Download Long-term pathological consequences of prenatal infection: beyond

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Common cold wikipedia , lookup

Immune system wikipedia , lookup

Social immunity wikipedia , lookup

Infection wikipedia , lookup

Inflammation wikipedia , lookup

Hospital-acquired infection wikipedia , lookup

Chickenpox wikipedia , lookup

Hepatitis B wikipedia , lookup

Innate immune system wikipedia , lookup

Sociality and disease transmission wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Immunomics wikipedia , lookup

Infection control wikipedia , lookup

Neonatal infection wikipedia , lookup

Hygiene hypothesis wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Transcript
Am J Physiol Regul Integr Comp Physiol 309: R1–R12, 2015.
First published April 29, 2015; doi:10.1152/ajpregu.00087.2015.
Review
Long-term pathological consequences of prenatal infection: beyond brain
disorders
Marie A. Labouesse, Wolfgang Langhans, and Urs Meyer
Physiology and Behavior Laboratory, ETH Zurich, Switzerland
Submitted 6 March 2015; accepted in final form 21 April 2015
autism; cytokines; gut microbiota; maternal immune activation; metabolic syndrome; schizophrenia
PRENATAL EXPOSURE TO INFECTIOUS pathogens or inflammatory
stimuli is increasingly recognized to play an important etiological role in neuropsychiatric and neurological disorders with
neurodevelopmental components (15, 21, 93, 99, 100). Significant associations between prenatal infection during pregnancy
and increased disease risk in later life have been revealed for
various brain disorders, including schizophrenia (19), autism
(7, 117), bipolar disorder (22, 116), mental retardation (61),
and cerebral palsy (31, 50). Hence, prenatal exposure to immune challenges may be best viewed as a general vulnerability
factor for neurodevelopmental brain disorders rather than a
disease-specific risk factor (52, 100). In this sense, the adverse
effects induced by prenatal infection may reflect an early entry
into a deviant neurodevelopmental route, but the specificity of
subsequent disease or symptoms is likely to be influenced by
the genetic and environmental context in which the prenatal
infectious process occurs. This concept would, indeed, be
consistent with the emerging evidence suggesting that seemingly remote disorders, such as schizophrenia, autism, attention-deficit/hyperactivity disorder, and major depression share
considerable amounts of risk factors and brain dysfunctions
(23, 92, 138). The presence of shared genetic and environ-
Address for reprint requests and other correspondence: M. A. Labouesse,
Physiology and Behavior Lab., Schorenstrasse 16, 8603 Schwerzenbach,
Switzerland (e-mail: [email protected]).
http://www.ajpregu.org
mental risks among those illnesses has led to the proposal
that they might lie along a continuum of genetically and
environmentally induced neurodevelopmental causalities
(103, 113), wherein prenatal infection may be one of the
many factors that shape the eventual pathological outcomes.
While the importance of prenatal immunological adversities
has been widely acknowledged in the fields of developmental
neuropsychiatry and neurology, less attention has been paid to
the possibility that prenatal exposure to infection may also play
an etiological role beyond central nervous system (CNS) disorders. Hence, the possible long-term effects of prenatal infection on disorders that are not primarily associated with CNS
dysfunctions may be somewhat underestimated. This seems
surprising for various reasons. The prenatal period is not only
a highly sensitive period for early brain development (99), but
also for other biological systems that develop in utero, including the innate and adaptive immune systems (47, 70), the
cardiovascular system (46, 60), the renal system (55, 80),
adipose tissue (140), and skeletal bones and muscles (20, 68).
It is, in fact, rather unlikely that maternal infection during
pregnancy would specifically disrupt fetal brain development;
instead, it may more generally represent a developmental
stressor for the entire organism. The underlying assumption for
this hypothesis is that infection leads to the secretion and
circulation of various immune system-related factors in the
maternal host and fetal environment, which, in turn, are likely
0363-6119/15 Copyright © 2015 the American Physiological Society
R1
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
Labouesse MA, Langhans W, Meyer U. Long-term pathological consequences of prenatal infection: beyond brain disorders. Am J Physiol Regul
Integr Comp Physiol 309: R1–R12, 2015. First published April 29, 2015;
doi:10.1152/ajpregu.00087.2015.—Prenatal immunological adversities such as
maternal infection have been widely acknowledged to contribute to an increased
risk of neurodevelopmental brain disorders. In recent years, epidemiological and
experimental evidence has accumulated to suggest that prenatal exposure to
immune challenges can also negatively affect various physiological and metabolic
functions beyond those typically associated with primary defects in CNS development. These peripheral changes include excessive accumulation of adipose tissue
and increased body weight, impaired glycemic regulation and insulin resistance,
altered myeloid lineage development, increased gut permeability, hyperpurinergia,
and changes in microbiota composition. Experimental work in animal models
further suggests that at least some of these peripheral abnormalities could directly
contribute to CNS dysfunctions, so that normalization of peripheral pathologies
could lead to an amelioration of behavioral deficits. Hence, seemingly unrelated
central and peripheral effects of prenatal infection could represent interrelated
pathological entities that emerge in response to a common developmental stressor.
Targeting peripheral abnormalities may thus represent a valuable strategy to
improve the wide spectrum of behavioral abnormalities that can emerge in subjects
with prenatal infection histories.
Review
R2
PERIPHERAL EFFECTS OF PRENATAL INFECTION
Obesity and Metabolic Dysfunctions
Epidemiological investigations in humans and experimental
work in animals both emphasize a critical role of the early-life
environment in shaping postnatal metabolic functions. Stimulated by the seminal work of David Barker and colleagues, the
concept of “early-life priming of adult disease” is now widely
accepted for various adverse health outcomes (37). In the
context of obesity and metabolic disorders, this concept refers
to the phenomenon that exposure to a specific environmental
factor during early prenatal or perinatal periods can induce
lifelong changes in metabolic functions, thereby predisposing
the organism to excessive adiposity and associated pathophysiological conditions, including impaired glucose homeostasis,
insulin resistance, Type 2 diabetes, and cardiovascular disease
(81, 89).
One of the most noticeable early-life adversities precipitating such metabolic dysfunctions is maternal obesity during
pregnancy (54, 141). Indeed, a robust association between
prenatal maternal obesity and an enhanced risk for metabolic
disorder in the offspring has been established by a plethora of
human epidemiological studies and translational animal models (41, 77). Because obesity is accompanied by chronic
low-grade inflammation (105, 131), it has been suggested that
enhanced systemic and placental inflammation in obese mothers may represent one of the mediating factors underlying the
developmental disruption of metabolic functions in the offspring (121, 127). Only relatively recently, however, epidemiologists have begun to explore more directly whether discrete
maternal exposure to infectious or inflammatory stimuli is
similarly associated with increased risk for metabolic disorder
in the offspring. A first line of evidence supporting this hypothesis comes from a human epidemiological study that used
a cross-sectional cohort design, in which more than 17,000
male singletons were included (25). The findings from this
study suggested a 34% increased risk of obesity [defined on the
basis of a body mass index (BMI) of 30 kg/m2 or more] after
being born to a mother with infection during pregnancy (25).
Interestingly, a similar positive association has also been found
following childhood infections (25), suggesting that the developmental period for infection-induced changes in adiposity
extends to the early postnatal life.
Work in developmental rodent models further supports the
hypothesis of causal effects between maternal immune challenge during pregnancy and long-term metabolic dysfunctions
in the offspring. For example, it has been shown that maternal
treatment with the bacterial endotoxin LPS during midpregnancy in rats induces a wide spectrum of metabolic
disturbances in the adult offspring, including increases in
body weight and adipose tissue, hyperphagia, and insulin
resistance (111). These effects were sex-specific and
emerged in male but not female rats, indicating that male
offspring are more vulnerable than females in terms of
developing metabolic abnormalities following maternal endotoxemia during pregnancy. Findings from other rodent
models of maternal immune activation further suggest that
the association between prenatal immune challenge during
pregnancy and development of metabolic disturbances is not
dependent on the precise identity of the infectious or inflammatory pathogen. In fact, such metabolic effects can
even be induced by prenatal exposure to specific inflammatory cytokines (29). Moreover, long-term metabolic abnormalities have also been observed in mouse offspring of
mothers that were exposed to the viral mimetic polyriboinosinic-polyribocytidilic acid (polyI:C), a synthetic analog
of double-stranded RNA that induces a virus-like acute
phase response (93). In this virus-like immune activation
model, offspring of polyI:C-treated mouse dams were
shown to develop altered glycemic regulation and abnormal
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
to affect peripheral organs and brain structures in the developing organism to the same extent.
Another reason why we should not ignore CNS-remote
effects of prenatal infection relates to the clinical observations
that individuals with major psychiatric disorders often display
signs of physiological and metabolic dysfunctions, including
insulin resistance, Type 2 diabetes, obesity, and cardiovascular
disease (90). Even though chronic psychopharmacotherapy
may facilitate or even induce such abnormalities (40, 139), it is
becoming increasingly evident that they cannot solely be accounted for by chronic drug exposure. Indeed, certain metabolic and physiological dysfunctions often occur (albeit in a
somewhat less severe form) in drug-naïve or minimally medicated first-episode patients with mental illnesses (63, 124, 143,
153). These findings have been taken as circumstantial evidence that at least some of the metabolic and pathophysiological comorbidities in major brain disorders may have a developmental origin and, thus, emerge prior to the onset of fullblown psychiatric illness.
