Elsevier

Brain, Behavior, and Immunity

Volume 67, January 2018, Pages 218-229
Brain, Behavior, and Immunity

Full-length Article
The microbiota influences cell death and microglial colonization in the perinatal mouse brain

https://doi.org/10.1016/j.bbi.2017.08.027Get rights and content

Highlights

  • Effects of the absence of a microbiota were examined in the neonatal mouse brain.

  • Pro-inflammatory cytokine expression was decreased in germ-free mice.

  • Microbiota influenced cell death and microglial labeling a region specific manner.

  • These effects were seen on the day of birth but not a day before.

  • Our results highlight the importance of the microbiota on early brain development.

Abstract

The mammalian fetus develops in a largely sterile environment, and direct exposure to a complex microbiota does not occur until birth. We took advantage of this to examine the effect of the microbiota on brain development during the first few days of life. The expression of anti- and pro-inflammatory cytokines, developmental cell death, and microglial colonization in the brain were compared between newborn conventionally colonized mice and mice born in sterile, germ-free (GF) conditions. Expression of the pro-inflammatory cytokines interleukin 1β and tumor necrosis factor α was markedly suppressed in GF newborns. GF mice also had altered cell death, with some regions exhibiting higher rates (paraventricular nucleus of the hypothalamus and the CA1 oriens layer of the hippocampus) and other regions exhibiting no change or lower rates (arcuate nucleus of the hypothalamus) of cell death. Microglial labeling was elevated in GF mice, due to an increase in both microglial cell size and number. The changes in cytokine expression, cell death and microglial labeling were evident on the day of birth, but were absent on embryonic day 18.5, approximately one-half day prior to expected delivery. Taken together, our results suggest that direct exposure to the microbiota at birth influences key neurodevelopmental events and does so within hours. These findings may help to explain some of the behavioral and neurochemical alterations previously seen in adult GF mice.

Introduction

Birth is an inflammatory event. The onset of labor depends on a state of ‘sterile inflammation,’ marked by a surge of maternal inflammatory cytokines (Golightly et al., 2011, Kobayashi, 2012, Thomson et al., 1999) that may reach the fetus (Dahlgren et al., 2006, Zaretsky et al., 2004) and stimulate brain cytokine expression (Dammann and Leviton, 1997). At parturition the fetus transitions from the relatively sterile environment of the womb to one teeming with microorganisms. Neonatal blood leukocyte populations expand rapidly following birth, likely due to both the stress of birth and sudden antigenic stimulation from maternal and environmental microbes (Marchini et al., 2000, Steinborn et al., 1999, Yektaei-Karin et al., 2007). Whether and how this microbial colonization affects the perinatal brain remains to be determined. However, effects of the microbiota on adult brain physiology and behavior have been reported in a variety of species including humans (Bravo et al., 2011, Clarke et al., 2013, Messaoudi et al., 2011).

One approach to address the question of the effects of the microbiota on perinatal brain development is to examine neonates born into germ-free (GF) conditions, because any role of microbial exposure would be absent in these animals. Neuronal cell death is a major neurodevelopmental event occurring around the time of birth in mice. Roughly 50% of the neurons that are initially produced are eliminated by apoptosis (Oppenheim, 1991). This large-scale pruning of neurons occurs primarily during the first week of life (Ahern et al., 2013) and is crucial for sculpting neuronal circuits. Although the importance of neuronal cell death is widely recognized, some surprisingly basic questions remain, such as what initiates the cell death period, or what accounts for the large regional differences in the magnitude of cell death. We recently found that cell death peaks just after birth in most forebrain regions of C57BL/6 mice (Mosley et al., 2017), suggesting that birth triggers or amplifies cell death.

Microglia are the resident immune cells of the brain and have been causally linked to developmental neuronal cell death (Marín-Teva et al., 2011). Microglia respond to perturbations by initiating an immune response involving the release of pro-inflammatory cytokines such as interleukin (IL) -6, IL-1β and tumor necrosis factor α (TNF-α; Olson and Miller, 2004), and are also activated by peripherally produced cytokines that reach the brain (Chen et al., 2012, Dantzer et al., 2008, Dilger and Johnson, 2008, Godbout et al., 2005, Qin et al., 2007). Microglial number increases during the first few weeks of postnatal life (Crain et al., 2013, Dalmau et al., 2003, Sharaf et al., 2013), and perinatal microglia have a relatively activated morphology and gene expression profile (Christensen et al., 2014, Crain et al., 2013, Lai et al., 2013, Schwarz et al., 2012, Strahan et al., 2017). Microglia may actively promote developmental cell death in the hippocampus and cerebellum (Marín-Teva et al., 2004, Wakselman et al., 2008), but enhance neuronal survival in the cerebral cortex (Arnoux et al., 2014, Ueno et al., 2013).

