Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 727
1.
Sci Rep ; 14(1): 10773, 2024 05 10.
Article En | MEDLINE | ID: mdl-38730262

The developing brain is vulnerable to maternal bacterial and viral infections which induce strong inflammatory responses in the mother that are mimicked in the offspring brain, resulting in irreversible neurodevelopmental defects, and associated cognitive and behavioural impairments. In contrast, infection during pregnancy and lactation with the immunoregulatory murine intestinal nematode, Heligmosomoides bakeri, upregulates expression of genes associated with long-term potentiation (LTP) of synaptic networks in the brain of neonatal uninfected offspring, and enhances spatial memory in uninfected juvenile offspring. As the hippocampus is involved in spatial navigation and sensitive to immune events during development, here we assessed hippocampal gene expression, LTP, and neuroimmunity in 3-week-old uninfected offspring born to H. bakeri infected mothers. Further, as maternal immunity shapes the developing immune system, we assessed the impact of maternal H. bakeri infection on the ability of offspring to resist direct infection. In response to maternal infection, we found an enhanced propensity to induce LTP at Schaffer collateral synapses, consistent with RNA-seq data indicating accelerated development of glutamatergic synapses in uninfected offspring, relative to those from uninfected mothers. Hippocampal RNA-seq analysis of offspring of infected mothers revealed increased expression of genes associated with neurogenesis, gliogenesis, and myelination. Furthermore, maternal infection improved resistance to direct infection of H. bakeri in offspring, correlated with transfer of parasite-specific IgG1 to their serum. Hippocampal immunohistochemistry and gene expression suggest Th2/Treg biased neuroimmunity in offspring, recapitulating peripheral immunoregulation of H. bakeri infected mothers. These findings indicate maternal H. bakeri infection during pregnancy and lactation alters peripheral and neural immunity in uninfected offspring, in a manner that accelerates neural maturation to promote hippocampal LTP, and upregulates the expression of genes associated with neurogenesis, gliogenesis, and myelination.


Hippocampus , Neuronal Plasticity , Animals , Female , Hippocampus/metabolism , Hippocampus/parasitology , Pregnancy , Mice , Nematode Infections/immunology , Nematode Infections/parasitology , Long-Term Potentiation , Prenatal Exposure Delayed Effects/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Male , Neuroimmunomodulation
2.
J Neuroinflammation ; 21(1): 118, 2024 May 07.
Article En | MEDLINE | ID: mdl-38715090

Maternal inflammation during gestation is associated with a later diagnosis of neurodevelopmental disorders including autism spectrum disorder (ASD). However, the specific impact of maternal immune activation (MIA) on placental and fetal brain development remains insufficiently understood. This study aimed to investigate the effects of MIA by analyzing placental and brain tissues obtained from the offspring of pregnant C57BL/6 dams exposed to polyinosinic: polycytidylic acid (poly I: C) on embryonic day 12.5. Cytokine and mRNA content in the placenta and brain tissues were assessed using multiplex cytokine assays and bulk-RNA sequencing on embryonic day 17.5. In the placenta, male MIA offspring exhibited higher levels of GM-CSF, IL-6, TNFα, and LT-α, but there were no differences in female MIA offspring. Furthermore, differentially expressed genes (DEG) in the placental tissues of MIA offspring were found to be enriched in processes related to synaptic vesicles and neuronal development. Placental mRNA from male and female MIA offspring were both enriched in synaptic and neuronal development terms, whereas females were also enriched for terms related to excitatory and inhibitory signaling. In the fetal brain of MIA offspring, increased levels of IL-28B and IL-25 were observed with male MIA offspring and increased levels of LT-α were observed in the female offspring. Notably, we identified few stable MIA fetal brain DEG, with no male specific difference whereas females had DEG related to immune cytokine signaling. Overall, these findings support the hypothesis that MIA contributes to the sex- specific abnormalities observed in ASD, possibly through altered neuron developed from exposure to inflammatory cytokines. Future research should aim to investigate how interactions between the placenta and fetal brain contribute to altered neuronal development in the context of MIA.