Finally, neurodevelopmentally acquired brain abnormalities
in response to maternal infection may also facilitate the development of metabolic and physiological dysfunctions across the
postnatal life span. For example, the development of homeostatic brain regions, such as the hypothalamus, encompasses a
number of steps that begin early in fetal life and continue
postnatally (135). Infection-induced maldevelopment of such
brain regions could thus impair the homeostatic control of
various physiological functions, including food intake, renal
functions, and reproduction (81, 89).
Here, we review the existing evidence indicating that the
long-term pathological consequences of prenatal immune
challenges are not solely confined to primary CNS disorders. Instead, they extend more globally to other physiological and metabolic dysfunctions in peripheral systems. Relevant studies were identified and selected using PubMed.
All articles published before December 2014 were screened
for relevance and were searched using the following search
criteria (in alphabetical order) in various combinations:
“animal model,” “autism,” “bipolar disorder,” “body
weight,” “cytokines,” “diabetes,” “dopamine,” “epidemiology,” “food intake,” “glucose,” “gut,” “homeostasis,” “hypothalamus,” “immune activation,” “infection,” “inflammation,” “LPS,” “maternal,” “metabolism,” “metabolic syndrome,” “microbiota,” “microglia,” “obesity,” “poly(I:C),”
“pregnancy,” “prenatal,” and “schizophrenia”. In addition to
reviewing the available literature, we also provide a conceptual framework suggesting that several apparently unrelated central and peripheral effects of prenatal infection may
not represent separate pathological entities but could, in
fact, result from interrelated pathological mechanisms that
are engaged in response to a common developmental stressor.
Review
PERIPHERAL EFFECTS OF PRENATAL INFECTION
Peripheral (and Central) Inflammation
Various rodent models demonstrate that maternal exposure
to infectious or immune system-activating agents leads to robust
post-acute immune changes at the maternal-fetal interface, including the placenta, amniotic fluid, and fetal organism (2, 6, 96, 97,
151, 154). The nature and/or severity of these changes are influenced by various factors, most notably the identity and/or intensity
of the pathogen (51, 94), the gestational timing of exposure (97),
and the genetic background of the infected host (2, 96, 154).
Despite this, it seems that maternal exposure to distinct infectious
or immune system-activating agents leads to partially overlapping
immune responses in the fetal system. These overlapping effects
are mostly characterized by increased fetal expression of inflammatory factors, such as proinflammatory cytokines and
chemokines (5). It is believed that abnormal expression of
inflammatory factors in the fetal brain contribute to, or even
mediate, abnormal brain and behavioral development following prenatal exposure to infection (79, 136). Indeed, as reviewed extensively elsewhere (95, 102), acute inflammation
during early fetal brain development may negatively affect
ongoing neurodevelopmental processes, such as neuronal/glial
cell differentiation, proliferation, migration, and survival, and,
thus, predispose the developing offspring to long-term brain
and behavioral dysfunctions.
Since signs of subchronic systemic (and central) inflammation are often present in at least a subset of patients with
neurodevelopmental disorders (64, 86, 92, 101, 150), considerable research has been undertaken to address the question of
whether maternal exposure to infectious or inflammatory
agents may lead to persistent inflammatory changes in the
offspring postnatally. The evidence for this hypothesis is, at
best, equivocal. Some studies using rodent models of bacterial
or viral maternal immune challenge have reported a significant
upregulation of circulating inflammatory factors such as proinflammatory cytokines or chemokines in the juvenile or adult
offspring (17, 43, 69). Other studies using the same animal
models, however, failed to find clear signs of systemic inflammation in juvenile or adult offspring born to immunologically
challenged mothers (96, 157), or they even reported opposite
effects that were characterized by blunted systemic inflammatory
activity following prenatal infection (115).
Inconclusive data also exist with respect to central inflammation. In the CNS, microglia and astrocytes are the major
immunocompetent cells, which drive both the induction and
limitation of inflammatory processes (122). This is achieved
through the synthesis of proinflammatory and anti-inflammatory cytokines, upregulation, or downregulation of various cell
surface receptors, such as pathogen recognition receptors, cytokine receptors, and numerous receptors crucial for antigen
presentation. Enhanced microglia activation in brain parenchyma, along with increased central production of secreted
inflammatory factors, is often taken as a sign of ongoing
inflammation in the CNS (48). The possibility that prenatal
infection leads to chronic signs of brain inflammation has been
supported only by some studies in rats and mice (17, 69),
whereas other rodent studies failed to find evidence for such
neuroinflammatory processes extending into neonatal or adult
life (5, 43, 96, 119, 151, 157).
The inconsistency surrounding the long-term effects of prenatal infection on the persistence of inflammatory processes
across the postnatal life span may be explained by various
factors, most notably the severity and/or chronicity of the
infectious process targeting the maternal host. Indeed, it appears that more marked postnatal inflammatory changes are
seen following relatively severe forms of maternal immune
challenge, such as chronic exposure to immune system-activating agents throughout the entire gestational period (17). In
contrast, acute or subchronic prenatal exposure to immune
system-activating stimuli in mice and rats appears to be largely
devoid of systemic and central inflammatory effects persisting
into the juvenile or adult period (5, 43, 96, 119, 151, 157). The
latter may not seem surprising because inflammation is typically counteracted by homeostatic processes that mount antiinflammatory and/or immunosuppressive responses upon the
induction of inflammation (132), which, in turn, dampen and
finally resolve the post-acute fetal inflammatory responses to
maternal immune activation (91).
Still, the developing organism might sustain latent inflammatory abnormalities that may not become apparent until
reexposure to specific immunogens postnatally. In support of
this hypothesis, it has been shown that prenatal virus-like
immune activation in mice leads to a persistent decrease in the
number of regulatory T cells (Tregs) in the spleen and mesenteric lymph nodes (58). Besides other functions, Tregs can
potently suppress innate and adaptive immune responses, so
that a disruption of normal Tregs functions can lead to exacerbated inflammatory reactions (24). Such an exacerbation is
seen in mouse offspring born to immunologically challenged
mothers, which mount a potentiated proinflammatory T-cell
response to immunogenic T-cell stimulation (58).
Prenatal virus-like immune activation in mice has further
been shown to induce long-term changes in macrophage function that persist into adulthood. More specifically, bone marrow-derived macrophages of prenatally poly(I:C)-exposed
mice show an augmented proinflammatory cytokine response
to in vitro LPS stimulation alone, or in combination with
interferon (IFN)-␥ (112). The costimulation with LPS and IL-4
does not result in a similar proinflammatory cytokine signature
(112). Collectively, these results indicate that prenatal viruslike immune activation potentiates the polarization of macrophages toward an M1 phenotype, which, in turn, is typically
associated with larger production of proinflammatory cytokines, such as IL-12, at the expense of reduced production of
anti-inflammatory cytokines, such as IL-10 (85, 106). Thus, it
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
ingestive behavior in adolescence and excess fat deposition
in adulthood (115). Similar results were obtained in a mouse
model, in which pregnant mice were infected with Ljungan
virus (LV), a virus that belongs to the Picornavirus family
and is virulent for laboratory rodents (126). Maternal LV
exposure in mice has been shown to predispose the offspring
to signs of Type 2 diabetes and obesity (110). These effects,
however, were particularly manifest in LV-exposed mice
that experienced additional stress during adolescence, suggesting that the severity of metabolic abnormalities following prenatal immune activation can be exacerbated by exposure to other environmental adversities, such as adolescent stress (110). These findings are particularly relevant for
the multifactorial etiology of obesity and Type 2 diabetes,
which are likely caused by a combination of environmental
(and genetic) factors (38, 81).
R3
Review
R4
PERIPHERAL EFFECTS OF PRENATAL INFECTION
Gastrointestinal Abnormalities and Dysbiosis
In recent years, there has been an increasing interest in the
relative potential of gastrointestinal (GI) functions to modulate
neuropsychological traits implicated in psychiatric disorders
(28, 87, 88). Epidemiological and clinical studies have repeatedly demonstrated a high comorbidity between GI inflammatory disorders and stress-related psychiatric symptoms, such as
anxiety or depressive behavior (28, 36). These findings have
been complemented by experimental studies in rodents showing that abnormal development of the gut microbiota leads to
neuroendocrine, neurochemical, and emotional abnormalities,
some of which are reminiscent of hormonal and behavioral
aberrations typically seen in depression and/or anxiety disorders. For example, ablation of the commensal GI microbiota in
germ-free mice or by chronic antimicrobial compounds can
lead to functional changes in the hypothalamus-pituitary-adrenal (HPA) axis and in the central dopaminergic, serotonergic,
and GABAergic neurotransmitter systems, and these effects
are further linked to the emergence of increased anxiety-like
behavior (11, 35).
Other clinical populations, for which GI abnormalities and
dysbiosis seem clinically relevant, comprise individuals suffering from neurodevelopmental psychiatric illnesses. Indeed, in
addition to the pathological symptoms traditionally attributed
to CNS dysfunctions, neurodevelopmental psychiatric illnesses, such as autism and schizophrenia, are also associated
with a number of GI dysfunctions. Such abnormalities include
chronic intestinal low-grade inflammation, increased intestinal
permeability (“leaky gut”), allergic reactions to dietary proteins, diarrhea, gastric dysmobility, and alterations in gut
microbiota (16, 26, 133, 136). By further undermining general
physical health and daily life quality, GI distress induces an
additional clinical burden to patients suffering from psychiatric
disorders (9, 53).