Adult GF mice have increased microglial numbers and altered microglial morphology (Erny et al., 2015), but whether effects of the microbiota on microglia are present early in life is unknown. Here, we investigated whether the normal exposure to microorganisms that occurs at birth influences cytokine expression, cell death, or microglial colonization of the newborn brain. Compared to conventionally colonized (CC) mice, we found markedly reduced levels of pro-inflammatory cytokine expression in the brains of GF mice on the day of birth and three days later. This was associated with brain-region specific changes in developmental neuronal cell death, and increased microglial density in GF mice. None of these changes were seen in the brains of GF embryos 12 h prior to expected birth. Together, our results suggest that the microbiota plays an important, region-specific role in brain development, and does so within hours of birth.

Section snippets

Animals

Swiss Webster mice (GF and CC) were obtained from our breeding program at Georgia State University. GF mice were kept under sterile conditions in a Park Bioservices isolator, as previously described (Chassaing et al., 2015), and CC mice were housed under conventional conditions. Mice were maintained in a 12:12 light dark cycle with ad libitum access to food and water. All procedures were approved by the Institutional Animal Care and Use Committee at Georgia State University and followed the

The microbiota increases the expression of inflammatory cytokines in the neonatal brain

We examined the expression of anti- (IL-10) and pro-inflammatory (IL-1β, IL-6, and TNF-α) cytokines in the mid- and hind-brain on P0 and P3. While there was no effect of microbiota status on the expression of IL-10 (F1, 76 = 0.18, p = 0.67; Fig. 1A), expression of the pro-inflammatory cytokines was markedly suppressed in the neonatal GF brain: IL-1β was reduced by 87% (F1, 76 = 11.38, p < 0.002; Fig. 1B) and TNF-α by 90% compared to CC mice (F1, 76 = 11.06, p < 0.002; Fig. 1C). There was also an 83%

Discussion

We find that lack of a microbiota alters neonatal brain development. Previously, altered brain physiology and behavior have been reported in adult GF mice. Compared to CC mice, GF adults exhibit a behavioral phenotype including reduced anxiety (Diaz Heijtz et al., 2011, Neufeld et al., 2011), a hyper-responsive stress response (Clarke et al., 2013, Sudo et al., 2004), impaired social behavior (Desbonnet et al., 2014), and impaired memory consolidation (Gareau et al., 2011). The production of

Conclusions

Taken together, our study highlights the importance of microbial exposure for neonatal brain development. Recently, differential exposure to microbiota at birth has been associated with the development of psychological disorders, such as autism (Curran et al., 2015). Millions of years of evolution have shaped the symbiotic relationship between mammalian species and their microbiota, so it is not surprising that our microbial symbionts influence key developmental processes. Moreover, because

Acknowledgments

We thank Geert de Vries, Mary Holder, Carla Cisternas, Nicole Peters, and Laura Cortes for critical comments on earlier versions of this manuscript. We also thank Daniel Cox and Atit Patel for RT-PCR training and Lucie Etienne-Mesmin for technical assistance. Supported by the National Science Foundation IOS-1743673 and the National Institutes of Health R21-MH108345 (to N.G.F). B.C. is a recipient of the Research Fellowship award from the Crohn’s and Colitis Foundation of America (CCFA).

References (90)

  • R.W. Oppenheim

    Naturally occurring cell death during neural development

    Trends Neurosci.

    (1985)
  • N.M. Orsi et al.

    Murine serum cytokines throughout the estrous cycle, pregnancy and post partum period

    Anim. Reprod. Sci.

    (2006)
  • B.P. Wann et al.

    Apoptosis detected in the amygdala following myocardial infarction in the rat

    Biol. Psychiatry

    (2006)
  • D. Wrona

    Neural-immune interactions: an integrative view of the bidirectional relationship between the brain and immune systems

    J. Neuroimmunol.

    (2006)
  • D.G. Zuloaga et al.

    Dexamethasone induces apoptosis in the developing rat amygdala in an age-, region-, and sex-specific manner

    Neuroscience

    (2011)
  • T.H. Ahern et al.

