1
|
Guma E, Chakravarty MM. Immune Alterations in the Intrauterine Environment Shape Offspring Brain Development in a Sex-Specific Manner. Biol Psychiatry 2025; 97:12-27. [PMID: 38679357 PMCID: PMC11511788 DOI: 10.1016/j.biopsych.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Exposure to immune dysregulation in utero or in early life has been shown to increase risk for neuropsychiatric illness. The sources of inflammation can be varied, including acute exposures due to maternal infection or acute stress, or persistent exposures due to chronic stress, obesity, malnutrition, or autoimmune diseases. These exposures may cause subtle alteration in brain development, structure, and function that can become progressively magnified across the lifespan, potentially increasing the likelihood of developing a neuropsychiatric conditions. There is some evidence that males are more susceptible to early-life inflammatory challenges than females. In this review, we discuss the various sources of in utero or early-life immune alteration and the known effects on fetal development with a sex-specific lens. To do so, we leveraged neuroimaging, behavioral, cellular, and neurochemical findings. Gaining clarity about how the intrauterine environment affects offspring development is critically important for informing preventive and early intervention measures that may buffer against the effects of these early-life risk factors.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, Maryland; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
2
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 DOI: 10.1016/j.neuropharm.2024.110084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Microglia play numerous important roles in brain development. From early embryonic stages through adolescence, these immune cells influence neuronal genesis and maturation, guide connectivity, and shape brain circuits. They also interact with other glial cells and structures, influencing the brain's supportive microenvironment. While this central role makes microglia essential, it means that early life perturbations to microglia can have widespread effects on brain development, potentially resulting in long-lasting behavioral impairments. Here, we will focus on the effects of early life psychosocial versus physiological stressors in rodent models. Psychosocial stress refers to perceived threats that lead to stress axes activation, including prenatal stress, or chronic postnatal stress, including maternal separation and resource scarcity. Physiological stress refers to physical threats, including maternal immune activation, postnatal infection, and traumatic brain injury. Differing sources of early life stress have varied impacts on microglia, and these effects are moderated by factors such as developmental age, brain region, and sex. Overall, these stressors appear to either 1) upregulate basal microglia numbers and activity throughout the lifespan, while possibly blunting their responsivity to subsequent stressors, or 2) shift the developmental curve of microglia, resulting in differential timing and function, impacting the critical periods they govern. Either could contribute to behavioral dysfunctions that occur after the resolution of early life stress. Exploring how different stressors impact microglia, as well as how multiple stressors interact to alter microglia's developmental functions, could deepen our understanding of how early life stress changes the brain's developmental trajectory. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Psychology, Ohio State University, Columbus, OH, USA.
| | - Kathryn M Lenz
- Department of Psychology, Ohio State University, Columbus, OH, USA; Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Ohio State University, Columbus, OH, USA
| |
Collapse
|
3
|
Chen C, Zhu S, Fu T, Chen Y, Chen D. The protective effects of Ferrostatin-1 against inflammation-induced preterm birth and fetal brain injury. J Reprod Immunol 2024; 164:104260. [PMID: 38761507 DOI: 10.1016/j.jri.2024.104260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/19/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
INTRODUCTION Recent studies have suggested the involvement of ferroptosis in preterm birth. Despite compelling evidence, the underlying mechanism remains unknown. This investigation aimed to determine the therapeutic effects of Ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, in preterm birth and fetal brain injury. METHODS Human placenta samples and clinical data of participants were collected to ascertain whether placental ferroptosis was associated with preterm birth. Lipopolysaccharide (LPS)-induced preterm birth mouse model was used to examine the protective effects of Fer-1 on preterm birth. Fetal brain tissues and offspring mice at 5 and 8 weeks were studied to determine the effects of Fer-1 on the cognitive function of offspring. RESULTS We examined the mechanism of spontaneous preterm birth and discovered that placental ferroptosis was associated with preterm birth. Fer-1 inhibited preterm birth by ameliorating placental ferroptosis and maternal inflammation, thus improving LPS-induced intrauterine inflammation to maintain pregnancy. Antenatal administration of Fer-1 prevented LPS-induced fetal brain damage in the acute phase and improved long-term neurodevelopmental impairments by improving placental neuroendocrine signaling and maintaining placental function. CONCLUSION Fer-1 inhibited preterm birth and fetal brain injury by inhibiting maternal inflammation and improving placental function. Our findings provide a novel therapeutic strategy for preterm birth.
Collapse
Affiliation(s)
- Chaolu Chen
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Shuaiying Zhu
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Tiantian Fu
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Yanmin Chen
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Danqing Chen
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China.
| |
Collapse
|
4
|
Terada S, Isumi A, Doi S, Tani Y, Fujiwara T. Association between gestational weight gain and behavioral problems of the offspring aged 6-7 years: A population-based study in Japan. Int J Gynaecol Obstet 2024; 166:804-811. [PMID: 38340033 DOI: 10.1002/ijgo.15410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/06/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVE To identify the optimal gestational weight gain (GWG) thresholds for behavioral problems and prosocial behavior in 6- to 7-year-old children. METHODS A retrospective cohort study was conducted using data from the Adachi Child Health Impact of Living Difficulty study, including all first-graders in public schools in Adachi, Tokyo, in 2017, 2019, and 2021 (n = 11 048, response rate = 80.1%). GWG was based on clinical records in the Mother and Child Health Handbook. Total difficulties and prosocial behavior were assessed using the Strength and Difficulties Questionnaire. Logistic regression models with restricted cubic splines, and quintile categories were employed to examine the association of GWG with the clinical range of total difficulties and prosocial behavior, controlling for covariates. RESULTS The association between GWG and total difficulties exhibited a reverse J-shaped pattern, with low GWG, but not high GWG, increasing the risk (odds ratio [OR] 1.20, 95% confidence interval [CI] 1.01-1.42, P = 0.039 for GWG < 7 kg; OR 1.03, 95% CI 0.85-1.24, P = 0.786 for GWG > 14 kg), referencing the median (10 kg). High GWG was associated with a lower risk of problems in prosocial behavior (OR = 0.77, 95% CI 0.62-0.95, P = 0.017 for GWG > 14 kg; OR 1.06, 95% CI 0.88-1.27, P = 0.532 for GWG <7 kg). CONCLUSION GWG less than 7 kg may increase the risk of total difficulties, whereas GWG over 14 kg may serve as a protective factor for prosocial behavior in 6- to 7-year-old children.
Collapse
Affiliation(s)
- Shuhei Terada
- Department of Global Health Promotion, Tokyo Medical and Dental University, Tokyo, Japan
| | - Aya Isumi
- Department of Health Policy, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satomi Doi
- Department of Health Policy, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yukako Tani
- Department of Global Health Promotion, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeo Fujiwara
- Department of Global Health Promotion, Tokyo Medical and Dental University, Tokyo, Japan
- Department of International Health, Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Davis AB, Lloyd KR, Bollinger JL, Wohleb ES, Reyes TM. Adolescent high fat diet alters the transcriptional response of microglia in the prefrontal cortex in response to stressors in both male and female mice. Stress 2024; 27:2365864. [PMID: 38912878 PMCID: PMC11228993 DOI: 10.1080/10253890.2024.2365864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Both obesity and high fat diets (HFD) have been associated with an increase in inflammatory gene expression within the brain. Microglia play an important role in early cortical development and may be responsive to HFD, particularly during sensitive windows, such as adolescence. We hypothesized that HFD during adolescence would increase proinflammatory gene expression in microglia at baseline and potentiate the microglial stress response. Two stressors were examined, a physiological stressor [lipopolysaccharide (LPS), IP] and a psychological stressor [15 min restraint (RST)]. From 3 to 7 weeks of age, male and female mice were fed standard control diet (SC, 20% energy from fat) or HFD (60% energy from fat). On P49, 1 h before sacrifice, mice were randomly assigned to either stressor exposure or control conditions. Microglia from the frontal cortex were enriched using a Percoll density gradient and isolated via fluorescence-activated cell sorting (FACS), followed by RNA expression analysis of 30 genes (27 target genes, three housekeeping genes) using Fluidigm, a medium throughput qPCR platform. We found that adolescent HFD induced sex-specific transcriptional response in cortical microglia, both at baseline and in response to a stressor. Contrary to our hypothesis, adolescent HFD did not potentiate the transcriptional response to stressors in males, but rather in some cases, resulted in a blunted or absent response to the stressor. This was most apparent in males treated with LPS. However, in females, potentiation of the LPS response was observed for select proinflammatory genes, including Tnfa and Socs3. Further, HFD increased the expression of Itgam, Ikbkb, and Apoe in cortical microglia of both sexes, while adrenergic receptor expression (Adrb1 and Adra2a) was changed in response to stressor exposure with no effect of diet. These data identify classes of genes that are uniquely affected by adolescent exposure to HFD and different stressor modalities in males and females.
Collapse
Affiliation(s)
- Alyshia B Davis
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Kelsey R Lloyd
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Teresa M Reyes
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
6
|
Abelaira HM, de Moura AB, Cardoso MM, de Pieri E, Abel JS, Luiz GP, Sombrio EM, Borghezan LA, Anastácio RS, Cruz LA, de Souza TG, Meab C, Lima IR, da Costa C, Dal Bó AG, Pcl S, Machado-de-Ávila RA. Sertraline associated with gold nanoparticles reduce cellular toxicity and induce sex-specific responses in behavior and neuroinflammation biomarkers in a mouse model of anxiety. Pharmacol Biochem Behav 2023; 233:173661. [PMID: 37879445 DOI: 10.1016/j.pbb.2023.173661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
This study aimed to evaluate the effects of sertraline associated with gold nanoparticles (AuNPs) in vitro cell viability and in vivo behavior and inflammatory biomarkers in a mouse model of anxiety. Sertraline associated with AuNPs were synthesized and characterized. For the in vitro study, NIH3T3 and HT-22 cells were treated with different doses of sertraline, AuNPs, and sertraline + AuNPs and their viability was evaluated using the MTT assay. For the in vivo study, pregnant Swiss mice were administered a single dose of lipopolysaccharide (LPS) on the ninth day of gestation. The female and male offspring were divided into five treatment groups on PND 60 and administered chronic treatment for 28 days. The animals were subjected to behavioral testing and were subsequently euthanized. Their brains were collected and analyzed for inflammatory biomarkers. Sertraline associated with AuNPs exhibited significant changes in surface characteristics and increased diameters. Different doses of sertraline + AuNPs showed higher cell viability in NIH3T3 and HT-22 cells compared with sertraline alone. The offspring of LPS-treated dams exhibited anxiety-like behavior and neuroinflammatory biomarker changes during adulthood, which were ameliorated via sertraline + AuNPs treatment. The treatment response was sex-dependent and brain region-specific. These results suggest that AuNPs, which demonstrate potential to bind to other molecules, low toxicity, and reduced inflammation, can be synergistically used with sertraline to improve drug efficacy and safety by decreasing neuroinflammation and sertraline toxicity.