In view of the etiological contribution of prenatal infection
to neurodevelopmental diseases, experimental research has
begun to explore whether early-life immune challenges may be
a relevant environmental risk factor for the development of GI
abnormalities. In a seminal recent study, Hsiao et al. (58)
demonstrated that prenatal polyI:C-induced immune activation
in mice can indeed cause long-term defects in intestinal integrity and alterations in the composition of the commensal
microbiota. The former effect was evident by increased translocation of dextran across the intestinal epithelium and was
associated with decreased colonic expression of tight junction
components. Interestingly, signs of intestinal disintegrity in
polyI:C-exposed mouse offspring were already present by
the age of 3 wk, suggesting that these abnormalities are
established during early life. Furthermore, the colons from
prenatally infected mice displayed increased expression of
the inflammatory cytokine IL-6, which is in line with the
notion that increased gut permeability is commonly associated with altered immune responses in the GI tract (148).
Offspring of immunologically exposed mouse dams also
markedly differed from control offspring in terms of their
gut microbiota composition, and this difference was mostly
accounted for by differences in the relative abundance of
Clostridia and Bacteroidia classes (58).
The study by Hsiao et al. (58), which used a mouse model of
virus-like immune activation, is, thus far, the only one that
directly assessed the influence of prenatal immune challenge
on the development of GI abnormalities in the offspring.
Hence, it remains essentially unknown whether similar effects
can also be induced by prenatal exposure to other infectious or
inflammatory agents. It should be noted, however, that remarkable changes in the gut microbiota composition and associated
intestinal abnormalities have also been identified in another
experimental model of neurodevelopmental disease, namely
prenatal exposure to valproic acid (VPA) in mice (34). Similar
to the polyI:C model (83), the VPA model is frequently used to
induce autism-related brain and behavioral abnormalities in
experimental rodents (123). Thus, it appears that prenatal
exposure to distinct environmental factors does not only induce
overlapping neurobehavioral phenotypes, but further induce
similar changes in GI functions and microbiota. It remains
currently unknown whether the latter commonalities may be
explained by mutual pathogenic mechanisms, whereby neurodevelopmentally acquired abnormalities could increase the
vulnerability to GI pathology through mechanisms involving a
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
seems that prenatal immune activation can prime the offspring’s peripheral immune system in such a way that it
mounts more excessive proinflammatory responses in the event
of postnatal pathogen (re-)exposures.
It should also be pointed out that prenatal immune activation can similarly lead to latent immune abnormalities in
the CNS. As reviewed in detail elsewhere (14, 50), numerous experimental studies in rodents show that immunological exposure in early (prenatal or neonatal) life can cause
the organism to mount differential (and often more vigorous) CNS inflammatory responses to subsequent immunological or nonimmunological challenges. For example, even
though prenatal virus-like immune activation per se does not
cause overt glial abnormalities and associated inflammatory
changes in the brain parenchyma of mouse offspring (5, 43,
157), signs of microglia overactivation and exacerbated
inflammatory cytokine secretion in CNS areas can be induced in these offspring when they are additionally exposed
to subchronic stress postnatally (44). This form of immune
sensitization or priming suggests that prenatal immune activation can markedly increase the vulnerability of the
offspring to brain immune changes in response to stress. A
similar microglia-priming effect by prenatal immune challenge has been demonstrated in a virus-like immune activation model, in which mouse offspring were subjected to
either prenatal poly(I:C) treatment alone, or in combination
with a second poly(I:C) treatment regimen in late adulthood
(69). In this model, mice exposed to both prenatal and
postnatal poly(I:C) treatment showed more extensive microglia activation and astrogliosis compared with either treatment alone (69).
Taken together, even though the long-term effects of prenatal immune activation on peripheral and central inflammatory
responses may not be apparent under basal conditions, they
may become so when the offspring are exposed to additional
environmental challenges, such as stress or acute infection
during the postnatal life. In addition, persistent inflammatory
abnormalities may develop more locally and may thus emerge
in specific peripheral organs. The latter possibility is discussed
in more detail in the subsequent section.
Review
PERIPHERAL EFFECTS OF PRENATAL INFECTION
R5
disruption of the normal top-down (CNS to GI tract) signaling.
This possibility is discussed in more detail below.
Do Peripheral Abnormalities Contribute to Neurobehavioral
Deficits Following Prenatal Infection?
Fig. 1. Graphical illustration of main top-down [central nervous system (CNS)
to periphery] and bottom-up (periphery to CNS) communication pathways and
summary of peripheral and central pathologies emerging following prenatal
infection. Top-down and bottom-up communication pathways are represented
by solid and dashed lines, respectively. CNS, central nervous system; CORT,
cortisol/corticosterone; NA, noradrenaline; Treg, regulatory T cells.
(58). On the basis of their subsequent findings showing that
prenatal virus-like immune challenge in mice leads to impaired GI
functions and dysbiosis, Hsiao et al. (59) went on to show that oral
treatment with the human commensal Bacteroides fragilis normalizes gut permeability and microbial composition and further
corrects autism-related behavioral deficits in mouse offspring born
to immunologically challenged mothers. Together, these findings
support the hypothesis that gut- and immune system-related peripheral abnormalities following prenatal infection have a direct
impact on CNS functions, so that normalization of the former
leads to beneficial effects on the latter.
A similar conclusion can be drawn from a recent investigation in mice demonstrating a link between prenatal virus-like
immune activation and the emergence of altered purine metabolism in plasma (108). Besides other metabolic changes, offspring of immunologically challenged mouse dams showed
marked signs of hyperpurinergia, a pathophysiological condition that is characterized by increased (plasma) levels of
purines. Importantly, Naviaux et al. (108) showed that an
antipurinergic therapy using acute administration of suramin
not only normalizes the peripheral metabolic abnormalities in
prenatally infected mice, but further corrects several of the
behavioral deficits typically observed in untreated offspring. In
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
Even though the evidence for altered peripheral functions in
prenatally infected subjects is emerging, it remains largely
elusive whether these abnormalities functionally contribute to
the manifestation of neurobehavioral deficits. In other words,
do peripheral and central abnormalities, which can both be
precipitated by early-life adversities such as prenatal infection,
represent separate pathological entities? Or is there a pathological connection between the two, so that functional abnormalities in the former can lead to pathological changes in the
latter?
It is obvious that the latter scenario would require bottom-up
(periphery to CNS) and top-down (CNS to periphery) pathways that allow a bidirectional communication between the
brain and the peripheral systems. As extensively reviewed
elsewhere (12, 27, 28, 88), such pathways, indeed, exist and are
evolutionarily well conserved across species. In fact, this
bidirectional communication between the CNS and periphery
is pivotal for body homeostasis and adaptive responses to
changes in environmental conditions. Various peripheral
signals such as GI peptides, cytokines, neuroactive bacterial
metabolites, and adipose-derived factors can be relayed to the
CNS via multiple routes, including the vagal afferents and the
blood circulation (12, 27, 28, 88). The latter allows transport of
systemic signals into the CNS through specialized transport
systems embedded in the blood-brain barrier (BBB), or at sites
lacking a complete BBB, such as the circumventricular organs.
Afferent vagal nerve fibers are the main neuronal pathways
through which peripheral signals can be conveyed to the brain
(Fig. 1). Vagal afferent neurons synapse bilaterally on the
nucleus tractus solitarii, from where neuronal signals are transmitted to other brain stem nuclei and to various forebrain
structures, such as the hypothalamus, nucleus accumbens,
amygdala, and prefrontal cortex (12). On the other hand, the
periphery is connected with and modulated by the CNS
through neuronal efferent branches of the sympathetic and
parasympathetic nervous systems, which regulate visceral
functions, both at rest and in response to activating stimuli
(134). The periphery is further regulated by factors secreted by
various neuroendocrine systems, such as the HPA axis, which
has a central role in regulating many homeostatic processes,
particularly in response to stress-related stimuli (10).
In view of these elaborated bidirectional communication
pathways, it may not be surprising that interventions directed at
peripheral targets have the potential to ameliorate neurobehavioral deficits in offspring exposed to prenatal infection. A proof
of concept for this notion has recently been realized using
prenatal virus-like immune activation models in mice. In a first
study, Hsiao et al. (58) demonstrated that autism-like behavioral
abnormalities, which typically emerge in prenatally polyI:Cinfected mouse offspring, can be rescued by transplantation of
bone marrow derived from control offspring. This elegant
rescue experiment was conducted on the basis of the aforementioned findings showing that prenatal virus-like immune
activation in mice impairs development and functions of Tregs,
and further alters the differentiation of the myeloid cell lineage
Review
R6
PERIPHERAL EFFECTS OF PRENATAL INFECTION
Do Neurodevelopmental Deficits Contribute to Peripheral
Abnormalities Following Prenatal Infection?
While a direct influence of peripheral abnormalities to CNS
dysfunctions is supported by at least some animal studies, it
remains essentially unknown whether the opposite scenario
holds true as well. Hence, the role of neurodevelopmentally
acquired CNS dysfunctions in promoting peripheral abnormalities following prenatal infection still awaits thorough examination. In view of the elaborated top-down (CNS to periphery)
communication pathways (Fig. 1), however, it seems feasible
that abnormal functioning of discrete neuronal and neuroendocrine systems can, at least to some extent, facilitate the development and/or maintenance of peripheral abnormalities.