    Cell death atlas of the postnatal mouse ventral forebrain and hypothalamus: effects of age and sex

    J. Comp. Neurol.

    (2013)
  • I. Arnoux et al.

    Paradoxical effects of minocycline in the developing mouse somatosensory cortex

    Glia

    (2014)
  • F. Bäckhed et al.

    The gut microbiota as an environmental factor that regulates fat storage

    Proc. Natl. Acad. Sci. U.S.A.

    (2004)
  • W.A. Banks et al.

    Entry of blood-borne cytokines into the central nervous system: effects on cognitive processes

    Neuroimmunomodulation

    (2002)
  • P. Bercik et al.

    The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice

    Gastroenterology

    (2011)
  • J.A. Bravo et al.

    Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve

    Proc. Natl. Acad. Sci. U.S.A.

    (2011)
  • K.M. Buller

    Neuroimmune stress responses: reciprocal connections between the hypothalamus and the brainstem

    Stress

    (2003)
  • B. Chassaing et al.

    Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome

    Nature

    (2015)
  • Z. Chen et al.

    Lipopolysaccharide-induced microglial activation and neuroprotection against experimental brain injury is independent of hematogenous TLR4

    J. Neurosci.

    (2012)
  • L.B. Christensen et al.

    Age-related differences in neuroinflammatory responses associated with a distinct profile of regulatory markers on neonatal microglia

    J. Neuroinflamm.

    (2014)
  • G. Clarke et al.

    The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner

    Mol. Psychiatry

    (2013)
  • M.C. Collado et al.

    Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid

    Sci. Rep.

    (2016)
  • J.C. Condon et al.

    Surfactant protein secreted by the maturing mouse fetal lung acts as a hormone that signals the initiation of parturition

    Proc. Natl. Acad. Sci. U.S.A.

    (2004)
  • J. Correale et al.

    The neuroprotective role of inflammation in nervous system injuries

    J. Neurol.

    (2004)
  • J.M. Crain et al.

    Microglia express distinct M1 and M2 phenotypic markers in the postnatal and adult central nervous system in male and female mice

    J. Neurosci. Res.

    (2013)
  • E.A. Curran et al.

    Research review: birth by caesarean section and development of autism spectrum disorder and attention-deficit/hyperactivity disorder: a systematic review and meta-analysis

    J. Child. Psychol. Psychiatry

    (2015)
  • J. Dahlgren et al.

    Interleukin-6 in the maternal circulation reaches the rat fetus in mid-gestation

    Pediatr. Res.

    (2006)
  • I. Dalmau et al.

    Dynamics of microglia in the developing rat brain

    J. Comp. Neurol.

    (2003)
  • O. Dammann et al.

    Maternal intrauterine infection, cytokines, and brain damage in the preterm newborn

    Pediatr. Res.

    (1997)
  • R. Dantzer et al.

    From inflammation to sickness and depression: when the immune system subjugates the brain

    Nat. Rev. Neurosci.

    (2008)
  • L. Desbonnet et al.

    Microbiota is essential for social development in the mouse

    Mol. Psychiatry

    (2014)
  • R. Diaz Heijtz et al.

    Normal gut microbiota modulates brain development and behavior

    Proc. Natl. Acad. Sci. U.S.A.

    (2011)
  • R.N. Dilger et al.

    Aging, microglial cell priming, and the discordant central inflammatory response to signals from the peripheral immune system

    Leukoc. Biol.

    (2008)
  • M.G. Dominguez-Bello et al.

    Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns

    Proc. Natl. Acad. Sci. U.S.A.

    (2010)
  • M.G. Dominguez-Bello et al.

    Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer

    Nat. Med.

    (2016)
  • D. Erny et al.

    Host microbiota constantly control maturation and function of microglia in the CNS

    Nat. Neurosci.

    (2015)
  • J.V. Faustino et al.

    Microglial cells contribute to endogenous brain defenses after acute neonatal focal stroke

    J. Neurosci.

    (2011)
  • C.K. Fleissner et al.

    Absence of intestinal microbiota does not protect mice from diet-induced obesity

    Br. J. Nutr.

    (2010)
  • M.G. Gareau et al.

    Bacterial infection causes stress-induced memory dysfunction in mice

    Gut

    (2011)
  • J.P. Godbout et al.

    Exaggerated neuroinflammation and sickness behavior in aged mice following activation of the peripheral innate immune system

    FASEB J.

    (2005)
  • Cited by (0)

    View full text