key: cord-0768748-9yhnixmk authors: Aguilar-Valles, Argel; Rodrigue, Brandon; Matta-Camacho, Edna title: Maternal Immune Activation and the Development of Dopaminergic Neurotransmission of the Offspring: Relevance for Schizophrenia and Other Psychoses date: 2020-08-21 journal: Front Psychiatry DOI: 10.3389/fpsyt.2020.00852 sha: b79d204dd9acd812063af5c76b0475e847e332c9 doc_id: 768748 cord_uid: 9yhnixmk Prenatal infections have been linked to the development of schizophrenia (SCZ) and other neurodevelopmental disorders in the offspring, and work in animal models indicates that this is to occur through the maternal inflammatory response triggered by infection. Several studies in animal models demonstrated that acute inflammatory episodes are sufficient to trigger brain alterations in the adult offspring, especially in the mesolimbic dopamine (DA) system, involved in the pathophysiology of SCZ and other disorders involving psychosis. In the current review, we synthesize the literature on the clinical studies implicating prenatal infectious events in the development of SCZ. Then, we summarize evidence from animal models of maternal immune activation (MIA) and the behavioral and molecular alterations relevant for the function of the DAergic system. Furthermore, we discuss the evidence supporting the involvement of maternal cytokines, such as interleukin 6 (IL-6) and leptin (a hormone with effects on inflammation) in mediating the effects of MIA on the fetal brain, leading to the long-lasting effects on the offspring. In particular, IL-6 has been involved in mediating the effects of MIA animal models in the offspring through actions on the placenta, induction of IL-17a, or triggering the decrease in non-heme iron (hypoferremia). Maternal infection is very likely interacting with additional genetic and environmental risk factors in the development of SCZ; systematically investigating how these interactions produce specific phenotypes is the next step in understanding the etiology of complex psychiatric disorders. We are currently undergoing a SARS-CoV-2 pandemic, which like previous viral outbreaks [e.g., Zika (1)] can leave behind sequelae of health complications, including direct effects in the nervous system (2) and alterations of brain development if infections occur during perinatal stages. Indeed, maternal infection has been identified as a risk factor for several neurodevelopmental disorders such as cerebral palsy, intellectual disability, autism spectrum disorder (ASD), bipolar disorder (BD), and schizophrenia (SCZ) (3) (4) (5) (6) (7) (8) (9) . We will focus on reviewing the effects of maternal infection on the dopaminergic neurotransmitter system and the link with psychosis, particularly SCZ. SCZ is one of the top leading causes of disability worldwide (10) and the seventh most costly medical illness in modern society (11, 12) . SCZ is characterized by psychotic symptoms such as delusions and hallucinations (also known as the positive symptom dimension); alterations in drive and volition, including lack of motivation, blunted affect, social withdrawal, and reduction in spontaneous speech (the negative symptom dimension) and alterations in neurocognition, including difficulties in memory, attention, and executive functioning (the cognitive symptom dimension) (13) (14) (15) . The positive symptoms of SCZ overlap with different psychiatric disorders. Indeed, psychosis is also frequent during mood episodes in BD, severe depression, substance use disorder and neurodegenerative disorders (16) (17) (18) . Intriguingly, some SCZ-like psychopathological abnormalities (i.e., paranoid delusional thinking and auditory hallucinations) are expressed in an attenuated form in 5-8% of the otherwise healthy population, especially in individuals with schizotypal or schizoid personality traits (13, 19) . This extensive overlapping of symptoms and genetic risk factors with other psychiatric and neurological conditions is suggestive of a common underlying neuropathophysiology for these disorders, which, rather than discrete diagnoses, may represent a continuum that extends to the general population (13, 19, 20) . The classical dopamine (DA) hypothesis of SCZ (21) states that the hyperactivity of the DA system is responsible for the symptoms of the disorder. More recently, this hypothesis was elaborated to include the proposal that the hyperactivity of the mesolimbic DA system ( Figure 1A ) contributes to positive symptoms in SCZ. Meanwhile, impaired function of the DA system in the prefrontal cortex (PFC, Figure 1A ) contributes to the cognitive symptom dimension (22, 23) . The DA hypothesis of SCZ derives, in part, from the identification of the mechanisms of action of antipsychotics, many of which act as DA receptor 2 (D2 receptor) blockers (15) . Furthermore, pharmacological studies show that a single exposure to amphetamine (AMPH), a stimulant drug that increases extracellular levels of DA in striatal and cortical regions via release and reverse transport (24, 25) , evokes or exacerbates positive symptoms in SCZ patients at doses which do not induce psychosis in