Collapse
Affiliation(s)
- H M Abelaira
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil.
| | - A B de Moura
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - M M Cardoso
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - E de Pieri
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - J S Abel
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - G P Luiz
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - E M Sombrio
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - L A Borghezan
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - R S Anastácio
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - L A Cruz
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - T G de Souza
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - Corrêa Meab
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - I R Lima
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - C da Costa
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - A G Dal Bó
- Laboratory of Advanced Polymer Processing, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - Silveira Pcl
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - R A Machado-de-Ávila
- Laboratory of Pathophysiology Experimental, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| |
Collapse
|
7
|
Goodman CV, Green R, DaCosta A, Flora D, Lanphear B, Till C. Sex difference of pre- and post-natal exposure to six developmental neurotoxicants on intellectual abilities: a systematic review and meta-analysis of human studies. Environ Health 2023; 22:80. [PMID: 37978510 PMCID: PMC10655280 DOI: 10.1186/s12940-023-01029-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Early life exposure to lead, mercury, polychlorinated biphenyls (PCBs), polybromide diphenyl ethers (PBDEs), organophosphate pesticides (OPPs), and phthalates have been associated with lowered IQ in children. In some studies, these neurotoxicants impact males and females differently. We aimed to examine the sex-specific effects of exposure to developmental neurotoxicants on intelligence (IQ) in a systematic review and meta-analysis. METHOD We screened abstracts published in PsychINFO and PubMed before December 31st, 2021, for empirical studies of six neurotoxicants (lead, mercury, PCBs, PBDEs, OPPs, and phthalates) that (1) used an individualized biomarker; (2) measured exposure during the prenatal period or before age six; and (3) provided effect estimates on general, nonverbal, and/or verbal IQ by sex. We assessed each study for risk of bias and evaluated the certainty of the evidence using Navigation Guide. We performed separate random effect meta-analyses by sex and timing of exposure with subgroup analyses by neurotoxicant. RESULTS Fifty-one studies were included in the systematic review and 20 in the meta-analysis. Prenatal exposure to developmental neurotoxicants was associated with decreased general and nonverbal IQ in males, especially for lead. No significant effects were found for verbal IQ, or postnatal lead exposure and general IQ. Due to the limited number of studies, we were unable to analyze postnatal effects of any of the other neurotoxicants. CONCLUSION During fetal development, males may be more vulnerable than females to general and nonverbal intellectual deficits from neurotoxic exposures, especially from lead. More research is needed to examine the nuanced sex-specific effects found for postnatal exposure to toxic chemicals.
Collapse
Affiliation(s)
- Carly V Goodman
- Faculty of Health, York University, Toronto, M3J 1P3, ON, Canada.
| | - Rivka Green
- Faculty of Health, York University, Toronto, M3J 1P3, ON, Canada
| | - Allya DaCosta
- Faculty of Health, York University, Toronto, M3J 1P3, ON, Canada
| | - David Flora
- Faculty of Health, York University, Toronto, M3J 1P3, ON, Canada
| | - Bruce Lanphear
- Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Christine Till
- Faculty of Health, York University, Toronto, M3J 1P3, ON, Canada
| |
Collapse
|
8
|
Ishida E, Furusho H, Renn TY, Shiba F, Chang HM, Oue H, Terayama R, Ago Y, Tsuga K, Miyauchi M. Mouse maternal odontogenic infection with Porphyromonas gingivalis induces cognitive decline in offspring. Front Pediatr 2023; 11:1203894. [PMID: 37635786 PMCID: PMC10450928 DOI: 10.3389/fped.2023.1203894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Porphyromonas gingivalis (P. gingivalis), a major periodontal pathogen, causes intrauterine infection/inflammation. Offspring exposed to intrauterine infection/inflammation have an increased risk of neurological disorders, regardless of gestational age. However, the relationship between maternal periodontitis and offspring functional/histological changes in the brain has not yet been elucidated. Methods In this study, we used a gestational mouse model to investigate the effects of maternal odontogenic infection of P. gingivalis on offspring behavior and brain tissue. Results The step-through passive avoidance test showed that the latency of the acquisition trial was significantly shorter in the P. gingivalis group (p < 0.05), but no difference in spontaneous motor/exploratory parameters by open-field test. P. gingivalis was diffusely distributed throughout the brain, especially in the hippocampus. In the hippocampus and amygdala, the numbers of neuron cells and cyclic adenosine monophosphate response element binding protein-positive cells were significantly reduced (p < 0.05), whereas the number of ionized calcium binding adapter protein 1-positive microglia was significantly increased (p < 0.05). In the hippocampus, the number of glial fibrillary acidic protein-positive astrocytes was also significantly increased (p < 0.05). Discussion The offspring of P. gingivalis-infected mothers have reduced cognitive function. Neurodegeneration/neuroinflammation in the hippocampus and amygdala may be caused by P. gingivalis infection, which is maternally transmitted. The importance of eliminating maternal P. gingivalis-odontogenic infection before or during gestation in maintenance healthy brain function in offspring should be addressed in near future.
Collapse
Affiliation(s)
- Eri Ishida
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hisako Furusho
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ting-Yi Renn
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Fumie Shiba
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hung-Ming Chang
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hiroshi Oue
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ryuji Terayama
- Department of Maxillofacial Anatomy and Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiro Tsuga
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mutsumi Miyauchi
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
9
|
Shepilov D, Osadchenko I, Kovalenko T, Yamada C, Chereshynska A, Smozhanyk K, Ostrovska G, Groppa S, Movila A, Skibo G. Maternal antibiotic administration during gestation can affect the memory and brain structure in mouse offspring. Front Cell Neurosci 2023; 17:1176676. [PMID: 37234915 PMCID: PMC10206017 DOI: 10.3389/fncel.2023.1176676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/18/2023] [Indexed: 05/28/2023] Open
Abstract
Maternal antibiotics administration (MAA) is among the widely used therapeutic approaches in pregnancy. Although published evidence demonstrates that infants exposed to antibiotics immediately after birth have altered recognition memory responses at one month of age, very little is known about in utero effects of antibiotics on the neuronal function and behavior of children after birth. Therefore, this study aimed to evaluate the impact of MAA at different periods of pregnancy on memory decline and brain structural alterations in young mouse offspring after their first month of life. To study the effects of MAA on 4-week-old offspring, pregnant C57BL/6J mouse dams (2-3-month-old; n = 4/group) were exposed to a cocktail of amoxicillin (205 mg/kg/day) and azithromycin (51 mg/kg/day) in sterile drinking water (daily/1 week) during either the 2nd or 3rd week of pregnancy and stopped after delivery. A control group of pregnant dams was exposed to sterile drinking water alone during all three weeks of pregnancy. Then, the 4-week-old offspring mice were first evaluated for behavioral changes. Using the Morris water maze assay, we revealed that exposure of pregnant mice to antibiotics at the 2nd and 3rd weeks of pregnancy significantly altered spatial reference memory and learning skills in their offspring compared to those delivered from the control group of dams. In contrast, no significant difference in long-term associative memory was detected between offspring groups using the novel object recognition test. Then, we histologically evaluated brain samples from the same offspring individuals using conventional immunofluorescence and electron microscopy assays. To our knowledge, we observed a reduction in the density of the hippocampal CA1 pyramidal neurons and hypomyelination in the corpus callosum in groups of mice in utero exposed to antibiotics at the 2nd and 3rd weeks of gestation. In addition, offspring exposed to antibiotics at the 2nd or 3rd week of gestation demonstrated a decreased astrocyte cell surface area and astrocyte territories or depletion of neurogenesis in the dentate gyrus and hippocampal synaptic loss, respectively. Altogether, this study shows that MAA at different times of pregnancy can pathologically alter cognitive behavior and brain development in offspring at an early age after weaning.
Collapse
Affiliation(s)
- Dmytro Shepilov
- Department of Cytology, Bogomoletz Institute of Physiology, NAS of Ukraine, Kyiv, Ukraine
| | - Iryna Osadchenko
- Department of Cytology, Bogomoletz Institute of Physiology, NAS of Ukraine, Kyiv, Ukraine
| | - Tetiana Kovalenko
- Department of Cytology, Bogomoletz Institute of Physiology, NAS of Ukraine, Kyiv, Ukraine
| | - Chiaki Yamada
- Department of Biomedical Sciences and Comprehensive Care, School of Dentistry, Indiana University, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anastasiia Chereshynska
- Department of Biomedical Sciences and Comprehensive Care, School of Dentistry, Indiana University, Indianapolis, IN, United States
| | - Kateryna Smozhanyk
- Department of Cytology, Bogomoletz Institute of Physiology, NAS of Ukraine, Kyiv, Ukraine
| | - Galyna Ostrovska
- Department of Cytology, Histology, and Reproductive Medicine, Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Stanislav Groppa
- Department of Neurology, Institute of Emergency Medicine, Chisinau, Moldova
- Department of Neurology, State University of Medicine and Pharmacy “Nicolae Testemiţanu”, Chisinau, Moldova
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, School of Dentistry, Indiana University, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Galyna Skibo
- Department of Cytology, Bogomoletz Institute of Physiology, NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
10
|
Zhang Y, Lu D, Guo VY, Wang Y, Qiu S, Zhang J, Zhang Y, Chen W, Wang B, Yang W. Association between maternal polycystic ovary syndrome and attention-deficit/hyperactivity disorder in offspring aged 3-6 years: A Chinese population-based study. Front Public Health 2023; 10:1032315. [PMID: 36699874 PMCID: PMC9868860 DOI: 10.3389/fpubh.2022.1032315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Background Maternal polycystic ovary syndrome (PCOS) may increase the risk of attention-deficit/hyperactivity disorder (ADHD) in offspring; however, their association remains unexplored in Asian populations. Hence, this study aimed to investigate the association between maternal PCOS and ADHD in offspring aged 3-6 years and whether it differed by offspring sex. Methods This was a district-wide population-based study of 87,081 preschoolers from 234 kindergartens in Longhua District, Shenzhen, China. The parents were invited to complete a self-administrated questionnaire covering information on socio-demographics, maternal disease history, and child behavior. ADHD symptoms were measured with the parent-rating 26-item Swanson, Nolan, and Pelham Rating Scale (SNAP-IV). Logistic regression was performed to examine the associations between maternal PCOS and ADHD symptoms in offspring. Results The response rate was 80% and 63,390 mother-child pairs were included. Of the mothers, 1,667 (2.6%) reported PCOS diagnoses. The mean age of children at ADHD assessment was 4.86 ± 0.84[SD] years, and 53.6% were boys. Children with maternal PCOS had a higher risk of developing ADHD symptoms than other children (12.0 vs. 9.4%, adjusted odds ratio [OR] = 1.32, 95% CI: 1.13-1.54). The risk estimate was significant in boys (adjusted OR = 1.38, 95% CI: 1.14-1.66) but not in girls (adjusted OR = 1.23, 95% CI: 0.94-1.57, P for interaction = 0.391). Treatment of PCOS tended to be associated with a lower risk of ADHD symptoms than untreated PCOS albeit risk confidence intervals were overlapped (treated: adjusted OR = 1.28, 95% CI: 1.06-1.54 vs. untreated: adjusted OR = 1.14, 95% CI: 1.08-1.83). Conclusion Maternal PCOS increases the risk of developing ADHD in offspring, especially boys. Further studies are warranted to confirm our findings, and early neurodevelopmental screening may be needed in children born to mothers with PCOS.