Brain , Cytokines , Mice, Inbred C57BL , Neurodevelopmental Disorders , Placenta , Prenatal Exposure Delayed Effects , Sex Characteristics , Female , Animals , Pregnancy , Male , Cytokines/metabolism , Cytokines/genetics , Mice , Brain/metabolism , Brain/immunology , Brain/embryology , Placenta/metabolism , Placenta/immunology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/immunology , Neurodevelopmental Disorders/metabolism , Poly I-C/toxicity , Transcriptome , Disease Models, Animal , Fetus/metabolism
3.
J Neuroimmunol ; 390: 578341, 2024 May 15.
Article En | MEDLINE | ID: mdl-38613873

Maternal allergic asthma (MAA) during pregnancy has been associated with increased risk of neurodevelopmental disorders in humans, and rodent studies have demonstrated that inducing a T helper-2-mediated allergic response during pregnancy leads to an offspring behavioral phenotype characterized by decreased social interaction and increased stereotypies. The interleukin (IL)-4 cytokine is hypothesized to mediate the neurobehavioral impact of MAA on offspring. Utilizing IL-4 knockout mice, this study assessed whether MAA without IL-4 signaling would still impart behavioral deficits. C57 and IL-4 knockout female mice were sensitized to ovalbumin, exposed to repeated MAA inductions, and their offspring performed social, cognitive, and motor tasks. Only C57 offspring of MAA dams displayed social and cognitive deficits, while IL-4 knockout mice showed altered motor activity compared with C57 mice. These findings highlight a key role for IL-4 signaling in MAA-induced behavioral deficits and more broadly in normal brain development.


Asthma , Interleukin-4 , Mice, Inbred C57BL , Mice, Knockout , Prenatal Exposure Delayed Effects , Animals , Female , Mice , Pregnancy , Asthma/immunology , Asthma/genetics , Interleukin-4/genetics , Interleukin-4/deficiency , Prenatal Exposure Delayed Effects/immunology , Behavior, Animal/physiology , Male , Ovalbumin/toxicity , Social Behavior , Motor Activity/physiology
4.
JAMA Psychiatry ; 81(5): 498-505, 2024 May 01.
Article En | MEDLINE | ID: mdl-38324324

Importance: Prenatal maternal inflammation has been associated with major depressive disorder in offspring in adulthood as well as with internalizing and externalizing symptoms in childhood; however, the association between prenatal inflammation and offspring depression in adolescence has yet to be examined. Objective: To determine whether maternal levels of inflammatory biomarkers during pregnancy are associated with depressive symptomatology in adolescent-aged offspring and to examine how gestational timing, offspring sex, and childhood psychiatric symptoms impact these associations. Design, Setting, and Participants: This was an observational study of a population-based birth cohort from the Child Health and Development Studies (CHDS), which recruited almost all mothers receiving obstetric care from the Kaiser Foundation Health Plan (KFHP) in Alameda County, California, between June 1959 and September 1966. Pregnancy data and blood sera were collected from mothers, and offspring psychiatric symptom data were collected in childhood (ages 9-11 years) and adolescence (ages 15-17 years). Mother-offspring dyads with available maternal prenatal inflammatory biomarkers during first and/or second trimesters and offspring depressive symptom data at adolescent follow-up were included. Data analyses took place between March 2020 and June 2023. Exposures: Levels of inflammatory biomarkers (interleukin 6 [IL-6], IL-8, IL-1 receptor antagonist [IL-1RA], and soluble tumor necrosis factor receptor-II) assayed from maternal sera in the first and second trimesters of pregnancy. Main Outcomes and Measures: Self-reported depressive symptoms at adolescent follow-up. Results: A total of 674 mothers (mean [SD] age, 28.1 [5.9] years) and their offspring (350 male and 325 female) were included in this study. Higher second trimester IL-6 was significantly associated with greater depressive symptoms in offspring during adolescence (b, 0.57; SE, 0.26); P = .03). Moderated mediation analyses showed that childhood externalizing symptoms significantly mediated the association between first trimester IL-6 and adolescent depressive symptoms in male offspring (b, 0.18; 95% CI, 0.02-0.47), while childhood internalizing symptoms mediated the association between second trimester IL-1RA and adolescent depressive symptoms in female offspring (b, 0.80; 95% CI, 0.19-1.75). Conclusions and Relevance: In this study, prenatal maternal inflammation was associated with depressive symptoms in adolescent-aged offspring. The findings of the study suggest that pathways to adolescent depressive symptomatology from prenatal risk factors may differ based on both the timing of exposure to prenatal inflammation and offspring sex.