healthy subjects (26-28). Imaging studies demonstrate that a significant number of non-medicated SCZ patients show marked elevation of AMPH-induced striatal dopamine release in comparison to healthy volunteers (29-31). This response correlates significantly with the emergence or worsening of positive symptoms (31-35). Understanding the etiology of SCZ is an active area of research. However, evidence accumulated in the last three decades on environmental risk factors that affect early (IL-6 ) and, in turn, IL-17a. Both of these cytokines affect brain development in the fetus, increasing the risk for neurodevelopmental disorders, such as SCZ (SCZ). These cytokines can act indirectly on the placenta, or in the case of IL-6 through the induction of hypoferremia, a reduction in circulating non-heme iron. Adipose tissue can also release hormones such as leptin, which affects fetal development. neurodevelopment during pregnancy has led to the proposal of the neurodevelopmental hypothesis for SCZ (36-38). In this sense, accumulating evidence suggests that perinatal insults also contribute to an increase risk of developing BD (9), particularly those cases with psychoses (8) . SCZ has been hypothesized to have a neurodevelopmental origin (22) : an outcome of an aberration in developmental processes within the brain, which begins long before the onset of the clinical symptoms (36, 39). There are numerous independent lines of evidence supporting this hypothesis. For example, there is a conspicuous absence of gross physical damage or signs of progressive neurodegeneration in SCZ (22, 39) . Besides, children that go on to develop SCZ present behavioral, physical and brain morphological alterations, before the clinical onset of psychosis (15, 36, (39) (40) (41) . Finally, individuals who develop SCZ are more likely to have experienced pre-or perinatal adverse events (22, 42) , or adolescent disturbances in brain development, compared to control individuals (36, 39, 41, 43). These adverse events include intrauterine growth retardation, pregnancy and birth complications (44), nutritional deficiencies (45, 46) maternal stress (47), and maternal infections (48). There is also mounting evidence for the role of neurodevelopmental disturbances in the etiology of BD, as thoroughly reviewed by (9) . In this regard, there is high comorbidity between BD and other developmental disorders such as attention-deficit/hyperactivity disorder (ADHD) and ASD (9) . Remarkably, there are increased rates of BD due to obstetric complications, cesarean section birth and perinatal infection (8, 9, 49, 50) . Ecological studies, including those based on the subjective report of illness, suggest that SCZ is more prevalent in the offspring of women that were pregnant during periods of influenza epidemics (51, 52), as well as other types of infections, including diphtheria, pneumonia, measles, varicella zoster, mumps and poliovirus (4, (53) (54) (55) (56) . Similarly, SCZ is more prevalent among individuals born to pregnancies that occur during winter, a season associated with an increased frequency of respiratory infections (36, 51, 57). The main limitation of these studies is that "exposure to infection" was defined solely by the fact that the individual was pregnant during the time of the epidemic (i.e., based on the date of birth of the offspring). It was later shown that SCZ in the offspring is significantly associated with maternal infections using individual biomarkers of illness in the maternal serum or clinical diagnoses (4). These included respiratory infections (58), influenza (59, 60), rubella (61, 62), Toxoplasma gondii (63, 64), herpes simplex virus-2 (HSV-2) (65, 66), maternal genital or reproductive infections (67) , and maternal bacterial infections (68) . Some of these studies used a broad definition of psychosis, where both non-affective (e.g., SCZ) and affective (e.g., major depression or BDs with psychotic features) psychiatric disorders were included (62, 66, 69) . This suggests that maternal infection may be involved in the development of psychotic features that may not be necessarily restricted to those that characterize SCZ, but several other disorders as well. Indeed, MIA involving influenza has been linked to BD (9) [and Toxoplasma gondii infections to a lesser extent (70)], especially for those patients that also develop psychotic features (8) . What remains unclear from these studies is the critical stage (s) of gestation during which the developing brain may be more vulnerable to this prenatal insult. Indeed, those studies that have tried to dissect a specific trimester of gestation where vulnerability to MIA may be increased, have provided evidence for all three trimesters (58, 59, 68, 69, 71, 72) . Overall, effect sizes of prenatal infection across gestation and development of SCZ in the offspring range from 1.5 to 7 for different infections (73) , suggesting the existence of additional factors that confer vulnerability or resilience (6) . The wide variety of infections associated with SCZ and BD with psychosis suggests that there may be a common factor underlying