Collapse
Affiliation(s)
- Yuying Zhang
- Department of Child Healthcare, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Dali Lu
- Department of Child Healthcare, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Vivian Yawei Guo
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuqing Wang
- Health Management Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Shuangyan Qiu
- Department of Child Healthcare, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Jingyu Zhang
- Department of Child Healthcare, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Yan Zhang
- Department of Child Healthcare, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Weiqing Chen
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Baoping Wang
- Department of Infertility and Reproductive Medicine, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China,Baoping Wang ✉
| | - Weikang Yang
- Department of Child Healthcare, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China,*Correspondence: Weikang Yang ✉
| |
Collapse
|
11
|
Breach MR, Lenz KM. Sex Differences in Neurodevelopmental Disorders: A Key Role for the Immune System. Curr Top Behav Neurosci 2023; 62:165-206. [PMID: 35435643 PMCID: PMC10286778 DOI: 10.1007/7854_2022_308] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sex differences are prominent defining features of neurodevelopmental disorders. Understanding the sex biases in these disorders can shed light on mechanisms leading to relative risk and resilience for the disorders, as well as more broadly advance our understanding of how sex differences may relate to brain development. The prevalence of neurodevelopmental disorders is increasing, and the two most common neurodevelopmental disorders, Autism Spectrum Disorder (ASD) and Attention-Deficit/Hyperactivity Disorder (ADHD) exhibit male-biases in prevalence rates and sex differences in symptomology. While the causes of neurodevelopmental disorders and their sex differences remain to be fully understood, increasing evidence suggests that the immune system plays a critical role in shaping development. In this chapter we discuss sex differences in prevalence and symptomology of ASD and ADHD, review sexual differentiation and immune regulation of neurodevelopment, and discuss findings from human and rodent studies of immune dysregulation and perinatal immune perturbation as they relate to potential mechanisms underlying neurodevelopmental disorders. This chapter will give an overview of how understanding sex differences in neuroimmune function in the context of neurodevelopmental disorders could lend insight into their etiologies and better treatment strategies.
Collapse
Affiliation(s)
- Michaela R Breach
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
12
|
Intrauterine Inflammation Leads to Select Sex- and Age-Specific Behavior and Molecular Differences in Mice. Int J Mol Sci 2022; 24:ijms24010032. [PMID: 36613475 PMCID: PMC9819857 DOI: 10.3390/ijms24010032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Sex-specific differences in behavior have been observed in anxiety and learning in children exposed to prenatal inflammation; however, whether these behaviors manifest differently by age is unknown. This study assesses possible behavioral changes due to in utero inflammation as a function of age in neonatal, juvenile, and adult animals and presents potential molecular targets for observed differences. CD-1 timed pregnant dams were injected in utero with lipopolysaccharide (LPS, 50 μg/animal) or saline at embryonic day 15. No differences in stress responses were measured by neonatal ultrasonic vocalizations between LPS- and saline-exposed groups of either sex. By contrast, prenatal inflammation caused a male-specific increase in anxiety in mature but not juvenile animals. Juvenile LPS-exposed females had decreased movement in open field testing that was not present in adult animals. We additionally observed improved memory retrieval after in utero LPS in the juvenile animals of both sexes, which in males may be related to a perseverative phenotype. However, there was an impairment of long-term memory in only adult LPS-exposed females. Finally, gene expression analyses revealed that LPS induced sex-specific changes in genes involved in hippocampal neurogenesis. In conclusion, intrauterine inflammation has age- and sex-specific effects on anxiety and learning that may correlate to sex-specific disruption of gene expression associated with neurogenesis in the hippocampus.
Collapse
|
13
|
Vilotić A, Nacka-Aleksić M, Pirković A, Bojić-Trbojević Ž, Dekanski D, Jovanović Krivokuća M. IL-6 and IL-8: An Overview of Their Roles in Healthy and Pathological Pregnancies. Int J Mol Sci 2022; 23:ijms232314574. [PMID: 36498901 PMCID: PMC9738067 DOI: 10.3390/ijms232314574] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Interleukin-6 (IL-6) is an acknowledged inflammatory cytokine with a pleiotropic action, mediating innate and adaptive immunity and multiple physiological processes, including protective and regenerative ones. IL-8 is a pro-inflammatory CXC chemokine with a primary function in attracting and activating neutrophils, but also implicated in a variety of other cellular processes. These two ILs are abundantly expressed at the feto-maternal interface over the course of a pregnancy and have been shown to participate in numerous pregnancy-related events. In this review, we summarize the literature data regarding their role in healthy and pathological pregnancies. The general information related to IL-6 and IL-8 functions is followed by an overview of their overall expression in cycling endometrium and at the feto-maternal interface. Further, we provide an overview of their involvement in pregnancy establishment and parturition. Finally, the implication of IL-6 and IL-8 in pregnancy-associated pathological conditions, such as pregnancy loss, preeclampsia, gestational diabetes mellitus and infection/inflammation is discussed.
Collapse
|
14
|
Infection of the murine placenta by Listeria monocytogenes induces sex-specific responses in the fetal brain. Pediatr Res 2022; 93:1566-1573. [PMID: 36127406 DOI: 10.1038/s41390-022-02307-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/06/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Epidemiological data indicate that prenatal infection is associated with an increased risk of several neurodevelopmental disorders in the progeny. These disorders display sex differences in presentation. The role of the placenta in the sex-specificity of infection-induced neurodevelopmental abnormalities is not well-defined. We used an imaging-based animal model of the bacterial pathogen Listeria monocytogenes to identify sex-specific effects of placental infection on neurodevelopment of the fetus. METHODS Pregnant CD1 mice were infected with a bioluminescent strain of Listeria on embryonic day 14.5 (E14.5). Excised fetuses were imaged on E18.5 to identify the infected placentas. The associated fetal brains were analyzed for gene expression and altered brain structure due to infection. The behavior of adult offspring affected by prenatal Listeria infection was analyzed. RESULTS Placental infection induced sex-specific alteration of gene expression patterns in the fetal brain and resulted in abnormal cortical development correlated with placental infection levels. Furthermore, male offspring exhibited abnormal social interaction, whereas females exhibited elevated anxiety. CONCLUSION Placental infection by Listeria induced sex-specific abnormalities in neurodevelopment of the fetus. Prenatal infection also affected the behavior of the offspring in a sex-specific manner. IMPACT Placental infection with Listeria monocytogenes induces sexually dichotomous gene expression patterns in the fetal brains of mice. Abnormal cortical lamination is correlated with placental infection levels. Placental infection results in autism-related behavior in male offspring and heightened anxiety levels in female offspring.
Collapse
|
15
|
Neuwirth LS, Verrengia MT, Harikinish-Murrary ZI, Orens JE, Lopez OE. Under or Absent Reporting of Light Stimuli in Testing of Anxiety-Like Behaviors in Rodents: The Need for Standardization. Front Mol Neurosci 2022; 15:912146. [PMID: 36061362 PMCID: PMC9428565 DOI: 10.3389/fnmol.2022.912146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Behavioral neuroscience tests such as the Light/Dark Test, the Open Field Test, the Elevated Plus Maze Test, and the Three Chamber Social Interaction Test have become both essential and widely used behavioral tests for transgenic and pre-clinical models for drug screening and testing. However, as fast as the field has evolved and the contemporaneous involvement of technology, little assessment of the literature has been done to ensure that these behavioral neuroscience tests that are crucial to pre-clinical testing have well-controlled ethological motivation by the use of lighting (i.e., Lux). In the present review paper, N = 420 manuscripts were examined from 2015 to 2019 as a sample set (i.e., n = ~20–22 publications per year) and it was found that only a meager n = 50 publications (i.e., 11.9% of the publications sampled) met the criteria for proper anxiogenic and anxiolytic Lux reported. These findings illustrate a serious concern that behavioral neuroscience papers are not being vetted properly at the journal review level and are being released into the literature and public domain making it difficult to assess the quality of the science being reported. This creates a real need for standardizing the use of Lux in all publications on behavioral neuroscience techniques within the field to ensure that contributions are meaningful, avoid unnecessary duplication, and ultimately would serve to create a more efficient process within the pre-clinical screening/testing for drugs that serve as anxiolytic compounds that would prove more useful than what prior decades of work have produced. It is suggested that improving the standardization of the use and reporting of Lux in behavioral neuroscience tests and the standardization of peer-review processes overseeing the proper documentation of these methodological approaches in manuscripts could serve to advance pre-clinical testing for effective anxiolytic drugs. This report serves to highlight this concern and proposes strategies to proactively remedy them as the field moves forward for decades to come.
Collapse
Affiliation(s)
- Lorenz S. Neuwirth
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
- *Correspondence: Lorenz S. Neuwirth
| | - Michael T. Verrengia
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Zachary I. Harikinish-Murrary
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Jessica E. Orens
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Oscar E. Lopez
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| |
Collapse
|
16
|
Anderson RC, O'Keeffe GW, McDermott KW. Characterisation of the consequences of maternal immune activation on distinct cell populations in the developing rat spinal cord. J Anat 2022; 241:938-950. [PMID: 35808977 PMCID: PMC9482694 DOI: 10.1111/joa.13726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
Abstract
Maternal immune activation (MIA) during gestation has been implicated in the development of neurological disorders such as schizophrenia and autism. Epidemiological studies have suggested that the effect of MIA may depend on the gestational timing of the immune challenge and the region of the central nervous system (CNS) in question. This study investigated the effects of MIA with 100 μg/kg lipopolysaccharide at either Embryonic days (E)12 or E16 on the oligodendrocytes, microglia and astrocytes of the offspring spinal cord. At E16, MIA decreased the number of olig2+ and Iba‐1+ cells in multiple grey and white matter regions of the developing spinal cord 5 h after injection. These decreases were not observed at postnatal day 14. In contrast, MIA at E12 did not alter Olig2+ or Iba‐1+ cell number in the developing spinal cord 5 h after injection, however, Olig2+ cell number was decreased in the ventral grey matter of the P14 spinal cord. No changes were observed in glial fibrillary acidic protein (GFAP) expression at P14 following MIA at either E12 or E16. These data suggest that E16 may be a window of immediate vulnerability to MIA during spinal cord development, however, the findings also suggest that the developmental process may be capable of compensation over time. Potential changes in P14 animals following the challenge at E12 are indicative of the complexity of the effects of MIA during the developmental process.
Collapse
Affiliation(s)
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | |
Collapse
|
17
|
Kirschen GW, Panda S, Burd I. Congenital Infection Influence on Early Brain Development Through the Gut-Brain Axis. Front Neurosci 2022; 16:894955. [PMID: 35844234 PMCID: PMC9280077 DOI: 10.3389/fnins.2022.894955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/23/2022] [Indexed: 11/21/2022] Open
Abstract
The mechanisms by which various pathogens cause congenital infections have been studied extensively, aiding in the understanding of the detrimental effects these infections can have on fetal/neonatal neurological development. Recent studies have focused on the gut-brain axis as pivotal in neurodevelopment, with congenital infections causing substantial disruptions. There remains controversy surrounding the purported sterility of the placenta as well as concerns regarding the effects of exposure to antibiotics used during pregnancy on neonatal microbiome development and how early exposure to microbes or antibiotics can shape the gut-brain axis. Long-term neurodevelopmental consequences, such as autism spectrum disorder, attention deficit hyperactivity disorder, and cerebral palsy, may be attributable, in part, to early life infection and changes in the immature gut microbiome. The goal of this review is thus to critically evaluate the current evidence related to early life infection affecting neurodevelopment through the gut-brain axis.