Depression , Inflammation , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Female , Adolescent , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/blood , Child , Inflammation/blood , Male , Depression/blood , Depression/epidemiology , Adult , Sex Factors , Biomarkers/blood , California/epidemiology , Pregnancy Complications/blood , Pregnancy Complications/immunology , Pregnancy Complications/psychology
5.
Environ Sci Technol ; 57(36): 13419-13428, 2023 09 12.
Article En | MEDLINE | ID: mdl-37649345

Per- and polyfluoroalkyl substances (PFAS) have been identified as environmental contributors to adverse birth outcomes. One potential mechanistic pathway could be through PFAS-related inflammation and cytokine production. Here, we examined associations between a PFAS mixture and inflammatory biomarkers during early and late pregnancy from participants enrolled in the Atlanta African American Maternal-Child Cohort (N = 425). Serum concentrations of multiple PFAS were detected in >90% samples at 8-14 weeks gestation. Serum concentrations of interferon-γ (IFN-γ), interleukin 6 (IL-6), interleukin 10 (IL-10), tumor necrosis factor-α (TNF-α), and C-reactive protein (CRP) were measured at up to two time points (8-14 weeks and 24-30 weeks gestation). The effect of the PFAS mixture on each inflammatory biomarker was examined using quantile g-computation, Bayesian kernel machine regression (BKMR), Bayesian Weighted Sums (BWS), and weighted quantile sum (WQS) regression. Across all models, the PFAS mixture was associated with increased IFN-γ, IL-10, and TNF-α at both time points, with the strongest effects being observed at 24-30 weeks. Using quantile g-computation, increasing concentrations of a PFAS mixture were associated with a 29% (95% confidence interval = 18.0%, 40.7%) increase in TNF-α at 24-30 weeks. Similarly, using BWS, the PFAS mixture was associated with increased TNF-α at 24-30 weeks (summed effect = 0.29, 95% highest posterior density = 0.17, 0.41). The PFAS mixture was also positively associated with TNF-α at 24-30 weeks using BKMR [75th vs 50th percentile: 17.1% (95% credible interval = 7.7%, 27.4%)]. Meanwhile, PFOS was consistently the main drivers of overall mixture effect across four methods. Our findings indicated an increase in prenatal PFAS exposure is associated with an increase in multiple pro-inflammatory cytokines, potentially contributing to adverse pregnancy outcomes.


Biomarkers , Black or African American , Fluorocarbons , Pregnancy Complications , Prenatal Exposure Delayed Effects , Female , Humans , Pregnancy , Bayes Theorem , Biomarkers/blood , Fluorocarbons/blood , Interleukin-10 , Tumor Necrosis Factor-alpha , Pregnancy Outcome , Pregnancy Complications/blood , Pregnancy Complications/immunology , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/immunology
6.
Nature ; 610(7931): 327-334, 2022 10.
Article En | MEDLINE | ID: mdl-36171283

Recent studies suggested that microglia, the primary brain immune cells, can affect circuit connectivity and neuronal function1,2. Microglia infiltrate the neuroepithelium early in embryonic development and are maintained in the brain throughout adulthood3,4. Several maternal environmental factors-such as an aberrant microbiome, immune activation and poor nutrition-can influence prenatal brain development5,6. Nevertheless, it is unknown how changes in the prenatal environment instruct the developmental trajectory of infiltrating microglia, which in turn affect brain development and function. Here we show that, after maternal immune activation (MIA) in mice, microglia from the offspring have a long-lived decrease in immune reactivity (blunting) across the developmental trajectory. The blunted immune response was accompanied by changes in chromatin accessibility and reduced transcription factor occupancy of the open chromatin. Single-cell RNA-sequencing analysis revealed that MIA does not induce a distinct subpopulation but, rather, decreases the contribution to inflammatory microglia states. Prenatal replacement of microglia from MIA offspring with physiological infiltration of naive microglia ameliorated the immune blunting and restored a decrease in presynaptic vesicle release probability onto dopamine receptor type-two medium spiny neurons, indicating that aberrantly formed microglia due to an adverse prenatal environment affect the long-term microglia reactivity and proper striatal circuit development.


Inflammation , Microglia , Mothers , Neural Pathways , Prenatal Exposure Delayed Effects , Animals , Chromatin/genetics , Chromatin/metabolism , Female , Inflammation/immunology , Inflammation/pathology , Mice , Microglia/immunology , Microglia/pathology , Neostriatum/cytology , Neural Pathways/pathology , Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/immunology , RNA-Seq , Receptors, Dopamine/metabolism , Single-Cell Analysis , Transcription Factors/metabolism
7.
Proc Natl Acad Sci U S A ; 119(15): e2113310119, 2022 04 12.
Article En | MEDLINE | ID: mdl-35377817