Perhaps one of the most obvious examples in this context
relates to the crucial role of the hypothalamus and interconnected structures in controlling food intake and energy balance
(104, 129). Given that maternal immune activation alters hypothalamic functions in rodent offspring (42, 78, 158), neurodevelopmentally acquired abnormalities in the hypothalamus
may contribute to the emergence of a hyperphagic phenotype
in offspring with prenatal infection (111, 115). An alternative
(but not mutually exclusive) mechanism underlying such
changes in food intake may be related to changes in the
mesocorticolimbic dopamine system. A plethora of investigations support a key role of dopamine in reward and incentive
values on the one hand, and in the associations between reward
and eating behavior on the other hand. These functional associations are highly complex and likely involve intricate interactions among homeostatic, hedonic, motivational, and associative processes (62, 104, 125). Prenatal immune activation in
rats and mice is well known to induce primary defects in early
dopaminergic development (91, 155) and to lead to long-term
dopaminergic abnormalities in the adult offspring (4, 98, 114,
155, 160). It seems, therefore, plausible that such dopaminergic
dysfunctions may be involved in the development of altered
food intake and energy balance, be it because of changes in
homeostatic, hedonic, motivational, and/or associative processes.
Another possible association that warrants careful examination in future studies is the potential impact of altered autonomic nervous system (ANS) functions in prenatally infected
animals. Initial evidence suggests that prenatal immune activation can cause hyperactivity of the sympathetic nervous
system, perhaps at the expense of diminished parasympathetic
nervous system functions. Hyperactivity of the sympathetic
nervous system typically leads to increased secretion of noradrenaline and corticosterone, both of which have been observed in mouse offspring that were exposed to prenatal immune challenge (115, 158). Prolonged secretion of these stressrelated factors can negatively influence GI physiology,
including gut motility, secretion, permeability, and composition of the gut microbiota (13, 67). Given that some of these GI
abnormalities have been shown to develop in mice exposed to
prenatal immune challenge (59), it would appear highly warranted to explore whether they may be causally related to a
hyperactivity of the sympathetic nervous system.
At the same time, diminished activity of the parasympathetic
branch of the ANS may contribute to the development of
altered inflammatory responses in the GI tract and other peripheral organs (120). Indeed, efferent vagal pathways originating from the dorsal motor nucleus of the vagus possess
anti-inflammatory activity by releasing ACh, which, in turn,
inhibits proinflammatory cytokine secretion by peripheral immune cells upon binding to ␣7 nicotinic receptors (118, 145).
This parasympathetic mechanism of neuronal anti-inflammatory signaling has been termed “the cholinergic antiinflammatory pathway” and seems to play an important role
in dampening peripheral inflammatory responses (118, 145).
Whether this pathway is altered by prenatal exposure to
infection still awaits direct examination, but it could provide
a contributing factor for the prenatal infection-induced disturbances in local inflammatory responses, including intestinal inflammation (59).
Possible Mechanisms Mediating the Effects of Maternal
Infection on the Offspring
The precise mechanisms responsible for mediating the pathological effects of maternal infection on the developing organism in utero remain to be determined. In fact, several plausible
mechanisms exist, whereby maternal infection can negatively
affect the normal development of peripheral (and central)
organs. As summarized in Fig. 2, the different pathological
mechanisms induced by infection may not be mutually exclusive, but may rather interact with each other to affect various
developmental processes that take place in prenatal life.
One prevalent hypothesis suggests that common immunological factors, in general, and inflammatory cytokines, in
particular, are key mediating factors changing developmental
trajectories in the offspring (4, 45, 79, 96, 102, 137). Inflammatory cytokines are typically induced during the acute phase
response to infection (146, 147) and may represent a major
developmental stressor for the organism (18, 57, 95). In the
event of maternal infection, an increase in fetal cytokine levels
may be caused by transplacental transfer of maternally pro-
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
addition to immunological and gut-related factors (58, 59),
alterations in blood purine metabolism, thus, seem to be another peripheral pathology that readily contributes to the emergence of behavioral deficits following prenatal infection (108).
Thus far, it remains unknown whether other metabolic
disturbances that are induced by prenatal infection, including
alterations in glycemic regulation, insulin insensitivity, and
increased adiposity, may also directly influence behavioral
functions in subjects with a history of prenatal infection.
Hence, it remains to be determined whether normalization of
these metabolic abnormalities may exert beneficial effects on
behavioral deficits in this population. Furthermore, the precise
link between individual peripheral abnormalities also remains
elusive. For example, it is not known whether the excessive
accumulation of adipose tissue in offspring of infected mothers
may facilitate excessive proinflammatory signaling. Such a
link has been demonstrated in various models of obesity, but it
still awaits direct verification in models of prenatal immune
challenge. Given the emerging functional relationships between the gut microbiome and obesity and between the gut
microbiome and Type 2 diabetes (144), it would also seem
warranted to explore such possible connections in the context
of pathologies that are associated with prenatal exposure to
infection.
Review
PERIPHERAL EFFECTS OF PRENATAL INFECTION
duced cytokines (30, 159), by placental production of cytokines (6, 56, 128), or by increased fetal cytokine synthesis (97).
In addition to its effects on inflammatory cytokine secretion,
infection and the subsequent induction of inflammatory responses are also strongly associated with numerous other
pathophysiological effects, including oxidative stress. Oxidative stress is referred to as an imbalance between the production and elimination of reactive oxygen species (ROS), some of
which are highly cytotoxic and promote tissue injury (66).
Upon activation, innate immune cells secrete ROS and reactive
nitrogen species (RNS) as a central part of killing invading
pathogens (107). Production of ROS and RNS is, thus, an
important downstream mechanism of inflammation-mediated
immune responses. Several lines of evidence from rodent
models suggest that infection-induced increases in fetal ROS
and RNS levels may, indeed, be involved in changing development trajectories in the offspring (74, 75).
Besides its effects on oxidative stress systems, exposure to
infection and the subsequent cytokine-associated inflammatory
reactions further lead to the activation of the HPA axis by
stimulating the release of corticotropin-releasing factor from
the hypothalamus and of adrenocorticotropic hormone from the
pituitary gland; this results eventually in an increase of glucocorticoid levels in the peripheral bloodstream (49). The cytokine-mediated effects on glucocorticoid secretion may be of
special interest because it has been suggested that prenatal
physiological stress triggered by high glucocorticoid levels can
interfere with normal physiological and metabolic development (130, 156). Activation of the innate immune system (in
response to infection) also changes the maternal and fetal
availability of several micronutrients, including iron and zinc,
both of which are highly important for the normal development
of peripheral and central organs (3, 71, 72, 149). In the case of
iron, it is well established that infection leads to a temporary
depletion of iron in the infected host. This process is mediated
to a great extent by the proinflammatory cytokines IL-1␤ and
IL-6 (76, 109) and serves to reduce the availability of this
essential nutrient to the invading pathogens as part of the host’s
inherent defense system (65). As part of the acute-phase
response to infection, proinflammatory cytokines also trigger
the induction of the zinc-binding protein metallothionein (152).
During the course of pregnancy, this process leads to maternal
and fetal zinc deficiency, which has further been associated
with teratogenicity and abnormal developmental processes in
utero (33, 142).
In addition to its effects on micronutrient availability, maternal infection during pregnancy may also impair the fetal
supply for macronutrients. Indeed, it is well established that
peripheral cytokine elevation in response to infection induces a
set of behavioral and physiological changes collectively referred to as sickness behavior (32). Sickness behavior typically
includes fever, malaise, and reduced exploratory, and social
investigation, as well as decreased food and water intake,
accompanied usually by weight loss. The effects of immune
challenge on weight loss may be particularly relevant because
prenatal malnutrition has also been implicated as a risk factor
for various developmental disturbances (73, 84).
Finally, it should also be noted that at least part of the
changes to the microbiome that emerge following prenatal
infection (59) may have an early prenatal origin. The conventional view is that microbial colonization begins at birth when
the neonate is first exposed to the microbiome of the mother
and the surrounding environment (39, 82), implying furthermore that the fetal environment is sterile and, therefore, lacks
a microbiome before birth. This view has recently been challenged by findings showing that the human placenta is not
sterile but, in fact, is colonized with nonpathogenic commensal
microbiota (1). Perhaps even more important are the findings
suggesting that the microbial composition of the human placenta can be modified by maternal infection during pregnancy,
even if the infectious process takes place during the time of
conception or in early gestation (1). One important implication
from these recent findings is that modifications of the placental
microbiome, be it as a result of maternal infection or by other
environmental factors, can critically shape the development of
the offspring’s microbiome and, thus, predispose the developing organism to dysbiosis and other microbiome-associated
abnormalities.
Defining the Next Steps
Prenatal immunological adversities, such as maternal infection, have been widely acknowledged to contribute to an
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
Fig. 2. Possible mechanisms mediating the pathological effects of maternal
infection on the developing organism in utero. Maternal infection during
pregnancy induces a number of pathophysiological responses in the maternal
host, including production of soluble immune factors, such as cytokines,
reactive oxygen species, and soluble endocrine factors, such as stress hormones. Some of these factors might cross the placental barrier and enter the
fetal environment, thereby causing fetal inflammation and oxidative stress.