increased susceptibility (74) . Therefore, it has been hypothesized that maternal immune activation (MIA), and the inflammatory mediators released following all types of infections (4), may be fundamentally involved. Epidemiological studies have provided some evidence supporting this hypothesis. Increased levels of maternal pro-inflammatory cytokines, specifically interleukin (IL)-8 (72), tumor necrosis factor (TNF)a (69, 71, 75, 76) , IL-6 (71, 75, 76), C-reactive protein (77) are associated with a higher risk of psychosis or SCZ in the offspring. Several animal models have been developed to investigate the immunological and neurobiological link between MIA and altered behavior in the offspring, with heavy emphasis in behavioral alterations. Initial approaches used prenatal infection with an influenza virus [at gestational day (GD) 9 in mice], followed by the application of a battery of behavioral tests relevant to SCZ in the adult offspring (78) . These studies showed that the adult offspring of infected mothers presented, compared to the offspring of control dams, decreased social interaction, reduced exploration in the open field, impaired performance in the novel object test, indicative of impaired working memory as observed in SCZ and diminished PPI of acoustic startle (78) . These behavioral alterations are analogous to aspects of SCZ. Further studies investigated the consequences of MIA using molecular immunogens in rats and mice. The viral mimic polyinosinic:polycytidylic acid (poly I:C) has been used to stimulate the maternal immune system (with one or multiple injections), at several stages of pregnancy in mice or rats, ranging from GD 8.5 until GD 18.5. The effects of these prenatal treatments have been extensively reviewed elsewhere (6, 7, (79) (80) (81) ; thus, we will focus on those consequences more closely relevant for psychosis. Prenatal stimulation with poly I:C induced deficits in an operational measure of sensorimotor gating (82) , pre-pulse inhibition of acoustic startle and increased sensitivity to the locomotor activating effects of cocaine, AMPH and methamphetamine, whose locomotor effects depend on the mesolimbic DA system . An overall trend regarding these two phenotypes is one where PPI deficits are more consistently observed when MIA occurs at gestational stages earlier than GD 16 in both mice and rats. At the same time, hyper-responsiveness to activators of the mesolimbic dopaminergic system appears when challenging the mothers at any developmental age [reviewed in (79) ]. The role of bacterial infection has also been investigated by using the Gram-negative bacterial cell wall component, lipopolysaccharide (LPS). In rats, injections at several stages of gestation, ranging from GD 9 until birth, induced, in the offspring, impairments in PPI, and increases in sensitivity to the locomotor effects of AMPH (106) (107) (108) (109) (110) (111) (112) (113) (114) (115) (116) (117) . LPS has also been administered either in alternate days (118) or daily throughout pregnancy (119, 120) . Similarly to acute LPS administration, these chronic prenatal treatments also induced impairments in PPI (118) (119) (120) . Turpentine (TURP) is an inflammatory agent whose injection [intramuscular (i.m.)] produces localized necrotic damage (121) and the sequential induction of TNFa and IL-1b at the site of injury, which trigger IL-6 release into the circulation (122, 123) . Using TURP at GD 15 or 18 in rats, we found that an earlier challenge with TURP induces greater maternal inflammatory response compared to later in gestation (124) . Furthermore, this difference in the inflammatory response during pregnancy correlates with the effect on the offspring, such that treatment at GD 15 induces impairment in PPI and hyper-responsiveness to AMPH, while the same treatment at GD 18 does not affect any of these behaviors (124) . Overall, some of the alterations in behavior induced by either polyI:C or LPS, have been shown to appear in the adult but not in the juvenile offspring (84, 93, 120, 125) , as occurs in SCZ patients. Also supporting the validity of the models toward the disorder is the observation that a number of these alterations, including deficits in PPI, were shown to be reversed by either acute or chronic treatment with several antipsychotic drugs in adult or adolescent animals [i.e., haloperidol, chlorpromazine, olanzapine, risperidone or clozapine, which constitute the primary pharmacological treatment for psychotic illness (78, 83-85, 89, 119, 126-132) ]. Given the central role of DA neurotransmission in SCZ, the findings on the exaggerated locomotor response to AMPH and other drugs that stimulate DAergic neurotransmission following MIA, and the effectiveness of antipsychotic treatments to reverse MIA effects, several studies investigated the effects of prenatal immune activation on this neurotransmitter system. One often used approach is the measurement of tissue DA content and its metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA, Figure 1B) . Prenatal poly I:C treatment at GD 15 induces enhanced release of DA from striatal