Gestational maternal immune activation (MIA) in mice induces persistent brain microglial activation and a range of neuropathologies in the adult offspring. Although long-term phenotypes are well documented, how MIA in utero leads to persistent brain inflammation is not well understood. Here, we found that offspring of mothers treated with polyriboinosinic­polyribocytidylic acid [poly(I:C)] to induce MIA at gestational day 13 exhibit blood­brain barrier (BBB) dysfunction throughout life. Live MRI in utero revealed fetal BBB hyperpermeability 2 d after MIA. Decreased pericyte­endothelium coupling in cerebral blood vessels and increased microglial activation were found in fetal and 1- and 6-mo-old offspring brains. The long-lasting disruptions result from abnormal prenatal BBB formation, driven by increased proliferation of cyclooxygenase-2 (COX2; Ptgs2)-expressing microglia in fetal brain parenchyma and perivascular spaces. Targeted deletion of the Ptgs2 gene in fetal myeloid cells or treatment with the inhibitor celecoxib 24 h after immune activation prevented microglial proliferation and disruption of BBB formation and function, showing that prenatal COX2 activation is a causal pathway of MIA effects. Thus, gestational MIA disrupts fetal BBB formation, inducing persistent BBB dysfunction, which promotes microglial overactivation and behavioral alterations across the offspring life span. Taken together, the data suggest that gestational MIA disruption of BBB formation could be an etiological contributor to neuropsychiatric disorders.


Blood-Brain Barrier , Cyclooxygenase 2 , Encephalitis , Maternal-Fetal Exchange , Microglia , Prenatal Exposure Delayed Effects , Animals , Blood-Brain Barrier/abnormalities , Blood-Brain Barrier/physiopathology , Celecoxib/pharmacology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Encephalitis/immunology , Female , Gene Deletion , Maternal-Fetal Exchange/immunology , Mice , Microglia/enzymology , Poly I-C/immunology , Pregnancy , Prenatal Exposure Delayed Effects/immunology
8.
Trends Immunol ; 43(3): 230-244, 2022 03.
Article En | MEDLINE | ID: mdl-35131181

Exposure to heightened inflammation in pregnancy caused by infections or other inflammatory insults has been associated with the onset of neurodevelopmental and psychiatric disorders in children. Rodent models have provided unique insights into how this maternal immune activation (MIA) disrupts brain development. Here, we discuss the key immune factors involved, highlight recent advances in determining the molecular and cellular pathways of MIA, and review how the maternal immune system affects fetal development. We also examine the roles of microbiomes in shaping maternal immune function and the development of autism-like phenotypes. A comprehensive understanding of the gut bacteria-immune-neuro interaction in MIA is essential for developing diagnostic and therapeutic measures for high-risk pregnant women and identifying targets for treating inflammation-induced neurodevelopmental disorders.


Microbiota , Prenatal Exposure Delayed Effects , Animals , Disease Models, Animal , Female , Humans , Immune System , Inflammation/immunology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology
9.
J Neuroimmunol ; 364: 577813, 2022 03 15.
Article En | MEDLINE | ID: mdl-35093761

Maternal immune activation (MIA) with poly(I:C) is a preclinical paradigm for schizophrenia and autism research. Methodological variations, including poly(I:C) molecular weight, contribute to inconsistencies in behavioural and molecular outcomes. We established in Wistar rats that 4 mg/kg high molecular weight (HMW)-poly(I:C) on GD19 induces maternal sickness, smaller litters and maternal elevations of serum cytokines, including increases in monocyte chemoattractants. In adult offspring, we found that males have higher serum cytokines than females, and MIA did not alter peripheral cytokines in either sex. Our study will contribute to the effective use of the MIA model to elucidate the neurobiology of neurodevelopmental disorders.


Monocyte Chemoattractant Proteins/immunology , Neurodevelopmental Disorders/immunology , Poly I-C/toxicity , Pregnancy Complications, Infectious/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Cytokines/blood , Cytokines/immunology , Disease Models, Animal , Female , Male , Poly I-C/immunology , Pregnancy , Rats , Rats, Wistar
10.
Int J Mol Sci ; 23(2)2022 Jan 16.
Article En | MEDLINE | ID: mdl-35055151

Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by the early onset of communication and behavioral problems. ASD is highly heritable; however, environmental factors also play a considerable role in this disorder. A significant part of both syndromic and idiopathic autism cases could be attributed to disorders caused by mammalian target of rapamycin (mTOR)-dependent translation deregulation. This narrative review analyzes both bioinformatic and experimental evidence that connects mTOR signaling to the maternal autoantibody-related (MAR) autism spectrum and autoimmune neuropsychiatric disorders simultaneously. In addition, we reconstruct a network presenting the interactions between the mTOR signaling and eight MAR ASD genes coding for ASD-specific maternal autoantibody target proteins. The research discussed in this review demonstrates novel perspectives and validates the need for a subtyping of ASD on the grounds of pathogenic mechanisms. The utter necessity of designing ELISA-based test panels to identify all antibodies related to autism-like behavior is also considered.