Collapse
Affiliation(s)
- Gregory W. Kirschen
- Department of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD, United States
- Integrated Center for Fetal Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Snigdha Panda
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Irina Burd
- Department of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD, United States
- Integrated Center for Fetal Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
18
|
Saito Reis CA, Ng PK, Kurashima CK, Padron J, Kendal-Wright CE. Fetal DNA Causes Sex-Specific Inflammation From Human Fetal Membranes. Front Physiol 2022; 13:901726. [PMID: 35812324 PMCID: PMC9257279 DOI: 10.3389/fphys.2022.901726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Inflammation is central to the mechanisms of parturition, but the lack of understanding of how it is controlled in normal parturition hampers our ability to understand how it may diverge resulting in preterm birth. Cell-free fetal DNA is found in the amniotic fluid, and it is thought to be able to activate inflammation as a danger-associated molecular pattern. Although its levels increases with gestational age, its effect has not been studied on the human fetal membranes. Thus, the aim of this study was to determine if the fetal DNA can trigger inflammation in the human fetal membranes and, thus, potentially contribute to the inflammatory load. Isolated human amniotic epithelial cells and fetal membrane explants were treated apically with fetal DNA causing the translocation of NF-KB into the nucleus of cells and throughout the cells of the explant layers with time. Fetal membrane explants were treated apically with either small or larger fragments of fetal DNA. IL-6, TNFα, and GM-CSF secretion was measured by ELISA, and pro-MMP2 and pro-MMP9 activity was measured by zymography from apical and basal media. Increased apical IL-6 secretion and basal pro-MMP2 activity was seen with small fragments of fetal DNA. When the data were disaggregated based on fetal sex, males had significant increases in IL-6 secretion and basal increased activity in pro-MMP2 and 9, whereas females had significantly increased basal secretion of TNFα. This was caused by the smaller fragments of fetal DNA, whereas the larger fragments did not cause any significant increases. Male fetal DNA had significantly lower percentages of methylation than females. Thus, when the cytokine and pro-MMP activity data were correlated with methylation percentage, IL-6 secretion significantly correlated negatively, whereas GM-CSF secretion positively correlated. These data support the role of fetal DNA as an inflammatory stimulus in the FM, as measured by increased NF-κB translocation, cytokine secretion, and increased pro-MMP activity. However, the data also suggested that the responses are different from FM tissues of male and female fetuses, and both the fragment size and methylation status of the fetal DNA can influence the magnitude and type of molecule secreted.
Collapse
Affiliation(s)
- Chelsea A. Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
| | - Po’okela K. Ng
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
| | | | - Justin Padron
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Claire Enid Kendal-Wright
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
- *Correspondence: Claire Enid Kendal-Wright,
| |
Collapse
|
19
|
Konsman JP, Laaker CJ, Lloyd KR, Hiltz A, Smith BL, Smail MA, Reyes TM. Translationally relevant mouse model of early life cancer and chemotherapy exposure results in brain and small intestine cytokine responses: A potential link to cognitive deficits. Brain Behav Immun 2022; 99:192-202. [PMID: 34655730 PMCID: PMC8842482 DOI: 10.1016/j.bbi.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 01/03/2023] Open
Abstract
Survivors of acute lymphoblastic leukemia (ALL), the most common childhood cancer, are at increased risk for long-term cognitive problems, including executive function deficits. The chemotherapeutic agent methotrexate (MTX) is used to treat most ALL patients and is closely associated with cognitive deficits. To address how early life cancer chemotherapy leads to cognitive deficits, we developed a translationally relevant mouse model of leukemia survival that exposed mice to leukemic cells and chemotherapeutic drugs (vincristine and MTX, with leucovorin rescue) in early life. Male and female mice were tested several weeks later using novel object recognition (recognition memory) and 5-choice serial reaction time task (executive function). Gene expression of proinflammatory, white matter and synapse-associated molecules was assessed in the prefrontal cortex and small intestine both acutely after chemotherapy and chronically after cognitive testing. Early life cancer-chemotherapy exposure resulted in recognition memory and executive function deficits in adult male mice. Prefrontal cortex expression of the chemokine Ccl2 was increased acutely, while small intestine expression of the proinflammatory cytokine tumor necrosis factor-alpha was elevated both acutely (both sexes) and chronically (males only). Inflammation in the small intestine was correlated with prefrontal cortical proinflammatory and synaptic gene expression changes, as well as to executive function deficits. Collectively, these data indicate that the current protocol results in a robust mouse model in which to study cognitive deficits in leukemia survivors, and suggest that small intestine inflammation may represent a novel contributor to adverse CNS consequences of early life chemotherapy.
Collapse
Affiliation(s)
- Jan Pieter Konsman
- Aquitaine Institute for Integrative and Cognitive Neuroscience (INCIA) UMR CNRS 5287, University of Bordeaux, 33076 Bordeaux, France
| | - Collin J. Laaker
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Kelsey R. Lloyd
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Adam Hiltz
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Brittany L. Smith
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Marissa A. Smail
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Teresa M. Reyes
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| |
Collapse
|
20
|
Disruption of Alternative Splicing in the Amygdala of Pigs Exposed to Maternal Immune Activation. IMMUNO 2021. [DOI: 10.3390/immuno1040035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The inflammatory response of gestating females to infection or stress can disrupt gene expression in the offspring’s amygdala, resulting in lasting neurodevelopmental, physiological, and behavioral disorders. The effects of maternal immune activation (MIA) can be impacted by the offspring’s sex and exposure to additional stressors later in life. The objectives of this study were to investigate the disruption of alternative splicing patterns associated with MIA in the offspring’s amygdala and characterize this disruption in the context of the second stress of weaning and sex. Differential alternative splicing was tested on the RNA-seq profiles of a pig model of viral-induced MIA. Compared to controls, MIA was associated with the differential alternative splicing (FDR-adjusted p-value < 0.1) of 292 and 240 genes in weaned females and males, respectively, whereas 132 and 176 genes were differentially spliced in control nursed female and male, respectively. The majority of the differentially spliced (FDR-adjusted p-value < 0.001) genes (e.g., SHANK1, ZNF672, KCNA6) and many associated enriched pathways (e.g., Fc gamma R-mediated phagocytosis, non-alcoholic fatty liver disease, and cGMP-PKG signaling) have been reported in MIA-related disorders including autism and schizophrenia in humans. Differential alternative splicing associated with MIA was detected in the gene MAG across all sex-stress groups except for unstressed males and SLC2A11 across all groups except unstressed females. Precise understanding of the effect of MIA across second stressors and sexes necessitates the consideration of splicing isoform profiles.
Collapse
|
21
|
Lewis EL, Tulina N, Anton L, Brown AG, Porrett PM, Elovitz MA. IFNγ-Producing γ/δ T Cells Accumulate in the Fetal Brain Following Intrauterine Inflammation. Front Immunol 2021; 12:741518. [PMID: 34675929 PMCID: PMC8524441 DOI: 10.3389/fimmu.2021.741518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/01/2021] [Indexed: 11/26/2022] Open
Abstract
Intrauterine inflammation impacts prenatal neurodevelopment and is linked to adverse neurobehavioral outcomes ranging from cerebral palsy to autism spectrum disorder. However, the mechanism by which a prenatal exposure to intrauterine inflammation contributes to life-long neurobehavioral consequences is unknown. To address this gap in knowledge, this study investigates how inflammation transverses across multiple anatomic compartments from the maternal reproductive tract to the fetal brain and what specific cell types in the fetal brain may cause long-term neuronal injury. Utilizing a well-established mouse model, we found that mid-gestation intrauterine inflammation resulted in a lasting neutrophil influx to the decidua in the absence of maternal systemic inflammation. Fetal immunologic changes were observed at 72-hours post-intrauterine inflammation, including elevated neutrophils and macrophages in the fetal liver, and increased granulocytes and activated microglia in the fetal brain. Through unbiased clustering, a population of Gr-1+ γ/δ T cells was identified as the earliest immune cell shift in the fetal brain of fetuses exposed to intrauterine inflammation and determined to be producing high levels of IFNγ when compared to γ/δ T cells in other compartments. In a case-control study of term infants, IFNγ was found to be elevated in the cord blood of term infants exposed to intrauterine inflammation compared to those without this exposure. Collectively, these data identify a novel cellular immune mechanism for fetal brain injury in the setting of intrauterine inflammation.
Collapse
Affiliation(s)
- Emma L Lewis
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, United States
| | - Natalia Tulina
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, United States
| | - Lauren Anton
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, United States
| | - Amy G Brown
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, United States
| | - Paige M Porrett
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, PA, United States
| | - Michal A Elovitz
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, United States.,Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
22
|
Lovick TA, Zangrossi H. Effect of Estrous Cycle on Behavior of Females in Rodent Tests of Anxiety. Front Psychiatry 2021; 12:711065. [PMID: 34531768 PMCID: PMC8438218 DOI: 10.3389/fpsyt.2021.711065] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/04/2021] [Indexed: 11/13/2022] Open
Abstract
Anxiety disorders are more prevalent in women than in men. In women the menstrual cycle introduces another variable; indeed, some conditions e.g., premenstrual syndrome, are menstrual cycle specific. Animal models of fear and anxiety, which form the basis for research into drug treatments, have been developed almost exclusively, using males. There remains a paucity of work using females and the available literature presents a confusing picture. One confound is the estrous cycle in females, which some authors consider, but many do not. Importantly, there are no accepted standardized criteria for defining cycle phase, which is important given the rapidly changing hormonal profile during the 4-day cycle of rodents. Moreover, since many behavioral tests that involve a learning component or that consider extinction of a previously acquired association require several days to complete; the outcome may depend on the phase of the cycle on the days of training as well as on test days. In this article we consider responsiveness of females compared to males in a number of commonly used behavioral tests of anxiety and fear that were developed in male rodents. We conclude that females perform in a qualitatively similar manner to males in most tests although there may be sex and strain differences in sensitivity. Tests based on unconditioned threatening stimuli are significantly influenced by estrous cycle phase with animals displaying increased responsiveness in the late diestrus phase of the cycle (similar to the premenstrual phase in women). Tests that utilize conditioned fear paradigms, which involve a learning component appear to be less impacted by the estrous cycle although sex and cycle-related differences in responding can still be detected. Ethologically-relevant tests appear to have more translational value in females. However, even when sex differences in behavior are not detected, the same outward behavioral response may be mediated by different brain mechanisms. In order to progress basic research in the field of female psychiatry and psychopharmacology, there is a pressing need to validate and standardize experimental protocols for using female animal models of anxiety-related states.