Abnormal fetal expression of these factors might impair the normal development of peripheral and central organs by modifying the differentiation, proliferation, and/or migration of target cells. These processes may involve
alterations in gene expression via epigenetic modifications. Maternal infection
during pregnancy can further induce inflammatory response in the placenta and
cause placental insufficiency, which, in turn can cause fetal hypoxemia. In
addition, infection can cause (temporary) states of macronutrient and micronutrient deficiency, which limit the fetal availability of essential nutrients
necessary for normal fetal development and growth. Finally, maternal infection
during pregnancy can modify the microbial composition of the placenta, which
might alter the development of the offspring’s microbiome and thus predispose
the developing organism to dysbiosis and other microbiome-associated
abnormalities.
R7
Review
R8
PERIPHERAL EFFECTS OF PRENATAL INFECTION
Perspectives and Significance
Several recent epidemiological and animal studies suggest
that infection-related prenatal adversities can negatively affect
various physiological and metabolic functions beyond those
typically associated with primary defects in CNS development.
These include excessive accumulation of adipose tissue and
increased body weight, impaired glycemic regulation and insulin resistance, altered myeloid lineage development in favor
of proinflammatory signaling, increased gut permeability, and
changes in microbiota composition. The existing data suggest
that these peripheral abnormalities can directly contribute to
the emergence of CNS abnormalities, so that a normalization
of peripheral symptoms effectively alleviates behavioral deficits induced by prenatal infection. Thus, it follows that seemingly unrelated central and peripheral effects of prenatal infection may not represent separate pathological entities, but rather,
they may be considered as pieces of the same puzzle. In fact,
there might be a “vicious circle” involving constant pathological interactions between developmentally acquired brain abnormalities and peripheral dysfunctions, in which deficient
peripheral functions facilitate or maintain CNS abnormalities,
and vice versa. Thus, targeting peripheral abnormalities may
represent a valuable strategy to improve the multitude of
behavioral abnormalities that can emerge in subjects with
prenatal infectious histories.
ACKNOWLEDGMENTS
Related work by the authors has been supported by grants from the Swiss
National Science Foundation (310030_146217; to U. Meyer) and from ETH
Zurich, Switzerland (ETH Research Grant 25_13-2; to U. Meyer and W.
Langhans).
Present address: U. Meyer, Institute of Pharmacology and Toxicology,
University of Zürich-Vetsuisse, Zürich, 8057 Switzerland.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
Author contributions: M.A.L. and U.M. prepared figures; M.A.L. and U.M.
drafted manuscript; M.A.L., W.L., and U.M. edited and revised manuscript;
M.A.L., W.L., and U.M. approved final version of manuscript.
REFERENCES
1. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J.
The placenta harbors a unique microbiome. Sci Transl Med 6: 237ra65,
2014.
2. Abazyan B, Nomura J, Kannan G, Ishizuka K, Tamashiro KL,
Nucifora F, Pogorelov V, Ladenheim B, Yang C, Krasnova IN, Cadet
JL, Pardo C, Mori S, Kamiya A, Vogel MW, Sawa A, Ross CA,
Pletnikov MV. Prenatal interaction of mutant DISC1 and immune
activation produces adult psychopathology. Biol Psychiatry 68: 1172–
1181.
3. Aguilar-Valles A, Flores C, Luheshi GN. Prenatal inflammation-induced hypoferremia alters dopamine function in the adult offspring in rat:
relevance for schizophrenia. PLoS One 5: e10967, 2010.
4. Aguilar-Valles A, Jung S, Poole S, Flores C, Luheshi GN. Leptin and
interleukin-6 alter the function of mesolimbic dopamine neurons in a
rodent model of prenatal inflammation. Psychoneuroendocrinology 37:
956 –969, 2012.
5. Arsenault D, St-Amour I, Cisbani G, Rousseau LS, Cicchetti F. The
different effects of LPS and poly I:C prenatal immune challenges on the
behavior, development and inflammatory responses in pregnant mice and
their offspring. Brain Behav Immun 38: 77–90, 2014.
6. Ashdown H, Dumont Y, Ng M, Poole S, Boksa P, Luheshi GN. The
role of cytokines in mediating effects of prenatal infection on the fetus:
implications for schizophrenia. Mol Psychiatry 11: 47–55, 2006.
7. Atladóttir HO, Thorsen P, Østergaard L, Schendel DE, Lemcke S,
Abdallah M, Parner ET. Maternal infection requiring hospitalization
during pregnancy and autism spectrum disorders. J Autism Dev Disord
40: 1423–1430, 2010.
8. Basu S, Haghiac M, Surace P, Challier JC, Guerre-Millo M, Singh K,
Waters T, Minium J, Presley L, Catalano PM, Hauguel-de Mouzon
S. Pregravid obesity associates with increased maternal endotoxemia and
metabolic inflammation. Obesity (Silver Spring) 19: 476 –482, 2011.
9. Bauman ML. Medical comorbidities in autism: challenges to diagnosis
and treatment. Neurotherapeutics 7: 320 –327, 2010.
10. Baumann N, Turpin JC. Neurochemistry of stress. An overview.
Neurochem Res 35: 1875–1879, 2010.
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
increased risk of neurodevelopmental brain disorders. Somewhat less attention has been given to the long-term consequences of prenatal infection on peripheral functions. Hence,
additional work is clearly warranted to further define the
putative effects of infection-related prenatal adversities on
postnatal physiology and metabolism. What is perhaps most
urgently needed to achieve this goal is more comprehensive
evidence from human epidemiological studies. To our knowledge, only one epidemiological study has yet directly explored
the putative association between maternal infection during
pregnancy and increased risk of obesity in the offspring (25).
Even though this study included a large number of subjects, it
requires replication and further extension to other physiological and metabolic parameters. It should also be mentioned that
exposures were broadly defined as “infections requiring hospitalization” in this first epidemiological study, so that the
possible relevance of pathogen specificity remained unexplored. Hence, we do not know whether long-term metabolic
abnormalities can similarly be induced by prenatal exposures
to various infectious pathogens, or alternatively, whether this
association is dependent on the precise identity of the pathogen. One powerful approach to overcome this limitation would
be the use of prospective epidemiological designs, in which a
specific infectious pathogen or inflammatory marker in prenatal life can be measured quantitatively. Such prospective epidemiological research has been proven indispensible for the
establishment of associations between prenatal exposure to
specific infectious pathogens and later risk of neuropsychiatric
disease (19, 22, 116). Hence, the future identification of possible long-term effects of prenatal infection on general physiology and metabolism might strongly benefit from such prospective epidemiological research.
For ethical and technical reasons, however, human epidemiological research will not be able to establish causality for such
associations and will be limited in its capacity to unravel the
downstream cellular and molecular mechanisms affecting normal physiological development. The examination of epidemiologically relevant risk factors in animal models, thus, remains
an important field of research to overcome these limitations. As
outlined above, several animal models of prenatal immune
challenge exist, so that their continuous implementation will be
pivotal to extend our knowledge on pathophysiological mechanisms underlying the association between prenatal infection
and peripheral abnormalities. In these attempts, a special emphasis should be placed on elucidating causal (and bidirectional) relationships between peripheral abnormalities and neurobehavioral deficits. These latter investigations may help to
establish holistic therapeutic interventions with beneficial effects on both central and peripheral dysfunctions.
Review
PERIPHERAL EFFECTS OF PRENATAL INFECTION
35. Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson
A, Hibberd ML, Forssberg H, Pettersson S. Normal gut microbiota
modulates brain development and behavior. Proc Natl Acad Sci USA
108: 3047–3052, 2011.
36. Dinan TG, Cryan JF. Melancholic microbes: a link between gut
microbiota and depression? Neurogastroenterol Motil 25: 713–719.
37. Dover GJ. The Barker hypothesis: how pediatricans will diagnose and
prevent common adult-onset diseases. Trans Am Clin Climatol Assoc
120: 199 –207, 2009.
38. Drong AW, Lindgren CM, McCarthy MI. The genetic and epigenetic
basis of type 2 diabetes and obesity. Clin Pharmacol Ther 92: 707–715,
2012.
39. Fanaro S, Chierici R, Guerrini P, Vigi V. Intestinal microflora in early
infancy: composition and development. Acta Paediatr Suppl 91: 48 –55,
2003.
40. Fernandez-Egea E, Miller B, Garcia-Rizo C, Bernardo M, Kirkpatrick B. Metabolic effects of olanzapine in patients with newly diagnosed
psychosis. J Clin Psychopharmacol 31: 154 –159, 2011.
41. Freeman DJ. Effects of maternal obesity on fetal growth and body
composition: implications for programming and future health. Semin
Fetal Neonatal Med 15: 113–118, 2010.
42. Galvão MC, Chaves-Kirsten GP, Queiroz-Hazarbassanov N, Carvalho VM, Bernardi MM, Kirsten TB. Prenatal zinc reduces stress
response in adult rat offspring exposed to lipopolysaccharide during
gestation. Life Sci 120: 54 –60, 2015.
43. Garay PA, Hsiao EY, Patterson PH, McAllister AK. Maternal immune activation causes age- and region-specific changes in brain cytokines in offspring throughout development. Brain Behav Immun 31:
54 –68, 2013.
44. Giovanoli S, Engler H, Engler A, Richetto J, Voget M, Willi R,
Winter C, Riva MA, Mortensen PB, Feldon J, Schedlowski M, Meyer
U. Stress in puberty unmasks latent neuropathological consequences of
prenatal immune activation in mice. Science 339: 1095–1099, 2013.