Autism Spectrum Disorder/pathology , Prenatal Exposure Delayed Effects/pathology , TOR Serine-Threonine Kinases/metabolism , Autism Spectrum Disorder/classification , Autism Spectrum Disorder/immunology , Autism Spectrum Disorder/metabolism , Autoantibodies/metabolism , Child , Computational Biology/methods , Female , Humans , Pregnancy , Prenatal Exposure Delayed Effects/classification , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/metabolism , Signal Transduction
11.
Respir Physiol Neurobiol ; 296: 103800, 2022 02.
Article En | MEDLINE | ID: mdl-34626831

Infants born with neonatal opioid withdrawal syndrome (NOWS) can display abnormal cardiorespiratory patterns including tachypnea, tachycardia, and impaired ventilatory responses to hypoxia (HVR) and hypercapnia (HCVR). Chronic morphine exposure is associated with increased midbrain microglial expression. Using a rat model of pre- and post-natal morphine exposure, we assessed cardiorespiratory features of NOWS (resting tachycardia and tachypnea) including the attenuated HVR and HCVR and whether they are associated with increased brainstem microglia expression. Pregnant rats (dams) received twice-daily subcutaneous injections of morphine (5 mg/kg) during the third (last) week of pregnancy to simulate 3rd trimester in utero opioid exposure. Offspring then received once-daily subcutaneous injections of morphine (0.5 mg/kg) until postnatal (P) day P10 days of age to simulate postnatal morphine therapy. Cardiorespiratory responses were assessed 24 h later (P11 days) following spontaneous withdrawal. Compared to saline-treated pups, morphine-exposed offspring exhibited tachycardia and tachypnea as well as an attenuated HVR and HCVR. Microglial cell counts were increased in the nucleus tractus solitarius (nTS), dorsal motor nucleus of the vagus (DMNV) and nucleus ambiguous (NAamb), but not the retrapezoid nucleus (RTN) or the non-cardiorespriatory region, the cuneate nucleus (CN). These data suggest that the cardiorespiratory features and autonomic dysregulation in NOWS infants may be associated with altered microglial function in specific brainstem cardiorespiratory control regions.


Brain Stem , Infant, Newborn, Diseases , Microglia , Opioid-Related Disorders , Substance Withdrawal Syndrome , Tachycardia , Tachypnea , Animals , Animals, Newborn , Brain Stem/immunology , Brain Stem/physiopathology , Disease Models, Animal , Female , Humans , Hypercapnia/immunology , Hypercapnia/physiopathology , Hypoxia/immunology , Hypoxia/physiopathology , Infant, Newborn , Infant, Newborn, Diseases/etiology , Infant, Newborn, Diseases/immunology , Infant, Newborn, Diseases/physiopathology , Microglia/immunology , Opioid-Related Disorders/complications , Opioid-Related Disorders/immunology , Opioid-Related Disorders/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Rats , Substance Withdrawal Syndrome/complications , Substance Withdrawal Syndrome/immunology , Substance Withdrawal Syndrome/physiopathology , Tachycardia/etiology , Tachycardia/immunology , Tachycardia/physiopathology , Tachypnea/etiology , Tachypnea/immunology , Tachypnea/physiopathology
12.
Behav Brain Res ; 417: 113581, 2022 01 24.
Article En | MEDLINE | ID: mdl-34530042