Collapse
Affiliation(s)
- Thelma A. Lovick
- Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Hélio Zangrossi
- Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
23
|
Breach MR, Dye CN, Joshi A, Platko S, Gilfarb RA, Krug AR, Franceschelli DV, Galan A, Dodson CM, Lenz KM. Maternal allergic inflammation in rats impacts the offspring perinatal neuroimmune milieu and the development of social play, locomotor behavior, and cognitive flexibility. Brain Behav Immun 2021; 95:269-286. [PMID: 33798637 PMCID: PMC8187275 DOI: 10.1016/j.bbi.2021.03.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 01/07/2023] Open
Abstract
Maternal systemic inflammation increases risk for neurodevelopmental disorders like autism, ADHD, and schizophrenia in offspring. Notably, these disorders are male-biased. Studies have implicated immune system dysfunction in the etiology of these disorders, and rodent models of maternal immune activation provide useful tools to examine mechanisms of sex-dependent effects on brain development, immunity, and behavior. Here, we employed an allergen-induced model of maternal inflammation in rats to characterize levels of mast cells and microglia in the perinatal period in male and female offspring, as well as social, emotional, and cognitive behaviors throughout the lifespan. Adult female rats were sensitized to ovalbumin (OVA), bred, and challenged intranasally on gestational day 15 of pregnancy with OVA or saline. Allergic inflammation upregulated microglia in the fetal brain, increased mast cell number in the hippocampus on the day of birth, and conferred region-, time- and sex- specific changes in microglia measures. Additionally, offspring of OVA-exposed mothers subsequently exhibited abnormal social behavior, hyperlocomotion, and reduced cognitive flexibility. These data demonstrate the long-term effects of maternal allergic challenge on offspring development and provide a basis for understanding neurodevelopmental disorders linked to maternal systemic inflammation in humans.
Collapse
Affiliation(s)
- Michaela R. Breach
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Courtney N. Dye
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Aarohi Joshi
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Steven Platko
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Rachel A. Gilfarb
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Annemarie R. Krug
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | | | - Anabel Galan
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Claire M. Dodson
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Kathryn M. Lenz
- Department of Psychology, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
24
|
Graham AM, Marr M, Buss C, Sullivan EL, Fair DA. Understanding Vulnerability and Adaptation in Early Brain Development using Network Neuroscience. Trends Neurosci 2021; 44:276-288. [PMID: 33663814 PMCID: PMC8216738 DOI: 10.1016/j.tins.2021.01.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 10/15/2020] [Accepted: 01/27/2021] [Indexed: 01/07/2023]
Abstract
Early adversity influences brain development and emerging behavioral phenotypes relevant for psychiatric disorders. Understanding the effects of adversity before and after conception on brain development has implications for contextualizing current public health crises and pervasive health inequities. The use of functional magnetic resonance imaging (fMRI) to study the brain at rest has shifted understanding of brain functioning and organization in the earliest periods of life. Here we review applications of this technique to examine effects of early life stress (ELS) on neurodevelopment in infancy, and highlight targets for future research. Building on the foundation of existing work in this area will require tackling significant challenges, including greater inclusion of often marginalized segments of society, and conducting larger, properly powered studies.
Collapse
Affiliation(s)
- Alice M Graham
- Department of Psychiatry, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Mollie Marr
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Claudia Buss
- Department of Medical Psychology, Charité University of Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; Development, Health, and Disease Research Program, University of California, Irvine, 837 Health Sciences Drive, Irvine, California, 92697, USA
| | - Elinor L Sullivan
- Department of Psychiatry, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA; Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA; Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Ave., Beaverton, OR, 97006, USA
| | - Damien A Fair
- The Masonic Institute of the Developing Brain, The University of Minnesota, Department of Pediatrics, The University of Minnesota Institute of Child Development, The University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
Regulation of Superoxide by BAP31 through Its Effect on p22 phox and Keap1/Nrf2/HO-1 Signaling Pathway in Microglia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1457089. [PMID: 33777312 PMCID: PMC7969104 DOI: 10.1155/2021/1457089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 11/28/2020] [Accepted: 12/23/2020] [Indexed: 11/20/2022]
Abstract
Reactive oxygen species (ROS) production by activation of microglia is considered to be a major cause of neuronal dysfunction, which can lead to damage and death through direct oxidative damage to neuronal macromolecules or derangement of neuronal redox signaling circuits. BAP31, an integral ER membrane protein, has been defined as a regulatory molecule in the CNS. Our latest studies have found that BAP31 deficiency leads to activation of microglia. In this study, we discovered that BAP31 deficiency upregulated LPS-induced superoxide anion production in BV2 cells and mice by upregulating the expression level of p22phox and by inhibiting the activation of Nrf2-HO-1 signaling. Knockdown of p22phox/keap1 or use of an NADPH oxidase inhibitor (apocynin) reversed the production of superoxide anion and inflammatory cytokines, which then reduced neuronal damage and death in vitro and in vivo. These results suggest that BAP31 deficiency contributes to microglia-related superoxide anion production and neuroinflammation through p22phox and keap1. Furthermore, the excess superoxide anion cooperated with inflammatory cytokines to induce the damage and death of neurons. Thus, we determined that BAP31 is an important regulator in superoxide anion production and neuroinflammation, and the downstream regulators or agonists of BAP31 could therefore be considered as potential therapeutic targets in microglial-related superoxide anion production and neuroinflammation.
Collapse
|
26
|
Hunter SK, Hoffman MC, D'Alessandro A, Noonan K, Wyrwa A, Freedman R, Law AJ. Male fetus susceptibility to maternal inflammation: C-reactive protein and brain development. Psychol Med 2021; 51:450-459. [PMID: 31787129 PMCID: PMC7263978 DOI: 10.1017/s0033291719003313] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Maternal inflammation in early pregnancy has been identified epidemiologically as a prenatal pathogenic factor for the offspring's later mental illness. Early newborn manifestations of the effects of maternal inflammation on human fetal brain development are largely unknown. METHODS Maternal infection, depression, obesity, and other factors associated with inflammation were assessed at 16 weeks gestation, along with maternal C-reactive protein (CRP), cytokines, and serum choline. Cerebral inhibition was assessed by inhibitory P50 sensory gating at 1 month of age, and infant behavior was assessed by maternal ratings at 3 months of age. RESULTS Maternal CRP diminished the development of cerebral inhibition in newborn males but paradoxically increased inhibition in females. Similar sex-dependent effects were seen in mothers' assessment of their infant's self-regulatory behaviors at 3 months of age. Higher maternal choline levels partly mitigated the effect of CRP in male offspring. CONCLUSIONS The male fetal-placental unit appears to be more sensitive to maternal inflammation than females. Effects are particularly marked on cerebral inhibition. Deficits in cerebral inhibition 1 month after birth, similar to those observed in several mental illnesses, including schizophrenia, indicate fetal developmental pathways that may lead to later mental illness. Deficits in early infant behavior follow. Early intervention before birth, including prenatal vitamins, folate, and choline supplements, may help prevent fetal development of pathophysiological deficits that can have life-long consequences for mental health.
Collapse
Affiliation(s)
- Sharon K Hunter
- Departments of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
| | - M Camille Hoffman
- Departments of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
- Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
| | - Angelo D'Alessandro
- Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
| | - Kathleen Noonan
- Departments of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
| | - Anna Wyrwa
- Departments of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
| | - Robert Freedman
- Departments of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
| | - Amanda J Law
- Departments of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
- Cell and Developmental Biology, University of Colorado Denver School of Medicine, Aurora, Colorado80045, USA
| |
Collapse
|
27
|
Rymut HE, Bolt CR, Caputo MP, Houser AK, Antonson AM, Zimmerman JD, Villamil MB, Southey BR, Rund LA, Johnson RW, Rodriguez-Zas SL. Long-Lasting Impact of Maternal Immune Activation and Interaction With a Second Immune Challenge on Pig Behavior. Front Vet Sci 2020; 7:561151. [PMID: 33330688 PMCID: PMC7732429 DOI: 10.3389/fvets.2020.561151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/28/2020] [Indexed: 01/30/2023] Open
Abstract
The combined effects on pig behavior of maternal immune challenge during gestation followed by a second immune challenge later in life have not been studied. Porcine reproductive and respiratory syndrome virus (PRRSV) infection during gestation can elicit maternal immune activation (MIA) yet the interactions with the offspring response to a second immune challenge after birth remains unexplored. Knowledge on the response to viral challenges in rodents has been gained through the use of the viral mimetic polyinosinic-polycytidylic acid (Poly(I:C)), yet the effects of this immune stimulant on pig behavior have not been assessed. This study advances the understanding of the combined effect of MIA and a second immune challenge later in life on female and male pig behavior. Three complementary experiments enabled the development of an effective Poly(I:C) challenge in pigs, and testing the interaction between PRRSV-elicited MIA, Poly(I:C) challenge at 60 days of age, and sex on behaviors. Individual-level observations on sickness, locomotor, and social behaviors were measured 1-3 h after Poly(I:C) challenge. Vomiting, panting, lethargy, walking, laying, playing, and touching behaviors were analyzed using generalized linear mixed effect models. Results indicated that a Poly(I:C) dose of 1 mg/kg within 1 h after injection increased the incidence of laying and sickness behavior. The Poly(I:C) challenge decreased the incidence of locomotor behaviors and activity levels. Pigs exposed to MIA had lower rates of social behaviors such as playing. The combined effect of PRRSV-elicited MIA and Poly(I:C) immune challenge further sensitized the pigs to behavior disruption across sexes including changes in sternal and lateral laying, walking, lethargy, and touching incidence. Notably, the effects of Poly(I:C) immune challenge alone on behaviors tended to be more extreme in males, whereas the effects of Poly(I:C) following MIA tended to be more extreme in females. Our findings demonstrate that MIA and Poly(I:C) affected behaviors, and the viral mimetic effects shortly after injection can offer insights into the prolonged effect of postnatal viral infections on feeding, social interactions and health status. Management practices that reduce the likelihood of gestational diseases and accommodate for behavioral disruptions in the offspring can minimize the impact of MIA.