explants in the adult offspring (83) . In addition, poly I:C treatment at GD 9 results in increases DA and DOPAC levels in the PFC and the globus pallidus (GP) and HVA in the nucleus accumbens (NAcc) and GP of adult mice (133) . Increases in DA are also found in the NAcc following poly I:C treatment at GD 9 (134) . Similarly, several injections of poly I:C (GD [12] [13] [14] [15] [16] [17] result in elevated levels of DOPAC and HVA in the adult STR (84) . Prenatal LPS treatment has been shown to have somewhat variable effects on DA. For example, daily administration of LPS throughout the entire pregnancy results in increased DA levels in the NAcc of adult animals (P 120, 170, or 400), but lower DA levels in younger animals (P 39) (119, 120) . Interestingly, a single LPS administration at GD 10, results in a decrease of DA in the dSTR (135) (136) (137) (138) (139) (140) . Decreased DA is also found in the NAcc, PFC, amygdala, hippocampus, and hypothalamus, accompanied by decreased levels of HVA in the NAcc and amygdala (P 120) (140) . Similarly, decreased DA levels in the NAcc at P 83 were found when escalating doses of LPS were administered daily from GD 15 until 19 (141) . Using MIA with TURP, we found increases in DA, DOPAC, and HVA in the NAcc, but not in the dorsal STR or the PFC of the male offspring in rats (142, 143) . Prenatal poly I:C treatment (GD 9) results in increased tyrosine hydroxylase (TH) immunoreactivity, the rate-limiting enzyme for the synthesis of DA, in the mesencephalon of embryonic mice at GD 13 and 17 (92) , as well as in the NAcc and SN of adult mice (P 120) (93). In the NAcc, TH immunoreactivity was decreased at P 35 (93) but increased at P 70 (90, 93) . DAT immunoreactivity is also found to be increased in the fetal mesencephalon (GD 17) (92), but decreased in the dSTR at P 35, as well as in the NAcc at GD 19 and P 35 (93) . Immunoreactivity of DA receptors, D1 and D2, is reduced in the adult mice's PFC (90, 144) and increased in the NAcc (for both D1 and D2) and dSTR (only D1) (93) . In contrast, Ozawa et al. reported that DA D2 receptor's binding is reduced in the STR of adult mice (84) . Most of these data are consistent with a scenario of increased synthesis of DA in the adult brain of MIA offspring, particularly in the meso-limbic, but not in the mesocortical pathway. The effects of prenatal LPS administration on these markers of DA neurotransmission are rather conflicting. Borrell et al. (118) found increased TH immunoreactivity in the NAcc and bed nucleus of the stria terminalis in adult rats whose mothers were treated with LPS on alternate days during the entire pregnancy. In contrast, Ling et al. reported in several studies that a single dose of LPS at GD 10 leads to a significant decrease in TH immunoreactivity, which was significant in the SN, at several postnatal ages (P 21, 120, 210, 420, 510) as well as in the VTA of post-weanling rats (135) (136) (137) (138) (139) (140) . As above, these results support the idea that models of bacterial MIA have different outcomes compared to those involving viral mimetics. Finally, prenatal TURP administration at GD 15 leads to an increase of TH levels in the NAcc, but not in other DA terminal areas such as the dorsal STR or the PFC, nor in the VTA or SN (124, 142, 143) . Overall, poly I:C and TURP induce molecular changes consistent with hyperactivity of mesolimbic DA neurotransmission, which may underlie the hyperactivity in response to AMPH and other behavioral alterations that can be corrected by administration of antipsychotics. A more causal role for elevated maternal cytokines in SCZ-related alterations has been established through the administration of exogenous cytokines to pregnant rats or mice. These manipulations have been shown to be sufficient to induce several molecular and behavioral effects in the offspring. For example, prenatal administration of IL-6 in mice (at GD 9, 5 mg, i.p.) results in impairments in PPI and other behaviors in the adult offspring, whereas a similar treatment with IFNg or TNFa does not affect the offspring (86) . Significantly, the effect of an influenza virus and poly I:C treatments on the fetal brain transcriptome overlapped to those of IL-6 administration in utero, supporting the idea that many effects of poly I:C are mediated by this cytokine (145) . Importantly, functional inhibition of poly I:C-induced IL-6 in pregnant mice prevented several of the behavioral effects of prenatal poly I:C in the offspring, including impaired PPI (86) . Also, the offspring of IL-6 "knock-out" mothers treated with poly I:C, do not present these alterations (86) . Similarly, knock-out of IL-6 receptor in the placental trophoblasts prevented several effects of prenatal poly I:C treatment (146), indicating a crucial role of this organ in mediating the effects of MIA. We also observed that co-treatment with an anti-IL-6 antibody during gestation and TURP prevented the development of a hyper-active DAergic system (143) . This prenatal treatment effectively rescued the exaggerated AMPH-induced hyperlocomotion and behavioral sensitization, elevated