BACKGROUND: The link between maternal immune activation (MIA) and the risk of developing schizophrenia (SCZ) later in life has been of major focus in recent years. This link could be bridged by activated inflammatory pathways and excessive cytokine release resulting in adverse effects on behavior, histology, and cytoarchitecture. The down-regulatory effects of immunomodulatory agents on the activated glial cells and their therapeutic effects on schizophrenic patients are consistent with this hypothesis. OBJECTIVE: We investigated whether treatment with the anti-inflammatory drug dimethyl fumarate (DMF) could rescue impacts of prenatal exposure to polyinosinic:polycytidylic acid [poly (I:C)]. METHODS: Pregnant dams were administered poly(I:C) at gestational day 9.5. Offspring born from these mothers were treated with DMF for fourteen consecutive days from postnatal day 80 and were assessed behaviorally before and after treatment. The brains were then stained with Cresyl Violet or Golgi-Cox. In addition to the estimation of stereological parameters, cytoarchitectural changes were also evaluated in the medial prefrontal cortex. RESULTS: MIA caused some abnormalities in behavior, as well as changes in the number of neurons and non-neurons. These alterations were also extended to pyramidal layer III neurons with a significant decrease in dendritic complexity and spine density which DMF treatment could prevent these changes. Furthermore, DMF treatment was also effective against abnormal exploratory and depression-related behavior, but not the changes in the number of cells. CONCLUSION: These findings support the idea of using anti-inflammatory agents as adjunctive therapy in patients with SCZ.


Dimethyl Fumarate/pharmacology , Poly I-C/pharmacology , Prefrontal Cortex/drug effects , Prenatal Exposure Delayed Effects/immunology , Schizophrenia/drug therapy , Animals , Brain/metabolism , Disease Models, Animal , Female , Male , Mice , Neurons/drug effects , Prefrontal Cortex/metabolism , Pregnancy , Schizophrenia/immunology
13.
J Immunol ; 208(1): 27-37, 2022 01 01.
Article En | MEDLINE | ID: mdl-34819390

Few studies have investigated immune cell ontogeny throughout the neonatal and early pediatric period, when there is often increased vulnerability to infections. In this study, we evaluated the dynamics of two critical T cell populations, T regulatory (Treg) cells and Th17 cells, over the first 36 wk of human life. First, we observed distinct CD4+ T cells phenotypes between cord blood and peripheral blood, collected within 12 h of birth, showing that cord blood is not a surrogate for newborn blood. Second, both Treg and Th17 cells expanded in a synchronous fashion over 36 wk of life. However, comparing infants exposed to HIV in utero, but remaining uninfected, with HIV-unexposed uninfected control infants, there was a lower frequency of peripheral blood Treg cells at birth, resulting in a delayed expansion, and then declining again at 36 wk. Focusing on birth events, we found that Treg cells coexpressing CCR4 and α4ß7 inversely correlated with plasma concentrations of CCL17 (the ligand for CCR4) and intestinal fatty acid binding protein, IL-7, and CCL20. This was in contrast with Th17 cells, which showed a positive association with these plasma analytes. Thus, despite the stereotypic expansion of both cell subsets over the first few months of life, there was a disruption in the balance of Th17 to Treg cells at birth likely being a result of gut damage and homing of newborn Treg cells from the blood circulation to the gut.


HIV Infections/immunology , HIV-1/physiology , Intestinal Mucosa/physiology , Prenatal Exposure Delayed Effects/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Cell Proliferation , Child, Preschool , Female , Homeostasis , Humans , Infant , Infant, Newborn , Lymphocyte Activation , Lymphopenia , Male , Pregnancy
14.
Am J Physiol Regul Integr Comp Physiol ; 322(2): R99-R111, 2022 02 01.
Article En | MEDLINE | ID: mdl-34874190

A prominent health issue nowadays is the COVID-19 pandemic, which poses acute risks to human health. However, the long-term health consequences are largely unknown and cannot be neglected. An especially vulnerable period for infection is pregnancy, when infections could have long-term health effect on the child. Evidence suggests that maternal immune activation (MIA) induced by either bacteria or viruses presents various effects on the offspring, leading to adverse phenotypes in many organ systems. This review compares the mechanisms of bacterial and viral MIA and the possible long-term outcomes for the offspring by summarizing the outcome in animal LPS and Poly I:C models. Both models are activated immune responses mediated by Toll-like receptors. The outcomes for MIA offspring include neurodevelopment, immune response, circulation, metabolism, and reproduction. Some of these changes continue to exist until later life. Besides different doses and batches of LPS and Poly I:C, the injection day, administration route, and also different animal species influence the outcomes. Here, we specifically aim to support colleagues when choosing their animal models for future studies.