45. Girard S, Tremblay L, Lepage M, Sébire G. IL-1 receptor antagonist
protects against placental and neurodevelopmental defects induced by
maternal inflammation. J Immunol 184: 3997–4005, 2010.
46. Gittenberger-de Groot AC, Bartelings MM, Poelmann RE, Haak
MC, Jongbloed MR. Embryology of the heart and its impact on
understanding fetal and neonatal heart disease. Semin Fetal Neonatal
Med 18: 237–244, 2013.
47. Golub R, Cumano A. Embryonic hematopoiesis. Blood Cells Mol Dis
51: 226 –231, 2013.
48. Graeber MB. Neuroinflammation: no rose by any other name. Brain
Pathol 24: 620 –622, 2014.
49. Haddad JJ, Saadé NE, Safieh-Garabedian B. Cytokines and neuroimmune-endocrine interactions: a role for the hypothalamic-pituitaryadrenal revolving axis. J Neuroimmunol 133: 1–19, 2002.
50. 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.
51. Harvey L, Boksa P. A stereological comparison of GAD67 and reelin
expression in the hippocampal stratum oriens of offspring from two
mouse models of maternal inflammation during pregnancy. Neuropharmacology 62: 1767–1776, 2012.
52. 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.
53. Heald A. Physical health in schizophrenia: a challenge for antipsychotic
therapy. Eur Psychiatry 25 Suppl 2: S6 –S11, 2010.
54. Heerwagen MJ, Miller MR, Barbour LA, Friedman JE. Maternal
obesity and fetal metabolic programming: a fertile epigenetic soil. Am J
Physiol Regul Integr Comp Physiol 299: R711–R722, 2010.
55. Hei MY, Yi ZW. Environmental factors for the development of fetal
urinary malformations. World J Pediatr 10: 17–23, 2014.
56. Holmlund U, Cebers G, Dahlfors AR, Sandstedt B, Bremme K,
Ekström ES, Scheynius A. Expression and regulation of the pattern
recognition receptors Toll-like receptor-2 and Toll-like receptor-4 in the
human placenta. Immunology 107: 145–151, 2002.
57. Howerton CL, Bale TL. Prenatal programing: at the intersection of
maternal stress and immune activation. Horm Behav 62: 237–242, 2012.
58. Hsiao EY, McBride SW, Chow J, Mazmanian SK, Patterson PH.
Modeling an autism risk factor in mice leads to permanent immune
dysregulation. Proc Natl Acad Sci USA 109: 12,776 –12,781, 2012.
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
11. Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, Deng Y,
Blennerhassett P, Macri J, McCoy KD, Verdu EF, Collins SM. The
intestinal microbiota affect central levels of brain-derived neurotropic
factor and behavior in mice. Gastroenterology 141: 599 –609, 2011.
12. Berthoud HR, Neuhuber WL. Functional and chemical anatomy of the
afferent vagal system. Auton Neurosci 85: 1–17, 2000.
13. Bhatia V, Tandon RK. Stress and the gastrointestinal tract. J Gastroenterol Hepatol 20: 332–339, 2005.
14. Bilbo SD, Schwarz JM. Early-life programming of later-life brain and
behavior: a critical role for the immune system. Front Behav Neurosci 3:
14, 2009.
15. Boksa P. Effects of prenatal infection on brain development and behavior: a review of findings from animal models. Brain Behav Immun 24:
881–897, 2010.
16. Borre YE, O’Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF.
Microbiota and neurodevelopmental windows: implications for brain
disorders. Trends Mol Med 20: 509 –518, 2014.
17. Borrell J, Vela JM, Arévalo-Martin A, Molina-Holgado E, Guaza C.
Prenatal immune challenge disrupts sensorimotor gating in adult rats.
Implications for the etiopathogenesis of schizophrenia. Neuropsychopharmacology 26: 204 –215, 2002.
18. Bronson SL, Bale TL. Prenatal stress-induced increases in placental
inflammation and offspring hyperactivity are male-specific and ameliorated by maternal antiinflammatory treatment. Endocrinology 155: 2635–
2646, 2014.
19. Brown AS, Derkits EJ. Prenatal infection and schizophrenia: a review
of epidemiologic and translational studies. Am J Psychiatry 167: 261–
280, 2010.
20. Brown LD. Endocrine regulation of fetal skeletal muscle growth: impact
on future metabolic health. J Endocrinol 221: R13–R29, 2014.
21. Brown AS. The environment and susceptibility to schizophrenia. Prog
Neurobiol 93: 23–58, 2011.
22. Canetta SE, Bao Y, Co MD, Ennis FA, Cruz J, Terajima M, Shen L,
Kellendonk C, Schaefer CA, Brown AS. Serological documentation of
maternal influenza exposure and bipolar disorder in adult offspring. Am
J Psychiatry 171: 557–563, 2014.
23. Cheung C, Yu K, Fung G, Leung M, Wong C, Li Q, Sham P, Chua
S, McAlonan G. Autistic disorders and schizophrenia: related or remote?
An anatomical likelihood estimation. PLoS One 5: e12233, 2010.
24. Chow Z, Banerjee A, Hickey MJ. Controlling the fire - tissue-specific
mechanisms of effector regulatory T-cell homing. Immunol Cell Biol 93:
355–363, 2015.
25. Cocoros NM, Lash TL, Nørgaard M, Farkas DK, DeMaria A Jr,
Sørensen HT. Hospitalized prenatal and childhood infections and obesity in Danish male conscripts. Ann Epidemiol 23: 307–313, 2013.
26. Critchfield JW, van Hemert S, Ash M, Mulder L, Ashwood P. The
potential role of probiotics in the management of childhood autism
spectrum disorders. Gastroenterol Res Pract 2011: 161358, 2011.
27. Critchley HD, Harrison NA. Visceral influences on brain and behavior.
Neuron 77: 624 –638, 2013.
28. Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the
gut microbiota on brain and behaviour. Nat Rev Neurosci 13: 701–712,
2012.
29. Dahlgren J, Nilsson C, Jennische E, Ho HP, Eriksson E, Niklasson A,
Björntorp P, Albertsson Wikland K, Holmäng A. Prenatal cytokine
exposure results in obesity and gender-specific programming. Am J
Physiol Endocrinol Metab 281: E326 –E334, 2001.
30. Dahlgren J, Samuelsson AM, Jansson T, Holmäng A. Interleukin-6 in
the maternal circulation reaches the rat fetus in mid-gestation. Pediatr
Res 60: 147–151, 2006.
31. Dammann O, Leviton A. Maternal intrauterine infection, cytokines, and
brain damage in the preterm newborn. Pediatr Res 42: 1–8, 1997.
32. Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW.
From inflammation to sickness and depression: when the immune system
subjugates the brain. Nat Rev Neurosci 9: 46 –56, 2008.
33. Daston GP, Overmann GJ, Baines D, Taubeneck MW, LehmanMcKeeman LD, Rogers JM, Keen CL. Altered Zn status by alphahederin in the pregnant rat and its relationship to adverse developmental
outcome. Reprod Toxicol 8: 15–24, 1994.
34. de Theije CG, Wopereis H, Ramadan M, van Eijndthoven T, Lambert J, Knol J, Garssen J, Kraneveld AD, Oozeer R. Altered gut
microbiota and activity in a murine model of autism spectrum disorders.
Brain Behav Immun 37: 197–206, 2014.
R9
Review
R10
PERIPHERAL EFFECTS OF PRENATAL INFECTION
81. Luo ZC, Xiao L, Nuyt AM. Mechanisms of developmental programming of the metabolic syndrome and related disorders. World J Diabetes
1: 89 –98, 2010.
82. Mackie RI, Sghir A, Gaskins HR. Developmental microbial ecology of
the neonatal gastrointestinal tract. Am J Clin Nutr 69: 1035S–1045S,
1999.
83. Malkova NV, Yu CZ, Hsiao EY, Moore MJ, Patterson PH. Maternal
immune activation yields offspring displaying mouse versions of the
three core symptoms of autism. Brain Behav Immun 26: 607–616, 2012.
84. Marques AH, Bjørke-Monsen AL, Teixeira AL, Silverman MN.
Maternal stress, nutrition and physical activity: Impact on immune
function, CNS development and psychopathology. Brain Res In press.
85. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation
and polarization. Front Biosci 13: 453–461, 2008.
86. Masi A, Quintana DS, Glozier N, Lloyd AR, Hickie IB, Guastella AJ.
Cytokine aberrations in autism spectrum disorder: a systematic review
and meta-analysis. Mol Psychiatry 20: 440 –446, 2015.
87. Mayer EA, Knight R, Mazmanian SK, Cryan JF, Tillisch K. Gut
microbes and the brain: paradigm shift in neuroscience. J Neurosci 34:
15,490 –15,496, 2014.
88. Mayer EA. Gut feelings: the emerging biology of gut-brain communication. Nat Rev Neurosci 12: 453–466, 2011.
89. McMillen IC, Robinson JS. Developmental origins of the metabolic
syndrome: prediction, plasticity, and programming. Physiol Rev 85:
571–633, 2005.
90. Meyer JM, Stahl SM. The metabolic syndrome and schizophrenia. Acta
Psychiatr Scand 119: 4 –14, 2009.
91. Meyer U, Engler A, Weber L, Schedlowski M, Feldon J. Preliminary
evidence for a modulation of fetal dopaminergic development by maternal immune activation during pregnancy. Neuroscience 154: 701–709,
2008.