DA, and TH in the NAcc in the offspring of TURP-treated mothers (143) . IL-6 can, in turn, act in more than one way to affect neurodevelopment ( Figure 1C ). One such mechanism is hypoferremia, a reduction in maternal circulating non-heme iron, which characterizes the acute phase response and is triggered by all types of infection (147, 148) . Proper iron homeostasis is fundamental for healthy brain development, especially for the DAergic neurons (149) . Indeed, we demonstrated that maternal iron supplementation, which counteracts inflammation-induced hypoferremia, prevented the development of exacerbated responses to a single AMPH injection and enhanced behavioral sensitization following repeated exposure to this drug in the offspring (142) . Furthermore, maternal iron supplementation during MIA also reversed the increased levels of TH, DA and its metabolites in the NAcc found in the offspring of mothers treated with TURP (142) . Notably, iron levels in the placenta were reduced by MIA (but not in the fetal brain), which were rescued by maternal iron supplementation (142) , supporting a role for this organ in mediating the effects of MIA in the development of the brain. Another potential mediator of MIA, downstream of IL-6, is IL-17a ( Figure 1C) , since blocking the latter cytokine with anti-IL-17a antibodies prevented cortical malformations and the emergence of abnormal behaviors in adult MIA offspring, including impaired social interaction and increases marbleburying behavior (150, 151) . Meanwhile, overexpression of the anti-inflammatory cytokine IL-10 in maternal macrophages prevented the MIA-induced deficits in PPI, although in itself, elevated IL-10 also induced other behavioral alterations (91) . In addition to IL-6, the hormone leptin has also been implicated on the effects of MIA in the DAergic system. Leptin is the product of ob gene (152), a hormone that regulates food intake and energy expenditure (153) (154) (155) . Leptin is primarily produced by adipose tissue and secreted into the circulation, where levels correlate positively with body fat mass (156, 157) . Leptin has a multitude of physiological roles, including regulation of inflammatory processes (158, 159) . For example, leptin treatment induces pro-inflammatory cytokines, including TNFa, IL-1b, IL-6, and IFN-g (160) (161) (162) . Inflammatory stimuli (e.g., TNFa, IL-1b, LPS, and TURP) in turn increase leptin synthesis (163) (164) (165) (166) (167) . During the acute inflammatory response, leptin is involved in the induction of several sickness-type responses, such as anorexia and fever (168) (169) (170) (171) (172) (173) . Despite its clear involvement in several aspects of the inflammatory response to infection, the role of leptin in brain development has not yet been extensively studied. We demonstrated that neutralization of leptin during MIA was effective in curtailing several alterations induced by prenatal TURP, including the hype-sensitized locomotor response to AMPH, and increases in DA in the NAcc ( Figure 1C ) (143) . Intriguingly, leptin could affect the development of the dopaminergic system, as constitutive leptin mutant mice have impaired locomotor response to AMPH, and diminished DA release in the NAcc (174) . Leptin can also exert impairing effects or the control of cytokines expression in the placenta (175) . MIA alters the development of the dopaminergic system and many other neurotransmitter systems and brain regions (5, 6, 79, 80) . Maternal cytokines, particularly IL-6, are central in mediating these effects (5) . However, other neuroendocrine factors, such as the adipokine leptin, are potentially involved and deserve further investigation. Maternal infections and other environmental risk factors for SCZ and neurodevelopmental disorders may independently account for a few clinical cases since exposure to them does not always generate the disorder or are implicated in several psychiatric illnesses (6, 39, 79, 80, 176, 177) . In this regard, heterogeneity of response characterizes all known environmental risk factors for psychopathology, including the most overwhelming of traumas (176) . Such response heterogeneity is associated with pre-existing genetic (175) or epigenetic (i.e., chromatin modifications) differences (178) . This hypothesis implies that in any given population, individual predisposition is directly responsible for the vulnerability or resilience to the environmental causes of many psychiatric conditions (176) , including SCZ (39, 179). Regarding vulnerability, there is a significant interaction between maternal HSV-2 seropositivity and GRIN2B genetic variation (GRIN2B encodes for a NMDA glutamate receptor) (180) . Also, exposure to maternal infection has been reported to increase the risk of SCZ only in cases with a family history of psychiatric disorders (181, 182) . Animal models of MIA support this notion, as the effects of poly I:C are enhanced when they occur in mice mutant for genes linked to SCZ and other disorders (183) (184) (185) . Furthermore, interaction with other environmental risk factors, such as maternal diet, gut