COVID-19/complications , COVID-19/immunology , Lipopolysaccharides/toxicity , Poly I-C/toxicity , Prenatal Exposure Delayed Effects/immunology , SARS-CoV-2 , Bacterial Infections/immunology , Female , Humans , Pregnancy
15.
Immunity ; 55(1): 145-158.e7, 2022 01 11.
Article En | MEDLINE | ID: mdl-34879222

Children with autism spectrum disorders often display dysregulated immune responses and related gastrointestinal symptoms. However, the underlying mechanisms leading to the development of both phenotypes have not been elucidated. Here, we show that mouse offspring exhibiting autism-like phenotypes due to prenatal exposure to maternal inflammation were more susceptible to developing intestinal inflammation following challenges later in life. In contrast to its prenatal role in neurodevelopmental phenotypes, interleukin-17A (IL-17A) generated immune-primed phenotypes in offspring through changes in the maternal gut microbiota that led to postnatal alterations in the chromatin landscape of naive CD4+ T cells. The transfer of stool samples from pregnant mice with enhanced IL-17A responses into germ-free dams produced immune-primed phenotypes in offspring. Our study provides mechanistic insights into why children exposed to heightened inflammation in the womb might have an increased risk of developing inflammatory diseases in addition to neurodevelopmental disorders.


Autism Spectrum Disorder/immunology , CD4-Positive T-Lymphocytes/immunology , Chromatin/metabolism , Gastrointestinal Microbiome/immunology , Inflammation/immunology , Interleukin-17/metabolism , Intestines/immunology , Neurodevelopmental Disorders/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Autism Spectrum Disorder/microbiology , Child , Disease Models, Animal , Fecal Microbiota Transplantation , Female , Humans , Immunization , Inflammation/microbiology , Mice , Neurodevelopmental Disorders/microbiology , Pregnancy , Prenatal Exposure Delayed Effects/microbiology
16.
Biol Reprod ; 106(1): 200-212, 2022 01 13.
Article En | MEDLINE | ID: mdl-34668971

We evaluated the influence of the hyperglycemic intrauterine environment and postweaning consumption of a high-fat diet (HFD) on the glycemia, insulin, lipid, and immunological profile of rat offspring in adulthood. Female rats received citrate buffer (Control-C) or Streptozotocin (a beta cell-cytotoxic drug to induce diabetes-D) on postnatal day 5. In adulthood, these rats were mated to obtain female offspring, who were fed a standard diet (SD) or HFD from weaning to adulthood (n = 10 rats/group). OC/SD and OC/HFD represent female offspring of control mothers and received SD or HFD, respectively; OD/SD and OD/HFD represent female offspring of diabetic mothers and received SD or HFD, respectively. At adulthood, the oral glucose tolerance test (OGTT) was performed and, next, the rats were anesthetized and euthanized. Pancreas was collected and analyzed, and adipose tissue was weighted. Blood samples were collected to determine biochemical and immunological profiles. The food intake was lower in HFD-fed rats and visceral fat weight was increased in the OD/HFD group. OC/HFD, OD/SD, and OD/HFD groups presented glucose intolerance and lower insulin secretion during OGTT. An impaired pancreatic beta-cell function was shown in the adult offspring of diabetic rats, regardless of diet. Interleukin (IL)-6 and IL-10 concentrations were lower in the OD/HFD group and associated to a low-grade inflammatory condition. The fetal programming was responsible for impaired beta cell function in experimental animals. The association of maternal diabetes and postweaning HFD are responsible for greater glucose intolerance, impaired insulin secretion and immunological change.


Diabetes Mellitus, Experimental/complications , Diet, High-Fat , Hyperglycemia/complications , Pregnancy Complications , Prenatal Exposure Delayed Effects , Adiposity , Animals , Female , Glucose Intolerance , Insulin Resistance , Insulin-Secreting Cells/physiology , Pregnancy , Pregnancy in Diabetics/physiopathology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Sprague-Dawley , Weaning
17.
Cells ; 10(12)2021 12 11.
Article En | MEDLINE | ID: mdl-34944011

Both in utero exposure to maternal immune activation and cannabis use during adolescence have been associated with increased risk for the development of schizophrenia; however, whether these exposures exert synergistic effects on brain function is not known. In the present study, mild maternal immune activation (MIA) was elicited in mice with prenatal exposure to polyinosinic-polycytidylic acid (poly(I:C)), and ∆9-tetrahydrocannabinol (THC) was provided throughout adolescence in cereal (3 mg/kg/day for 5 days). Neither THC nor MIA pretreatments altered activity in assays used to characterize hyperdopaminergic states in adulthood: amphetamine hyperlocomotion and prepulse inhibition of the acoustic startle reflex. Adolescent THC treatment elicited deficits in spatial memory and enhanced spatial reversal learning in adult female mice in the Morris water maze, while exposure to MIA elicited female-specific deficits in fear extinction learning in adulthood. There were no effects in these assays in adult males, nor were there interactions between THC and MIA in adult females. While doses of poly(I:C) and THC were sufficient to elicit behavioral effects, particularly relating to cognitive performance in females, there was no evidence that adolescent THC exposure synergized with the risk imposed by MIA to worsen behavioral outcomes in adult mice of either sex.