92. Meyer U, Feldon J, Dammann O. Schizophrenia and autism: both
shared and disorder-specific pathogenesis via perinatal inflammation?
Pediatr Res 69: 26R–33R, 2011.
93. Meyer U, Feldon J, Fatemi SH. In-vivo rodent models for the experimental investigation of prenatal immune activation effects in neurodevelopmental brain disorders. Neurosci Biobehav Rev 33: 1061–1079,
2009.
94. Meyer U, Feldon J, Schedlowski M, Yee BK. Towards an immunoprecipitated neurodevelopmental animal model of schizophrenia. Neurosci Biobehav Rev 29: 913–947, 2005.
95. Meyer U, Feldon J, Yee BK. A review of the fetal brain cytokine
imbalance hypothesis of schizophrenia. Schizophr Bull 35: 959 –972,
2009.
96. Meyer U, Murray PJ, Urwyler A, Yee BK, Schedlowski M, Feldon J.
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.
97. Meyer U, Nyffeler M, Engler A, Urwyler A, Schedlowski M, Knuesel
I, Yee BK, Feldon J. The time of prenatal immune challenge determines
the specificity of inflammation-mediated brain and behavioral pathology.
J Neurosci 26: 4752–4762, 2006.
98. Meyer U, Nyffeler M, Schwendener S, Knuesel I, Yee BK, Feldon J.
Relative prenatal and postnatal maternal contributions to schizophreniarelated neurochemical dysfunction after in utero immune challenge.
Neuropsychopharmacology 33: 441–456, 2008.
99. Meyer U, Yee BK, Feldon J. The neurodevelopmental impact of
prenatal infections at different times of pregnancy: the earlier the worse?
Neuroscientist 13: 241–256, 2007.
100. Meyer U. Prenatal poly(I:C) exposure and other developmental immune
activation models in rodent systems. Biol Psychiatry 75: 307–315, 2014.
101. Miller BJ, Buckley P, Seabolt W, Mellor A, Kirkpatrick B. Metaanalysis of cytokine alterations in schizophrenia: clinical status and
antipsychotic effects. Biol Psychiatry 70: 663–671, 2011.
102. Miller BJ, Culpepper N, Rapaport MH, Buckley P. Prenatal inflammation and neurodevelopment in schizophrenia: a review of human
studies. Prog Neuropsychopharmacol Biol Psychiatry 42: 92–100, 2013.
103. Moreno-De-Luca A, Myers SM, Challman TD, Moreno-De-Luca D,
Evans DW, Ledbetter DH. Developmental brain dysfunction: revival
and expansion of old concepts based on new genetic evidence. Lancet
Neurol 12: 406 –414, 2013.
104. Morton GJ, Meek TH, Schwartz MW. Neurobiology of food intake in
health and disease. Nat Rev Neurosci 15: 367–378, 2014.
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
59. Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T,
Codelli JA, Chow J, Reisman SE, Petrosino JF, Patterson PH,
Mazmanian SK. Microbiota modulate behavioral and physiological
abnormalities associated with neurodevelopmental disorders. Cell 155:
1451–1463, 2013.
60. Huhta J, Linask KK. Environmental origins of congenital heart disease:
the heart-placenta connection. Semin Fetal Neonatal Med 18: 245–250,
2013.
61. Johnson J, Anderson B, Pass RF. Prevention of maternal and congenital cytomegalovirus infection. Clin Obstet Gynecol 55: 521–530, 2012.
62. Kenny PJ. Common cellular and molecular mechanisms in obesity and
drug addiction. Nat Rev Neurosci 12: 638 –651, 2011.
63. Kirkpatrick B, Miller BJ, Garcia-Rizo C, Fernandez-Egea E, Bernardo M. Is abnormal glucose tolerance in antipsychotic-naive patients
with nonaffective psychosis confounded by poor health habits? Schizophr
Bull 38: 280 –284, 2012.
64. Kirkpatrick B, Miller BJ. Inflammation and schizophrenia. Schizophr
Bull 39: 1174 –1179, 2013.
65. Kluger MJ, Rothenburg BA. Fever and reduced iron: their interaction
as a host defense response to bacterial infection. Science 203: 374 –376,
1979.
66. Kohen R, Nyska A. Oxidation of biological systems: oxidative stress
phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol 30: 620 –650, 2002.
67. Konturek PC, Brzozowski T, Konturek SJ. Stress and the gut: pathophysiology, clinical consequences, diagnostic approach and treatment
options. J Physiol Pharmacol 62: 591–599, 2011.
68. Kovacs CS. Bone development and mineral homeostasis in the fetus and
neonate: roles of the calciotropic and phosphotropic hormones. Physiol
Rev 94: 1143–1218, 2014.
69. Krstic D, Madhusudan A, Doehner J, Vogel P, Notter T, Imhof C,
Manalastas A, Hilfiker M, Pfister S, Schwerdel C, Riether C, Meyer
U, Knuesel I. Systemic immune challenges trigger and drive Alzheimerlike neuropathology in mice. J Neuroinflammation 9: 151, 2012.
70. Kunzmann S, Collins JJ, Kuypers E, Kramer BW. Thrown off
balance: the effect of antenatal inflammation on the developing lung and
immune system. Am J Obstet Gynecol 208: 429 –437, 2013.
71. Kwik-Uribe CL, Gietzen D, German JB, Golub MS, Keen CL.
Chronic marginal iron intakes during early development in mice result in
persistent changes in dopamine metabolism and myelin composition. J
Nutr 130: 2821–2830, 2000.
72. Kwik-Uribe CL, Golub MS, Keen CL. Chronic marginal iron intakes
during early development in mice alter brain iron concentrations and
behavior despite postnatal iron supplementation. J Nutr 130: 2040 –2048,
2000.
73. Lakshmy R. Metabolic syndrome: role of maternal undernutrition and
fetal programming. Rev Endocr Metab Disord 14: 229 –240, 2013.
74. Lanté F, Meunier J, Guiramand J, De Jesus Ferreira MC, Cambonie
G, Aimar R, Cohen-Solal C, Maurice T, Vignes M, Barbanel G. Late
N-acetylcysteine treatment prevents the deficits induced in the offspring
of dams exposed to an immune stress during gestation. Hippocampus 18:
602–609, 2008.
75. Lanté F, Meunier J, Guiramand J, Maurice T, Cavalier M, de Jesus
Ferreira MC, Aimar R, Cohen-Solal C, Vignes M, Barbanel G.
Neurodevelopmental damage after prenatal infection: role of oxidative
stress in the fetal brain. Free Radic Biol Med 42: 1231–1245, 2007.
76. Lee P, Peng H, Gelbart T, Wang L, Beutler E. Regulation of hepcidin
transcription by interleukin-1 and interleukin-6. Proc Natl Acad Sci USA
102: 1906 –1910, 2005.
77. Li M, Sloboda DM, Vickers MH. Maternal obesity and developmental
programming of metabolic disorders in offspring: evidence from animal
models. Exp Diabetes Res 2011: 592408, 2011.
78. Lin YL, Lin SY, 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.
79. Lipina TV, Zai C, Hlousek D, Roder JC, Wong AH. Maternal immune
activation during gestation interacts with Disc1 point mutation to exacerbate schizophrenia-related behaviors in mice. J Neurosci 33: 7654 –
7666, 2013.
80. Lumbers ER, Pringle KG, Wang Y, Gibson KJ. The renin-angiotensin
system from conception to old age: the good, the bad and the ugly. Clin
Exp Pharmacol Physiol 40: 743–752, 2013.
Review
PERIPHERAL EFFECTS OF PRENATAL INFECTION
128. Schaefer TM, Fahey JV, Wright JA, Wira CR. Innate immunity in the
human female reproductive tract: antiviral response of uterine epithelial
cells to the TLR3 agonist poly(I:C). J Immunol 174: 992–1002, 2005.
129. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central
nervous system control of food intake. Nature 404: 661–671, 2000.
130. Seckl JR, Holmes MC. Mechanisms of disease: glucocorticoids, their
placental metabolism and fetal ‘programming’ of adult pathophysiology.
Nat Clin Pract Endocrinol Metab 3: 479 –488, 2007.
131. Sell H, Eckel J. Adipose tissue inflammation: novel insight into the role
of macrophages and lymphocytes. Curr Opin Clin Nutr Metab Care 13:
366 –370, 2010.
132. Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol 6: 1191–1197, 2005.
133. Severance EG, Yolken RH, Eaton WW. Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: more than a
gut feeling. Schizophr Res In press.
134. Shields RW Jr. Functional anatomy of the autonomic nervous system. J
Clin Neurophysiol 10: 2–13, 1993.
135. Shimogori T, Lee DA, Miranda-Angulo A, Yang Y, Wang H, Jiang
L, Yoshida AC, Kataoka A, Mashiko H, Avetisyan M, Qi L, Qian J,
Blackshaw S. A genomic atlas of mouse hypothalamic development. Nat
Neurosci 13: 767–775, 2010.
136. Short DD, Hawley JM, McCarthy MF. Management of schizophrenia
with medical disorders: cardiovascular, pulmonary, and gastrointestinal.
Psychiatr Clin North Am 32: 759 –773, 2009.
137. Smith SE, Li J, Garbett K, Mirnics K, Patterson PH. Maternal
immune activation alters fetal brain development through interleukin-6. J
Neurosci 27: 10,695–10,702, 20107.