microbiota, or experiences of peripubertal trauma, can have a synergistic effect with maternal infection or prevent its detrimental effect (6, 79, 186) . Therefore, systematically generating translational models of the interaction between genetic and environmental (or environmental and environmental) risk factors for SCZ and other neurodevelopmental and psychiatric disorders appears to be the next step in understanding the etiology of mental illnesses. AA-V, BR, and EM-C wrote and edited the manuscript. Zika Virus and Birth Defects-Reviewing the Evidence for Causality Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and the Central Nervous System Cytokines and CNS development Prenatal Infection and Schizophrenia: A Review of Epidemiologic and Translational Studies Maternal immune activation: Implications for neuropsychiatric disorders Neurodevelopmental Resilience and Susceptibility to Maternal Immune Activation Maternal immune activation and abnormal brain development across CNS disorders Serological documentation of maternal influenza exposure and bipolar disorder in adult offspring Neurodevelopmental pathways in bipolar disorder 2017-Disease-Injury-Incidence-Prevalence-Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study Global economic burden of schizophrenia: a systematic review Schizophrenia: etiology and course Neurobiology of schizophrenia On the diagnostic and neurobiological origins of bipolar disorder Neuropsychiatric aspects of Parkinson disease psychopharmacology: Insights from circuit dynamics Behavioral and psychological symptoms in Alzheimer's dementia and vascular dementia Markers of Psychosis Risk in the General Population Genetics and genomics of psychiatric disease Effect of Chlorpromazine or Haloperidol on Formation of 3methoxytyramine and Normetanephrine in Mouse Brain Implications of normal brain development for the pathogenesis of schizophrenia Dopamine in schizophrenia: a review and reconceptualization Amphetamine redistributes dopamine from synaptic vesicles to the cytosol and promotes reverse transport Toxoplasma gondii as a risk factor for early-onset schizophrenia: analysis of filter paper blood samples obtained at birth Maternal exposure to herpes simplex virus and risk of psychosis among adult offspring Maternal infections and subsequent psychosis among offspring Prenatal exposure to maternal genital and reproductive infections and adult schizophrenia Association between prenatal exposure to bacterial infection and risk of schizophrenia Maternal cytokine levels during pregnancy and adult psychosis Systematic assessment of environmental risk factors for bipolar disorder: an umbrella review of systematic reviews and meta-analyses Maternal inflammation during pregnancy and offspring psychiatric symptoms in childhood: Timing and sex matter Elevated maternal interleukin-8 levels and risk of schizophrenia in adult offspring Prenatal maternal infection, neurodevelopment and adult schizophrenia: a systematic review of population-based studies Exposure to infection and brain development: cytokines in the pathogenesis of schizophrenia Elevated maternal cytokine levels at birth and risk for psychosis in adult offspring Prenatal maternal immune disruption and sex-dependent risk for psychoses Elevated maternal C-reactive protein and increased risk of schizophrenia in a national birth cohort Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring Maternal Immune Activation by Poly I:C as a preclinical Model for Neurodevelopmental Disorders: A focus on Autism and Schizophrenia Brain changes in a maternal immune activation model of neurodevelopmental brain disorders Prenatal Poly(I:C) Exposure and Other Developmental Immune Activation Models in Rodent Systems Neural circuit regulation of prepulse inhibition of startle in the rat: current knowledge and future challenges 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 Immune activation during pregnancy in mice leads to dopaminergic hyperfunction and cognitive impairment in the offspring: a neurodevelopmental animal model of schizophrenia Maternal immune activation leads to behavioral and pharmacological changes in the adult offspring Maternal immune activation alters fetal brain development through interleukin-6 Prenatal and postnatal maternal contributions in the infection model of schizophrenia The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology Evaluating Early Preventive Antipsychotic and Antidepressant Drug Treatment in an Infection-Based Neurodevelopmental Mouse Model of Schizophrenia Relative prenatal and postnatal maternal contributions to schizophrenia-related neurochemical dysfunction after in utero immune challenge Adult behavioral and pharmacological dysfunctions following disruption of the fetal brain balance between pro-inflammatory and IL-10-mediated antiinflammatory signaling Preliminary evidence for a modulation of fetal dopaminergic development by maternal immune activation during pregnancy A longitudinal examination of the neurodevelopmental impact of prenatal immune activation in mice reveals primary defects in dopaminergic development relevant