Aging/physiology , Behavior, Animal/drug effects , Dronabinol/pharmacology , Prenatal Exposure Delayed Effects/immunology , Amphetamine , Animals , Conditioning, Classical , Extinction, Psychological/drug effects , Fear/drug effects , Female , Locomotion/drug effects , Male , Maze Learning/physiology , Mice, Inbred C57BL , Pregnancy , Prepulse Inhibition/drug effects , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Swimming
18.
Sci Rep ; 11(1): 23841, 2021 12 13.
Article En | MEDLINE | ID: mdl-34903784

Exposure to infection in utero predisposes towards psychiatric diseases such as autism, depression and schizophrenia in later life. The mechanisms involved are typically studied by administering mimetics of double-stranded (ds) virus or bacterial infection to pregnant rats or mice. The effect of single-stranded (ss) virus mimetics has been largely ignored, despite evidence linking prenatal ss virus exposure with psychiatric disease. Understanding the effects of gestational ss virus exposure has become even more important with recent events. In this study, in pregnant mice, we compare directly the effects, on the maternal blood, placenta and the embryonic brain, of maternal administration of ds-virus mimetic poly I:C (to activate Toll-like receptor 3, TLR3) and ss-virus mimetic resiquimod (to activate TLR7/8). We find that, 4 h after the administration, both poly I:C and resiquimod elevated the levels of IL-6, TNFα, and chemokines including CCL2 and CCL5, in maternal plasma. Both agents also increased placental mRNA levels of IL-6 and IL-10, but only resiquimod increased placental TNFα mRNA. In foetal brain, poly I:C produced no detectable immune-response-related increases, whereas pronounced increases in cytokine (e.g. Il-6, Tnfα) and chemokine (e.g. Ccl2, Ccl5) expression were observed with maternal resiquimod administration. The data show substantial differences between the effect of maternal exposure to a TLR7/8 activator as compared to a TLR3 activator. There are significant implications for future modelling of diseases where maternal ss virus exposure contributes to environmental disease risk in offspring.


Membrane Glycoproteins/immunology , Placenta/metabolism , Prenatal Exposure Delayed Effects/immunology , Schizophrenia/immunology , Toll-Like Receptor 3/immunology , Toll-Like Receptor 7/immunology , Animals , Chemokines/metabolism , Female , Imidazoles/toxicity , Interleukin-6/metabolism , Male , Membrane Glycoproteins/agonists , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Schizophrenia/etiology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 7/agonists , Tumor Necrosis Factor-alpha/metabolism
19.
Viruses ; 13(11)2021 11 18.
Article En | MEDLINE | ID: mdl-34835104

Zika virus (ZIKV) is a mosquito-borne flavivirus that became widely recognized due to the epidemic in Brazil in 2015. Since then, there has been nearly a 20-fold increase in the incidence of microcephaly and birth defects seen among women giving birth in Brazil, leading the Centers for Disease Control and Prevention (CDC) to officially declare a causal link between prenatal ZIKV infection and the serious brain abnormalities seen in affected infants. Here, we used a unique rat model of prenatal ZIKV infection to study three possible long-term outcomes of congenital ZIKV infection: (1) behavior, (2) cell proliferation, survival, and differentiation in the brain, and (3) immune responses later in life. Adult offspring that were prenatally infected with ZIKV exhibited motor deficits in a sex-specific manner, and failed to mount a normal interferon response to a viral immune challenge later in life. Despite undetectable levels of ZIKV in the brain and serum in these offspring at P2, P24, or P60, these results suggest that prenatal exposure to ZIKV results in lasting consequences that could significantly impact the health of the offspring. To help individuals already exposed to ZIKV, as well as be prepared for future outbreaks, we need to understand the full spectrum of neurological and immunological consequences that could arise following prenatal ZIKV infection.


Maternal Exposure/adverse effects , Nervous System Malformations/etiology , Neurodevelopmental Disorders/etiology , Pregnancy Complications, Infectious/immunology , Prenatal Exposure Delayed Effects/immunology , Zika Virus Infection , Animals , Animals, Newborn , Female , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Zika Virus Infection/immunology , Zika Virus Infection/physiopathology
...