obstetric complications, stressful events, childhood adverse events, cannabis use, and serum folate levels have provided reliable evidence of an association with schizophrenia (Belbasis et al., 2018).

    • Maternal immune activation in rodent models: A systematic review of neurodevelopmental changes in gene expression and epigenetic modulation in the offspring brain

      2021, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      Of note, while having no other incidences of inter-study analysis overlaps, when expression of Nos1 or Nos2 has been identified as significantly altered in mIA-affected offspring, there has been a unilateral increase relative to control offspring (Table 3). By comparison, for the neuro-patterning protein and nuclear receptor groups, all replicated results were mostly non-significant: Fgf8 growth factor, had no change in expression in a rat-poly(I:C) model but did demonstrate a significant increase in expression in a mouse-poly(I:C) model 2d post-induction while Shh, a developmental morphogen, was non-significant in both (Meyer et al., 2008a; Ohkawara et al., 2015) and Nr2f1, a steroid hormone receptor, was replicated as having unaltered expression in the adult hippocampus (Fatemi et al., 2012; Tang et al., 2013). Results for synaptic proteins and interneuron markers were more variable, with the exception of Pvalb, encoding parvalbumin, a calcium binding protein enriched in a subset of GABAergic interneurons, which was unchanged in every tissue analysed, a result which was replicated in the adult cerebral cortex (Duchatel et al., 2019; Rahman et al., 2020).

    View all citing articles on Scopus
    View full text