Collapse
Affiliation(s)
- Haley E Rymut
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Courtni R Bolt
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Megan P Caputo
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Alexandra K Houser
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Adrienne M Antonson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Jalisa D Zimmerman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Maria B Villamil
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Laurie A Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Rodney W Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Sandra L Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,C. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
28
|
Delage CI, Cornil CA. Estrogen‐dependent sex difference in microglia in the developing brain of Japanese quail (
Coturnix japonica
). Dev Neurobiol 2020; 80:239-262. [DOI: 10.1002/dneu.22781] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/26/2022]
Affiliation(s)
| | - Charlotte Anne Cornil
- Laboratory of Neuroendocrinology GIGA Neurosciences University of Liège Liège Belgium
| |
Collapse
|
29
|
Reddaway J, Brydges NM. Enduring neuroimmunological consequences of developmental experiences: From vulnerability to resilience. Mol Cell Neurosci 2020; 109:103567. [PMID: 33068720 PMCID: PMC7556274 DOI: 10.1016/j.mcn.2020.103567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/14/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
The immune system is crucial for normal neuronal development and function (neuroimmune system). Both immune and neuronal systems undergo significant postnatal development and are sensitive to developmental programming by environmental experiences. Negative experiences from infection to psychological stress at a range of different time points (in utero to adolescence) can permanently alter the function of the neuroimmune system: given its prominent role in normal brain development and function this dysregulation may increase vulnerability to psychiatric illness. In contrast, positive experiences such as exercise and environmental enrichment are protective and can promote resilience, even restoring the detrimental effects of negative experiences on the neuroimmune system. This suggests the neuroimmune system is a viable therapeutic target for treatment and prevention of psychiatric illnesses, especially those related to stress. In this review we will summarise the main cells, molecules and functions of the immune system in general and with specific reference to central nervous system development and function. We will then discuss the effects of negative and positive environmental experiences, especially during development, in programming the long-term functioning of the neuroimmune system. Finally, we will review the sparse but growing literature on sex differences in neuroimmune development and response to environmental experiences. The immune system is essential for development and function of the central nervous system (neuroimmune system) Environmental experiences can permanently alter neuroimmune function and associated brain development Altered neuroimmune function following negative developmental experiences may play a role in psychiatric illnesses Positive experiences can promote resilience and rescue the effects of negative experiences on the neuroimmune system The neuroimmune system is therefore a viable therapeutic target for preventing and treating psychiatric illnesses
Collapse
Affiliation(s)
- Jack Reddaway
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Nichola M Brydges
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK.
| |
Collapse
|
30
|
Chamera K, Kotarska K, Szuster-Głuszczak M, Trojan E, Skórkowska A, Pomierny B, Krzyżanowska W, Bryniarska N, Basta-Kaim A. The prenatal challenge with lipopolysaccharide and polyinosinic:polycytidylic acid disrupts CX3CL1-CX3CR1 and CD200-CD200R signalling in the brains of male rat offspring: a link to schizophrenia-like behaviours. J Neuroinflammation 2020; 17:247. [PMID: 32829711 PMCID: PMC7444338 DOI: 10.1186/s12974-020-01923-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/10/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The bidirectional communication between neurons and microglia is fundamental for the homeostasis and biological function of the central nervous system. Maternal immune activation (MIA) is considered to be one of the factors affecting these interactions. Accordingly, MIA has been suggested to be involved in several neuropsychiatric diseases, including schizophrenia. The crucial regulatory systems for neuron-microglia crosstalk are the CX3CL1-CX3CR1 and CD200-CD200R axes. METHODS We aimed to clarify the impact of MIA on CX3CL1-CX3CR1 and CD200-CD200R signalling pathways in the brains of male Wistar rats in early and adult life by employing two neurodevelopmental models of schizophrenia based on the prenatal challenge with lipopolysaccharide (LPS) and polyinosinic:polycytidylic acid (Poly I:C). We also examined the effect of MIA on the expression of microglial markers and the profile of cytokines released in the brains of young offspring, as well as the behaviour of adult animals. Moreover, we visualized the localization of ligand-receptor systems in the hippocampal regions (CA1, CA3 and DG) and the frontal cortex of young rats exposed to MIA. The differences between groups were analysed using Student's t test. RESULTS We observed that MIA altered developmental trajectories in neuron-microglia communication in the brains of young offspring, as evidenced by the disruption of CX3CL1-CX3CR1 and/or CD200-CD200R axes. Our data demonstrated the presence of abnormalities after LPS-induced MIA in levels of Cd40, Il-1β, Tnf-α, Arg1, Tgf-β and Il-10, as well as IBA1, IL-1β and IL-4, while after Poly I:C-generated MIA in levels of Cd40, iNos, Il-6, Tgf-β, Il-10, and IBA1, IL-1β, TNF-α, IL-6, TGF-β and IL-4 early in the life of male animals. In adult male rats that experienced prenatal exposure to MIA, we observed behavioural changes resembling a schizophrenia-like phenotype. CONCLUSIONS Our study provides evidence that altered CX3CL1-CX3CR1 and/or CD200-CD200R pathways, emerging after prenatal immune challenge with LPS and Poly I:C, might be involved in the aetiology of schizophrenia.
Collapse
Affiliation(s)
- Katarzyna Chamera
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Katarzyna Kotarska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Magdalena Szuster-Głuszczak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Alicja Skórkowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Collegium Medicum, 9 Medyczna St, 30-688, Kraków, Poland
| | - Bartosz Pomierny
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Collegium Medicum, 9 Medyczna St, 30-688, Kraków, Poland
| | - Weronika Krzyżanowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Collegium Medicum, 9 Medyczna St, 30-688, Kraków, Poland
| | - Natalia Bryniarska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland.
| |
Collapse
|
31
|
Freedman R, Hunter SK, Noonan K, Wyrwa A, Christians U, Law AJ, Hoffman MC. Maternal Prenatal Depression in Pregnancies With Female and Male Fetuses and Developmental Associations With C-reactive Protein and Cortisol. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 6:310-320. [PMID: 33060035 DOI: 10.1016/j.bpsc.2020.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/01/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Prenatal depression has lasting effects on development in offspring, including later mental illness risk. Maternal responses to depression include inflammation and hypothalamic-pituitary-adrenal axis stimulation. Effects on development of cerebral inhibitory neurocircuits may differ for female and male fetuses. METHODS Mothers (N = 181) were assessed periodically, beginning at 16 weeks' gestation, using the Center for Epidemiologic Studies-Depression Scale. Maternal prenatal C-reactive protein and hair cortisol and cortisone levels were determined. Cortisone was determined in neonatal hair. Development of cerebral inhibitory neurocircuits was assessed in 162 1-month-old newborns by inhibition of P50 electrophysiological responses to repeated sounds. RESULTS Maternal depression was associated with decreased newborn P50 inhibition in both sexes. Maternal C-reactive protein levels were significantly associated with depression only in pregnancies with male fetuses and with decreased newborn P50 inhibition only in male newborns. Maternal cortisol levels were significantly associated with depression only in pregnancies with female fetuses and with decreased newborn P50 inhibition only in female newborns. In pregnancies with male fetuses compared with pregnancies with female fetuses, cortisol was more robustly metabolized to cortisone, which does not activate cortisol receptors. CONCLUSIONS This study finds sex-specific associations of C-reactive protein and cortisol levels with prenatal depression in women and with decreased development of newborn P50 inhibition. Sex-based differences in maternal response to depression with inflammation or cortisol and their developmental effects may reflect evolutionary influences to promote survival in adversity. Decreased newborn P50 inhibition is associated with later childhood behavioral problems, and decreased P50 inhibition is a pathophysiological feature of several mental illnesses.
Collapse
Affiliation(s)
- Robert Freedman
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado.
| | - Sharon K Hunter
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - Kathleen Noonan
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - Anna Wyrwa
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - Uwe Christians
- Department of Psychiatry, iC42 Clinical Research and Development, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - Amanda J Law
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado; Department of Anesthesiology, Department of Cell and Developmental Biology, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - M Camille Hoffman
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado; Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| |
Collapse
|
32
|
The associations of maternal polycystic ovary syndrome and hirsutism with behavioral problems in offspring. Fertil Steril 2020; 113:435-443. [PMID: 32106995 DOI: 10.1016/j.fertnstert.2019.09.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To study the associations between maternal polycystic ovary syndrome (PCOS) and hirsutism with offspring attention-deficit/hyperactivity disorder (ADHD), anxiety, conduct disorder, and behavioral problems. DESIGN Prospective birth cohort study. SETTING Not applicable. PATIENT(S) A total of 1,915 mother-child dyads. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Maternal report of offspring ADHD, anxiety, or conduct disorder diagnosis at 7 to 8 years; emotional symptoms, behavioral problems (including peer relationship, conduct, hyperactivity/inattention), and prosocial problems measured with the Strengths and Difficulties Questionnaire (SDQ) at 7 years. RESULT(S) Prevalence of PCOS and hirsutism were 12.0% and 3.9%; 84% of women with hirsutism had PCOS. After adjustment for sociodemographic covariates, prepregnancy body mass index, and parental history of affective disorders, children born to mothers with PCOS had higher risk of anxiety (adjusted risk ratio [aRR] 1.62; 95% confidence interval [CI], 1.02-2.57) and borderline emotional symptoms (aRR 1.66; 95% CI, 1.18-2.33) compared with children born to mothers without PCOS. The associations between maternal PCOS and offspring ADHD were positive but imprecise. Maternal hirsutism was related to a higher risk of children's ADHD (aRR 2.33; 95% CI, 1.28-4.24), conduct disorder (aRR 2.54; 95% CI 1.18-5.47), borderline emotional symptoms, peer relationship problems, and conduct problems (aRRs 2.61; 95% CI, 1.69-4.05; 1.92; 95% CI, 1.16-3.17; and 2.22; 95% CI, 1.30-3.79, respectively). CONCLUSION(S) Maternal PCOS was associated with offspring anxiety, and hirsutism was related to other offspring behavioral problems. These findings should be interpreted with caution as replication is needed in prospective cohort studies that assess PCOS and hirsutism diagnoses using medical records.
Collapse
|
33
|
Maternal Immune Activation Sensitizes Male Offspring Rats to Lipopolysaccharide-Induced Microglial Deficits Involving the Dysfunction of CD200-CD200R and CX3CL1-CX3CR1 Systems. Cells 2020; 9:cells9071676. [PMID: 32664639 PMCID: PMC7407118 DOI: 10.3390/cells9071676] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
Early life challenges resulting from maternal immune activation (MIA) may exert persistent effects on the offspring, including the development of psychiatric disorders, such as schizophrenia. Recent evidence has suggested that the adverse effects of MIA may be mediated by neuron-microglia crosstalk, particularly CX3CL1-CX3CR1 and CD200-CD200R dyads. Therefore, the present study assessed the behavioural parameters resembling schizophrenia-like symptoms in the adult male offspring of Sprague-Dawley rats that were exposed to MIA and to an additional acute lipopolysaccharide (LPS) challenge in adulthood, according to the "two-hit" hypothesis of schizophrenia. Simultaneously, we aimed to clarify the role of the CX3CL1-CX3CR1 and CD200-CD200R axes and microglial reactivity in the brains of adult offspring subjected to MIA and the "second hit" wit LPS. In the present study, MIA generated a range of behavioural changes in the adult male offspring, including increased exploratory activity and anxiety-like behaviours. The most intriguing finding was observed in the prepulse inhibition (PPI) test, where the deficit in the sensorimotor gating was age-dependent and present only in part of the rats. We were able to distinguish the occurrence of two groups: responsive and non-responsive (without the deficit). Concurrently, based on the results of the biochemical studies, MIA disrupted mainly the CD200-CD200R system, while the changes of the CX3CL1-CX3CR1 axis were less evident in the frontal cortex of adult non-responsive offspring. MIA markedly affected the immune regulators of the CD200-CD200R pathway as we observed an increase in cortical IL-6 release in the responsive group and IL-4 in the non-responsive offspring. Importantly, the "second hit" generated disturbances at the behavioural and biochemical levels mostly in the non-responsive adult animals. Those offspring were characterized both by disturbed PPI and "priming" microglia. Altogether, the exposure to MIA altered the immunomodulatory mechanisms, including the CD200-CD200R axis, in the brain and sensitized animals to subsequent immunological challenges, leading to the manifestation of schizophrenia-like alterations.