138. Smoller W, Craddock N, Kendler K, Lee PH, Neale BM, CrossDisorder Group of the Psychiatric Genomics Consortium. Identification of risk loci with shared effects on five major psychiatric disorders:
A genome-wide analysis. Lancet 381: 1371–1379, 2013.
139. Stahl SM, Mignon L, Meyer JM. Which comes first: atypical antipsychotic treatment or cardiometabolic risk? Acta Psychiatr Scand 119:
171–179, 2009.
140. Symonds ME, Mostyn A, Pearce S, Budge H, Stephenson T. Endocrine and nutritional regulation of fetal adipose tissue development. J
Endocrinol 179: 293–299, 2003.
141. Symonds ME, Sebert SP, Hyatt MA, Budge H. Nutritional programming of the metabolic syndrome. Nat Rev Endocrinol 5: 604 –610.
142. Taubeneck MW, Daston GP, Rogers JM, Gershwin ME, Ansari A,
Keen CL. Tumor necrosis factor-alpha alters maternal and embryonic
zinc metabolism and is developmentally toxic in mice. J Nutr 125:
908 –919, 1995.
143. Thakore JH, Mann JN, Vlahos I, Martin A, Reznek R. Increased
visceral fat distribution in drug-naive and drug-free patients with schizophrenia. Int J Obes Relat Metab Disord 26: 137–141, 2002.
144. Tilg H, Kaser A. Gut microbiome, obesity, and metabolic dysfunction.
J Clin Invest 121: 2126 –2132, 2011.
145. Tracey KJ. The inflammatory reflex. Nature 420: 853–859, 2002.
146. Traynor TR, Majde JA, Bohnet SG, Krueger JM. Intratracheal
double-stranded RNA plus interferon-gamma: a model for analysis of the
acute phase response to respiratory viral infections. Life Sci 74: 2563–
2576, 2004.
147. Triantafilou M, Triantafilou K. Lipopolysaccharide recognition: CD14,
TLRs, and the LPS-activation cluster. Trends Immunol 23: 301–304,
2002.
148. Turner JR. Intestinal mucosal barrier function in health and disease. Nat
Rev Immunol 9: 799 –809, 2009.
149. Unger EL, Paul T, Murray-Kolb LE, Felt B, Jones BC, Beard JL.
Early iron deficiency alters sensorimotor development and brain monoamines in rats. J Nutr 137: 118 –124, 2007.
150. Upthegrove R, Manzanares-Teson N, Barnes NM. Cytokine function
in medication-naive first episode psychosis: a systematic review and
meta-analysis. Schizophr Res 155: 101–108, 2014.
151. Urakubo A, Jarskog LF, Lieberman JA, Gilmore JH. Prenatal exposure to maternal infection alters cytokine expression in the placenta,
amniotic fluid, and fetal brain. Schizophr Res 47: 27–36, 2001.
152. Vallee BL, Falchuk KH. The biochemical basis of zinc physiology.
Physiol Rev 73: 79 –118.
153. Verma SK, Subramaniam M, Liew A, Poon LY. Metabolic risk factors
in drug-naive patients with first-episode psychosis. J Clin Psychiatry 70:
997–1000, 2009.
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
105. Mraz M, Haluzik M. The role of adipose tissue immune cells in obesity
and low-grade inflammation. J Endocrinol 222: R113–R127, 2014.
106. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S,
Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, Locati M,
Mantovani A, Martinez FO, Mege JL, Mosser DM, Natoli G, Saeij
JP, Schultze JL, Shirey KA, Sica A, Suttles J, Udalova I, van
Ginderachter JA, Vogel SN, Wynn TA. Macrophage activation and
polarization: nomenclature and experimental guidelines. Immunity 41:
14 –20, 2014.
107. Nathan C, Shiloh MU. Reactive oxygen and nitrogen intermediates in
the relationship between mammalian hosts and microbial pathogens.
Proc Natl Acad Sci USA 97: 8841–8848, 2000.
108. Naviaux JC, Schuchbauer MA, Li K, Wang L, Risbrough VB, Powell
SB, Naviaux RK. Reversal of autism-like behaviors and metabolism in
adult mice with single-dose antipurinergic therapy. Transl Psychiatry 4:
e400, 2014.
109. Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK,
Ganz T. IL-6 mediates hypoferremia of inflammation by inducing the
synthesis of the iron regulatory hormone hepcidin. J Clin Invest 113:
1271–1276, 2007.
110. Niklasson B, Samsioe A, Blixt M, Sandler S, Sjöholm A, Lagerquist
E, Lernmark A, Klitz W. Prenatal viral exposure followed by adult
stress produces glucose intolerance in a mouse model. Diabetologia 49:
2192–2129, 2006.
111. Nilsson C, Larsson BM, Jennische E, Eriksson E, Björntorp P, York
DA, Holmäng A. Maternal endotoxemia results in obesity and insulin
resistance in adult male offspring. Endocrinology 142: 2622–2630, 2001.
112. Onore CE, Schwartzer JJ, Careaga M, Berman RF, Ashwood P.
Maternal immune activation leads to activated inflammatory macrophages in offspring. Brain Behav Immun 38: 220 –226, 2014.
113. Owen MJ. Intellectual disability and major psychiatric disorders: A
continuum of neurodevelopmental causality. Br J Psychiatry 200: 268 –
269, 2012.
114. Ozawa K, Hashimoto K, Kishimoto T, Shimizu E, Ishikura H, Iyo M.
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.
115. Pacheco-López G, Giovanoli S, Langhans W, Meyer U. Priming of
metabolic dysfunctions by prenatal immune activation in mice: relevance
to schizophrenia. Schizophr Bull 39: 319 –329, 2013.
116. Parboosing R, Bao Y, Shen L, Schaefer CA, Brown AS. Gestational
influenza and bipolar disorder in adult offspring. JAMA Psychiatry 70:
677–685, 2013.
117. Patterson PH. Maternal infection and immune involvement in autism.
Trends Mol Med 17: 389 –394, 2011.
118. Pavlov VA, Tracey KJ. The vagus nerve and the inflammatory reflex—
linking immunity and metabolism. Nat Rev Endocrinol 8: 743–754,
2012.
119. Pineda E, Shin D, You SJ, Auvin S, Sankar R, Mazarati A. Maternal
immune activation promotes hippocampal kindling epileptogenesis in
mice. Ann Neurol 74: 11–19, 2013.
120. Pongratz G, Straub RH. Role of peripheral nerve fibres in acute and
chronic inflammation in arthritis. Nat Rev Rheumatol 9: 117–126, 2013.
121. Radaelli T, Uvena-Celebrezze J, Minium J, Huston-Presley L, Catalano P, Hauguel-de Mouzon S. Maternal interleukin-6: marker of fetal
growth and adiposity. J Soc Gynecol Investig 13: 53–57, 2006.
122. Ransohoff RM, Perry VH. Microglial physiology: unique stimuli,
specialized responses. Annu Rev Immunol 27: 119 –145, 2009.
123. Roullet FI, Lai JK, Foster JA. In utero exposure to valproic acid and
autism—a current review of clinical and animal studies. Neurotoxicol
Teratol 36: 47–56, 2013.
124. Ryan MC, Collins P, Thakore JH. Impaired fasting glucose tolerance
in first-episode, drug-naive patients with schizophrenia. Am J Psychiatry
160: 284 –289, 2003.
125. Salamone JD, Correa M. The mysterious motivational functions of
mesolimbic dopamine. Neuron 76: 470 –485, 2012.
126. Salisbury AM, Begon M, Dove W, Niklasson B, Stewart JP. Ljungan
virus is endemic in rodents in the UK. Arch Virol 159: 547–551, 2014.
127. Sanders TR, Kim DW, Glendining KA, Jasoni CL. Maternal obesity
and IL-6 lead to aberrant developmental gene expression and deregulated
neurite growth in the fetal arcuate nucleus. Endocrinology 155: 2566 –
2577, 2014.
R11
Review
R12
PERIPHERAL EFFECTS OF PRENATAL INFECTION
154. Vuillermot S, Joodmardi E, Perlmann T, Ögren SO, Feldon J, Meyer
U. Prenatal immune activation interacts with genetic Nurr1 deficiency in
the development of attentional impairments. J Neurosci 32: 436 –451,
2012.
155. Vuillermot S, Weber L, Feldon J, Meyer U. A longitudinal examination of the neurodevelopmental impact of prenatal immune activation in
mice reveals primary defects in dopaminergic development relevant to
schizophrenia. J Neurosci 30: 1270 –1287, 2010.
156. Welberg LA, Seckl JR. Prenatal stress, glucocorticoids and the programming of the brain. J Neuroendocrinol 13: 113–128, 2001.
157. Willi R, Harmeier A, Giovanoli S, Meyer U. Altered GSK3␤ signaling
in an infection-based mouse model of developmental neuropsychiatric
disease. Neuropharmacology 73: 56 –65.
158. Zager A, Andersen ML, Tufik S, Palermo-Neto J. Maternal immune activation increases the corticosterone response to acute stress
without affecting the hypothalamic monoamine content and sleep
patterns in male mice offspring. Neuroimmunomodulation 21: 37–44,
2014.
159. Zaretsky MV, Alexander JM, Byrd W, Bawdon RE. Transfer of
inflammatory cytokines across the placenta. Obstet Gynecol 103: 546 –
550, 2004.
160. 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.
Downloaded from http://ajpregu.physiology.org/ by 10.220.32.246 on June 17, 2017
AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00087.2015 • www.ajpregu.org