to schizophrenia Maternal Vitamin D Prevents Abnormal Dopaminergic Development and Function in a Mouse Model of Prenatal Immune Activation Effects of prenatal immune activation on amphetamine-induced addictive behaviors: Contributions from animal models The risk for behavioural deficits is determined by the maternal immune response to prenatal immune challenge in a neurodevelopmental model Altered GSK3beta signaling in an infection-based mouse model of developmental neuropsychiatric disease Risperidone administered during asymptomatic period of adolescence prevents the emergence of brain structural pathology and behavioral abnormalities in an animal model of schizophrenia Abnormal trajectories of neurodevelopment and behavior following in utero insult in the rat Prospective Analysis of the Effects of Maternal Immune Activation on Rat Cytokines during Pregnancy and Behavior of the Male Offspring Relevant to Adolescent olanzapine sensitization is correlated with hippocampal stem cell proliferation in a maternal immune activation rat model of schizophrenia Late prenatal immune activation in mice leads to behavioral and neurochemical abnormalities relevant to the negative symptoms of schizophrenia Effects of early or late prenatal immune activation in mice on behavioral and neuroanatomical abnormalities relevant to schizophrenia in the adulthood Abnormal context-reward associations in an immune-mediated neurodevelopmental mouse model with relevance to schizophrenia Prenatal versus postnatal maternal factors in the development of infection-induced working memory impairments in mice Maternal exposure to bacterial endotoxin during pregnancy enhances amphetamine-induced locomotion and startle responses in adult rat offspring Effects of prenatal infection on prepulse inhibition in the rat depend on the nature of the infectious agent and the stage of pregnancy Endotoxin exposure in utero increases ethanol consumption in adult male offspring IL-1 receptor antagonist protects against placental and neurodevelopmental defects induced by maternal inflammation Developmental motor deficits induced by combined fetal exposure to lipopolysaccharide and early neonatal hypoxia/ischemia: a novel animal model for cerebral palsy in very premature infants Late N-acetylcysteine treatment prevents the deficits induced in the offspring of dams exposed to an immune stress during gestation Neurodevelopmental damage after prenatal infection: role of oxidative stress in the fetal brain Age-and gender-dependent impairments of neurobehaviors in mice whose mothers were exposed to lipopolysaccharide during pregnancy Normal aging of offspring mice of mothers with induced inflammation during pregnancy Specific neurodevelopmental damage in mice offspring following maternal inflammation during pregnancy Alterations in behavior in adult offspring mice following maternal inflammation during pregnancy Maternal immune activation leads to age-related behavioral and immunological changes in male rat offspring -the effect of antipsychotic drugs Prenatal immune challenge disrupts sensorimotor gating in adult rats. Implications for the etiopathogenesis of schizophrenia Neurobehavioral and Immunological Consequences of Prenatal Immune Activation in Rats. Influence of Antipsychotics Ontogeny of sensorimotor gating and immune impairment induced by prenatal immune challenge in rats: implications for the etiopathology of schizophrenia Protein metabolism after injury with turpentine: a rat model for clinical trauma Interleukin-6 is an afferent signal to the hypothalamo-pituitary-adrenal axis during local inflammation in mice Attenuated fever in rats during late pregnancy is linked to suppressed interleukin-6 production after localized inflammation with turpentine Alterations in cognitive function and behavioral response to amphetamine induced by prenatal inflammation are dependent on the stage of pregnancy Post-pubertal emergence of disrupted latent inhibition following prenatal immune activation Schizophrenia: more dopamine, more D2 receptors Paliperidone reverts Toll-like receptor 3 signaling pathway activation and cognitive deficits in a maternal immune activation mouse model of schizophrenia Chronic clozapine treatment improves prenatal infection-induced working memory deficits without influencing adult hippocampal neurogenesis Evaluating early preventive antipsychotic and antidepressant drug treatment in an infection-based neurodevelopmental mouse model of schizophrenia Maternal immune activation produces neonatal excitability defects in offspring hippocampal neurons from pregnant rats treated with poly I Effects of risperidone treatment in adolescence on hippocampal neurogenesis, parvalbumin expression, and vascularization following prenatal immune activation in rats Effects of risperidone and paliperidone pre-treatment on locomotor response following prenatal immune activation Prenatal immune activation leads to multiple changes in basal neurotransmitter levels in the adult brain: implications for brain disorders of neurodevelopmental origin such as schizophrenia Stress in puberty unmasks latent