Collapse
|
34
|
Beyeler SA, Hodges MR, Huxtable AG. Impact of inflammation on developing respiratory control networks: rhythm generation, chemoreception and plasticity. Respir Physiol Neurobiol 2020; 274:103357. [PMID: 31899353 DOI: 10.1016/j.resp.2019.103357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/17/2019] [Accepted: 12/02/2019] [Indexed: 10/25/2022]
Abstract
The respiratory control network in the central nervous system undergoes critical developmental events early in life to ensure adequate breathing at birth. There are at least three "critical windows" in development of respiratory control networks: 1) in utero, 2) newborn (postnatal day 0-4 in rodents), and 3) neonatal (P10-13 in rodents, 2-4 months in humans). During these critical windows, developmental processes required for normal maturation of the respiratory control network occur, thereby increasing vulnerability of the network to insults, such as inflammation. Early life inflammation (induced by LPS, chronic intermittent hypoxia, sustained hypoxia, or neonatal maternal separation) acutely impairs respiratory rhythm generation, chemoreception and increases neonatal risk of mortality. These early life impairments are also greater in young males, suggesting sex-specific impairments in respiratory control. Further, neonatal inflammation has a lasting impact on respiratory control by impairing adult respiratory plasticity. This review focuses on how inflammation alters respiratory rhythm generation, chemoreception and plasticity during each of the three critical windows. We also highlight the need for additional mechanistic studies and increased investigation into how glia (such as microglia and astrocytes) play a role in impaired respiratory control after inflammation. Understanding how inflammation during critical windows of development disrupt respiratory control networks is essential for developing better treatments for vulnerable neonates and preventing adult ventilatory control disorders.
Collapse
Affiliation(s)
- Sarah A Beyeler
- Department of Human Physiology, University of Oregon, Eugene, OR, 97403, United States
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Adrianne G Huxtable
- Department of Human Physiology, University of Oregon, Eugene, OR, 97403, United States.
| |
Collapse
|
35
|
Brydges NM, Reddaway J. Neuroimmunological effects of early life experiences. Brain Neurosci Adv 2020; 4:2398212820953706. [PMID: 33015371 PMCID: PMC7513403 DOI: 10.1177/2398212820953706] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022] Open
Abstract
Exposure to adverse experiences during development increases the risk of psychiatric illness later in life. Growing evidence suggests a role for the neuroimmune system in this relationship. There is now substantial evidence that the immune system is critical for normal brain development and behaviour, and responds to environmental perturbations experienced early in life. Severe or chronic stress results in dysregulated neuroimmune function, concomitant with abnormal brain morphology and function. Positive experiences including environmental enrichment and exercise exert the opposite effect, promoting normal brain and immune function even in the face of early life stress. The neuroimmune system may therefore provide a viable target for prevention and treatment of psychiatric illness. This review will briefly summarise the neuroimmune system in brain development and function, and review the effects of stress and positive environmental experiences during development on neuroimmune function. There are also significant sex differences in how the neuroimmune system responds to environmental experiences early in life, which we will briefly review.
Collapse
Affiliation(s)
- Nichola M. Brydges
- Neuroscience and Mental Health Research
Institute, Cardiff University, Cardiff, UK
| | - Jack Reddaway
- Neuroscience and Mental Health Research
Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
36
|
Shaw GA, Dupree JL, Neigh GN. Adolescent maturation of the prefrontal cortex: Role of stress and sex in shaping adult risk for compromise. GENES BRAIN AND BEHAVIOR 2019; 19:e12626. [DOI: 10.1111/gbb.12626] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Gladys A. Shaw
- Department of Anatomy and NeurobiologyVirginia Commonwealth University Richmond Virginia
| | - Jeffrey L. Dupree
- Department of Anatomy and NeurobiologyVirginia Commonwealth University Richmond Virginia
- Research ServiceHunter Holmes McGuire VA Medical Center Richmond Virginia
| | - Gretchen N. Neigh
- Department of Anatomy and NeurobiologyVirginia Commonwealth University Richmond Virginia
| |
Collapse
|
37
|
"Females Are Not Just 'Protected' Males": Sex-Specific Vulnerabilities in Placenta and Brain after Prenatal Immune Disruption. eNeuro 2019; 6:ENEURO.0358-19.2019. [PMID: 31611335 PMCID: PMC6838689 DOI: 10.1523/eneuro.0358-19.2019] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/27/2019] [Indexed: 11/21/2022] Open
Abstract
Current perceptions of genetic and environmental vulnerabilities in the developing fetus are biased toward male outcomes. An argument is made that males are more vulnerable to gestational complications and neurodevelopmental disorders, the implication being that an understanding of disrupted development in males is sufficient to understand causal mechanisms that are assumed to be similar but attenuated in females. Here we examine this assumption in the context of immune-driven alterations in fetal brain development and related outcomes in female and male mice. Pregnant C57BL/6 mice were treated with low-dose lipopolysaccharide at embryonic day 12.5. Placental pathology, acute fetal brain inflammation and hypoxia, long-term changes in adult cortex cytoarchitecture, altered densities and ratio of excitatory (Satb2+) to inhibitory (parvalbumin+) neuronal subtypes, postnatal growth, and behavior outcomes were compared between male and female offspring. We find that while males experience more pronounced placental pathology, fetal brain hypoxia, depleted PV and Satb2+ densities, and social and learning-related behavioral abnormalities, females exhibit unique acute inflammatory signaling in fetal brain, postnatal growth delay, opposite alterations in cortical PV densities, changes in juvenile behavior, delayed postnatal body growth, and elevated anxiety-related behavior as adults. While males are more severely impacted by prenatal immune disruption by several measures, females exposed to the same insult exhibit a unique set of vulnerabilities and developmental consequences that is not present in males. Our results clearly outline disparate sex-specific features of prenatal vulnerability to inflammatory insults and warn against the casual extrapolation of male disease mechanisms to females.
Collapse
|
38
|
VanRyzin JW, Marquardt AE, Pickett LA, McCarthy MM. Microglia and sexual differentiation of the developing brain: A focus on extrinsic factors. Glia 2019; 68:1100-1113. [PMID: 31691400 DOI: 10.1002/glia.23740] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022]
Abstract
Microglia, the innate immune cells of the brain, have recently been removed from the position of mere sentinels and promoted to the role of active sculptors of developing circuits and cells. Alongside their functions in normal brain development, microglia coordinate sexual differentiation of the brain, a set of processes which vary by region and endpoint like that of microglia function itself. In this review, we highlight the ways microglia are both targets and drivers of brain sexual differentiation. We examine the factors that may drive sex differences in microglia, with a special focus on how changing microenvironments in the developing brain dictate microglia phenotypes and discuss how their diverse functions sculpt lasting sex-specific changes in the brain. Finally, we consider how sex-specific early life environments contribute to epigenetic programming and lasting sex differences in microglia identity.
Collapse
Affiliation(s)
- Jonathan W VanRyzin
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ashley E Marquardt
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lindsay A Pickett
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland
| | - Margaret M McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
39
|
Chávez CE, Oyarzún JE, Avendaño BC, Mellado LA, Inostroza CA, Alvear TF, Orellana JA. The Opening of Connexin 43 Hemichannels Alters Hippocampal Astrocyte Function and Neuronal Survival in Prenatally LPS-Exposed Adult Offspring. Front Cell Neurosci 2019; 13:460. [PMID: 31680871 PMCID: PMC6797550 DOI: 10.3389/fncel.2019.00460] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/27/2019] [Indexed: 01/19/2023] Open
Abstract
Clinical evidence has revealed that children born from mothers exposed to viral and bacterial pathogens during pregnancy are more likely to suffer various neurological disorders including schizophrenia, autism bipolar disorder, major depression, epilepsy, and cerebral palsy. Despite that most research has centered on the impact of prenatal inflammation in neurons and microglia, the potential modifications of astrocytes and neuron-astrocyte communication have received less scrutiny. Here, we evaluated whether prenatally LPS-exposed offspring display alterations in the opening of astrocyte hemichannels and pannexons in the hippocampus, together with changes in neuroinflammation, intracellular Ca2+ and nitric oxide (NO) signaling, gliotransmitter release, cell arborization, and neuronal survival. Ethidium uptake recordings revealed that prenatal LPS exposure enhances the opening of astrocyte Cx43 hemichannels and Panx1 channels in the hippocampus of adult offspring mice. This enhanced channel activity occurred by a mechanism involving a microglia-dependent production of IL-1β/TNF-α and the stimulation of p38 MAP kinase/iNOS/[Ca2+]i-mediated signaling and purinergic/glutamatergic pathways. Noteworthy, the activity of Cx43 hemichannels affected the release of glutamate, [Ca2+]i handling, and morphology of astrocytes, whereas also disturbed neuronal function, including the dendritic arbor and spine density, as well as survival. We speculate that excitotoxic levels of glutamate triggered by the activation of Cx43 hemichannels may contribute to hippocampal neurotoxicity and damage in prenatally LPS-exposed offspring. Therefore, the understanding of how astrocyte-neuron crosstalk is an auspicious avenue toward the development of broad treatments for several neurological disorders observed in children born to women who had a severe infection during gestation.
Collapse
Affiliation(s)
- Carolina E Chávez
- Departamento de Neurología, Facultad de Medicina, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan E Oyarzún
- Departamento de Neurología, Facultad de Medicina, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Beatriz C Avendaño
- Departamento de Neurología, Facultad de Medicina, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis A Mellado
- Departamento de Neurología, Facultad de Medicina, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carla A Inostroza
- Departamento de Neurología, Facultad de Medicina, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tanhia F Alvear
- Departamento de Neurología, Facultad de Medicina, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan A Orellana
- Departamento de Neurología, Facultad de Medicina, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
40
|
Brown AG, Maubert ME, Anton L, Heiser LM, Elovitz MA. The tracking of lipopolysaccharide through the feto-maternal compartment and the involvement of maternal TLR4 in inflammation-induced fetal brain injury. Am J Reprod Immunol 2019; 82:e13189. [PMID: 31495009 PMCID: PMC6899932 DOI: 10.1111/aji.13189] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 11/28/2022] Open
Abstract
Problem Exposure to intrauterine inflammation (IUI) has been shown to induce fetal brain injury and increase the risk of acquiring a neurobehavioral disorder. The trafficking of the inflammatory mediator, lipopolysaccharide (LPS), in the pregnant female reproductive tract in the setting of IUI and the precise mechanisms by which inflammation induces fetal brain injury are not fully understood. Method of study FITC‐labeled LPS was utilized to induce IUI on E15, tissues were collected, and fluorescence was visualized via the Spectrum IVIS. Embryo transfer was utilized to create divergent maternal and fetal genotypes. Wild‐type (WT) embryos were transferred into TLR4−/− pseudopregnant dams (TLR4−/−mat/WTfet). On E15, TLR4−/−mat/WTfet dams or their WT controls (WTmat/WTfet) received an intrauterine injection of LPS or phosphate‐buffered saline (PBS). Endotoxin and IL‐6 levels were assessed in amniotic fluid, and cytokine expression was measured via QPCR. Results Lipopolysaccharide trafficked to the uterus, fetal membranes, placenta, and the fetus and was undetectable in other tissues. Endotoxin was present in the amniotic fluid of all animals exposed to LPS. However, the immune response was blunted in TLR4−/−mat/WTfet compared with WT controls. Conclusion Intrauterine administered LPS is capable of accessing the entire feto‐placental unit with or without a functional maternal TLR4. Thus, bacteria or bacterial byproducts in the uterus may negatively impact fetal development regardless of the maternal genotype or endotoxin response. Despite the blunted immune response in the TLR4‐deficient dams, an inflammatory response is still ignited in the amniotic cavity and may negatively impact the fetus. IL‐6 protein expression in the amniotic fluid of WTmat/WTfet and TLR4‐/‐mat/WTfet Pregnant females were treated with an intrauterine dose of LPS (250 μg) or PBS on E15. LPS injection resulted in significantly increased IL‐6 protein in WT animals (*, P = 0.0017) compared to controls. LPS did not significantly elevate IL‐6 levels in the TLR4‐/‐mat/WTfet animals. The WTmat/WTfet dams had a significantly higher immune response compared to their TLR4‐/‐mat/WTfet counterparts (#, P = 0.015).