neuropathological consequences of prenatal immune activation in mice In utero bacterial endotoxin exposure causes loss of tyrosine hydroxylase neurons in the postnatal rat midbrain Combined toxicity of prenatal bacterial endotoxin exposure and postnatal 6-hydroxydopamine in the adult rat midbrain Rotenone potentiates dopamine neuron loss in animals exposed to lipopolysaccharide prenatally Progressive dopamine neuron loss following supra-nigral lipopolysaccharide (LPS) infusion into rats exposed to LPS prenatally Prenatal lipopolysaccharide does not accelerate progressive dopamine neuron loss in the rat as a result of normal aging Dopaminergic and serotoninergic deficiencies in young adult rats prenatally exposed to the bacterial lipopolysaccharide Prenatal immune challenge affects growth, behavior, and brain dopamine in offspring Prenatal inflammation-induced hypoferremia alters dopamine function in the adult offspring in rat: relevance for schizophrenia Leptin and interleukin-6 alter the function of mesolimbic dopamine neurons in a rodent model of prenatal inflammation Adult brain and behavioral pathological markers of prenatal immune challenge during early/middle and late fetal development in mice Effects of maternal immune activation on gene expression patterns in the fetal brain The placental interleukin-6 signaling controls fetal brain development and behavior Fever and survival: the role of serum iron Fever and reduced iron: their interaction as a host defense response to bacterial infection Iron status and neural functioning The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring Positional cloning of the mouse obese gene and its human homologue Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks Weight-reducing effects of the plasma protein encoded by the obese gene Effects of the obese gene product on body weight regulation in ob/ob mice Leptin Levels Reflect Body Lipid-Content in Mice -Evidence for Diet-Induced Resistance to Leptin Action Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects Obesity and its metabolic complications: the role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease Adipokines in inflammation and metabolic disease High-dose leptin activates human leukocytes via receptor expression on monocytes Ghrelin inhibits leptin-and activation-induced proinflammatory cytokine expression by human monocytes and T cells Leptin modulates beta cell expression of IL-1 receptor antagonist and release of IL-1beta in human islets Multiple cytokines and acute inflammation raise mouse leptin levels: potential role in inflammatory anorexia Endotoxin and cytokines induce expression of leptin, the ob gene product, in hamsters. A role for leptin in the anorexia of infection IL-1 beta mediates leptin induction during inflammation Elevated serum leptin concentrations induced by experimental acute inflammation Lipopolysaccharide-induced leptin release is neurally controlled Circulating leptin mediates lipopolysaccharide-induced anorexia and fever in rats Interleukin-6 and leptin mediate lipopolysaccharide-induced fever and sickness behavior Leptin actions on food intake and body temperature are mediated by IL-1 The effects of melanocortin agonists and antagonists on leptin-induced fever in rats Leptin receptor missense mutation in the fatty Zucker rat Leptin induces cyclooxygenase-2 via an interaction with interleukin-1beta in the rat brain Leptin regulation of the mesoaccumbens dopamine pathway Leptin and adiponectin stimulate the release of proinflammatory cytokines and prostaglandins from human placenta and maternal adipose tissue via nuclear factor-kappaB, peroxisomal proliferator-activated receptor-gamma and extracellularly regulated kinase 1/2 Gene-environment interactions in psychiatry: joining forces with neuroscience The developmental neurobiology of autism spectrum disorder Epigenetic Modifications in Schizophrenia and Related Disorders: Molecular Scars of Environmental Exposures and Source of Phenotypic Variability Recent advances in postmortem pathology and neurochemistry in schizophrenia Association of GRIN1 and GRIN2A-D with schizophrenia and genetic interaction with maternal herpes simplex virus-2 infection affecting disease risk Genome-wide study of association and interaction with maternal cytomegalovirus infection suggests new schizophrenia loci Evidence for an interaction between familial liability and prenatal exposure to infection in the causation of schizophrenia Prenatal interaction of mutant DISC1 and immune activation produces adult psychopathology Maternal immune activation during gestation interacts with Disc1 point mutation to exacerbate schizophrenia-related behaviors in mice Modulation of schizophrenia-related genes in the forebrain of adolescent and adult rats exposed to maternal immune activation Joint Effects of Exposure to Prenatal Infection and Peripubertal Psychological Trauma in Schizophrenia The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.Copyright © 2020 Aguilar-Valles, Rodrigue and Matta-Camacho. This is an openaccess article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.