Collapse
Affiliation(s)
- Amy G Brown
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Monique E Maubert
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren Anton
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Laura M Heiser
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michal A Elovitz
- Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
41
|
Exposure to in utero inflammation increases locomotor activity, alters cognitive performance and drives vulnerability to cognitive performance deficits after acute immune activation. Brain Behav Immun 2019; 80:56-65. [PMID: 30797960 DOI: 10.1016/j.bbi.2019.02.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/31/2018] [Accepted: 02/20/2019] [Indexed: 12/24/2022] Open
Abstract
Fetal exposure to intrauterine inflammation (IUI) affects brain development. Using intrauterine lipopolysaccharide (LPS) administration to induce a localized, rather than a systemic, inflammation, we have previously shown that IUI increases cytokine expression and microglia number, and reduces white matter in the brains of exposed offspring. Clinical data suggest that IUI may increase the risk for cognitive and neurodevelopmental disorders, however, IUI is often found in the context of preterm birth, making it difficult to disentangle the adverse effects of inflammation from those related to prematurity. Therefore, using a mouse model of IUI that does not involve preterm birth, operant tasks were used to evaluate motivation, attention, impulsivity, and locomotion. IUI-exposed offspring were found to have increased locomotion and increased motivation (females only), and testing in the 5-choice serial reaction time task (5-CSRTT) showed that IUI-exposed offspring performed more trials and could respond accurately at a shorter stimulus length. We have previously shown that IUI animals have a potentiated cytokine response to a "second hit" (acute LPS injection) in adulthood, so animals' performance in the 5CSRTT was evaluated following an acute injection of LPS. As opposed to the improved performance observed under baseline conditions, IUI exposed animals demonstrated a greater decrease in performance after an acute LPS administration. To identify putative molecular mechanisms underlying this potentiated decline in cognitive performance, PFC samples were collected immediately after post-LPS cognitive testing and targeted gene expression analysis was correlated with specific measures of cognitive performance. Three receptors important for neuron-microglia crosstalk were found to correlate with task performance in the males following acute LPS administration. These data demonstrate that early life exposure to localized inflammation of the uterus, in the absence of prematurity, increases locomotor activity and improves some aspects of cognitive performance, but drives a vulnerability for adult cognitive performance deficits in response to acute infection.
Collapse
|
42
|
Barrientos RM, Brunton PJ, Lenz KM, Pyter L, Spencer SJ. Neuroimmunology of the female brain across the lifespan: Plasticity to psychopathology. Brain Behav Immun 2019; 79:39-55. [PMID: 30872093 PMCID: PMC6591071 DOI: 10.1016/j.bbi.2019.03.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Accepted: 03/09/2019] [Indexed: 02/06/2023] Open
Abstract
The female brain is highly dynamic and can fundamentally remodel throughout the normal ovarian cycle as well as in critical life stages including perinatal development, pregnancy and old-age. As such, females are particularly vulnerable to infections, psychological disorders, certain cancers, and cognitive impairments. We will present the latest evidence on the female brain; how it develops through the neonatal period; how it changes through the ovarian cycle in normal individuals; how it adapts to pregnancy and postpartum; how it responds to illness and disease, particularly cancer; and, finally, how it is shaped by old age. Throughout, we will highlight female vulnerability to and resilience against disease and dysfunction in the face of environmental challenges.
Collapse
Affiliation(s)
- R M Barrientos
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH 43210, United States
| | - P J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, Scotland, UK; Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, International Campus, Haining, Zhejiang 314400, PR China
| | - K M Lenz
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychology, Department of Neuroscience, The Ohio State University, Columbus, OH 43210, United States
| | - L Pyter
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States
| | - S J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic. 3083, Australia.
| |
Collapse
|
43
|
Dunn GA, Nigg JT, Sullivan EL. Neuroinflammation as a risk factor for attention deficit hyperactivity disorder. Pharmacol Biochem Behav 2019; 182:22-34. [PMID: 31103523 PMCID: PMC6855401 DOI: 10.1016/j.pbb.2019.05.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 01/08/2023]
Abstract
Attention Deficit Hyperactivity Disorder (ADHD) is a persistent, and impairing pediatric-onset neurodevelopmental condition. Its high prevalence, and recurrent controversy over its widespread identification and treatment, drive strong interest in its etiology and mechanisms. Emerging evidence for a role for neuroinflammation in ADHD pathophysiology is of great interest. This evidence includes 1) the above-chance comorbidity of ADHD with inflammatory and autoimmune disorders, 2) initial studies indicating an association with ADHD and increased serum cytokines, 3) preliminary evidence from genetic studies demonstrating associations between polymorphisms in genes associated with inflammatory pathways and ADHD, 4) emerging evidence that early life exposure to environmental factors may increase risk for ADHD via an inflammatory mechanism, and 5) mechanistic evidence from animal models of maternal immune activation documenting behavioral and neural outcomes consistent with ADHD. Prenatal exposure to inflammation is associated with changes in offspring brain development including reductions in cortical gray matter volume and the volume of certain cortical areas -parallel to observations associated with ADHD. Alterations in neurotransmitter systems, including the dopaminergic, serotonergic and glutamatergic systems, are observed in ADHD populations. Animal models provide strong evidence that development and function of these neurotransmitters systems are sensitive to exposure to in utero inflammation. In summary, accumulating evidence from human studies and animal models, while still incomplete, support a potential role for neuroinflammation in the pathophysiology of ADHD. Confirmation of this association and the underlying mechanisms have become valuable targets for research. If confirmed, such a picture may be important in opening new intervention routes.
Collapse
Affiliation(s)
| | - Joel T Nigg
- Oregon Health and Science University, United States of America
| | - Elinor L Sullivan
- University of Oregon, United States of America; Oregon Health and Science University, United States of America; Oregon National Primate Research Center, United States of America.
| |
Collapse
|
44
|
Sex-Dependent Effects of Perinatal Inflammation on the Brain: Implication for Neuro-Psychiatric Disorders. Int J Mol Sci 2019; 20:ijms20092270. [PMID: 31071949 PMCID: PMC6539135 DOI: 10.3390/ijms20092270] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
Individuals born preterm have higher rates of neurodevelopmental disorders such as schizophrenia, autistic spectrum, and attention deficit/hyperactivity disorders. These conditions are often sexually dimorphic and with different developmental trajectories. The etiology is likely multifactorial, however, infections both during pregnancy and in childhood have emerged as important risk factors. The association between sex- and age-dependent vulnerability to neuropsychiatric disorders has been suggested to relate to immune activation in the brain, including complex interactions between sex hormones, brain transcriptome, activation of glia cells, and cytokine production. Here, we will review sex-dependent effects on brain development, including glia cells, both under normal physiological conditions and following perinatal inflammation. Emphasis will be given to sex-dependent effects on brain regions which play a role in neuropsychiatric disorders and inflammatory reactions that may underlie early-life programming of neurobehavioral disturbances later in life.
Collapse
|
45
|
Nelson LH, Saulsbery AI, Lenz KM. Small cells with big implications: Microglia and sex differences in brain development, plasticity and behavioral health. Prog Neurobiol 2019; 176:103-119. [PMID: 30193820 PMCID: PMC8008579 DOI: 10.1016/j.pneurobio.2018.09.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 07/17/2018] [Accepted: 09/01/2018] [Indexed: 12/20/2022]
Abstract
Brain sex differences are programmed largely by sex hormone secretions and direct sex chromosome effects in early life, and are subsequently modulated by early life experiences. The brain's resident immune cells, called microglia, actively contribute to brain development. Recent research has shown that microglia are sexually dimorphic, especially during early life, and may participate in sex-specific organization of the brain and behavior. Likewise, sex differences in immune cells and their signaling in the adult brain have been found, although in most cases their function remains unclear. Additionally, immune cells and their signaling have been implicated in many disorders in which brain development or plasticity is altered, including autism, schizophrenia, pain disorders, major depression, and postpartum depression. This review summarizes what is currently known about sex differences in neuroimmune function in development and during other major phases of brain plasticity, as well as the current state of knowledge regarding sex-specific neuroimmune function in psychiatric disorders.
Collapse
Affiliation(s)
- Lars H Nelson
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Angela I Saulsbery
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
46
|
Gandhi K, Montoya‐Uribe V, Martinez S, David S, Jain B, Shim G, Li C, Jenkins S, Nathanielsz P, Schlabritz‐Loutsevitch N. Ontogeny and programming of the fetal temporal cortical endocannabinoid system by moderate maternal nutrient reduction in baboons (Papio spp.). Physiol Rep 2019; 7:e14024. [PMID: 30912236 PMCID: PMC6434170 DOI: 10.14814/phy2.14024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 12/24/2022] Open
Abstract
Poor nutrition during pregnancy is a worldwide public health problem. Maternal nutrient reduction (MNR) is associated with maternal and fetal stress and a sex-dependent decrease in nonhuman primate (NHP) cognitive performance. Early life stress potentiates epileptogenesis in a sex-specific manner, and temporal lobe (TL) epilepsy is associated with neurocognitive disorders. The endogenous cannabinoid system (ECS) demonstrates remarkable developmental changes and plays a key role in aging-related diseases (e.g., dementia). Baboons have been studied as a natural model of epilepsy and express all ECS system components. We therefore evaluated baboon fetal temporal cortex ECS ontogenic and MNR-dependent changes. At 120 days gestational age (dGA) (term 185 days), maternal, fetal, and placental morphometry were similar between control and MNR pregnancies. MNR maternal weight gain was decreased compared with controls at 165 dGA independent of fetal sex. In male fetuses, expression of ECS synthesizing and degrading enzymes was gestational age-dependent, with the exception of fatty acid amide hydrolase (FAAH). MNR had a sex-specific effect on the protein expression of CB1R during development: CB1R protein expression was decreased in fetal temporal cortex of male fetuses at 120 and 140 dGA. Our data reveal that the MNR has sex-specific effects on temporal cortical expression of the ECS in baboon offspring and shows vulnerability of ECS in male fetuses during gestation.
Collapse
MESH Headings
- Amidohydrolases/genetics
- Amidohydrolases/metabolism
- Animal Nutritional Physiological Phenomena
- Animals
- Caloric Restriction
- Endocannabinoids/genetics
- Endocannabinoids/metabolism
- Female
- Fetal Development
- Gene Expression Regulation, Developmental
- Gene Expression Regulation, Enzymologic
- Gestational Age
- Male
- Maternal Nutritional Physiological Phenomena
- Papio
- Pregnancy
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/genetics
- Receptor, Cannabinoid, CB2/metabolism
- Sex Factors
- Signal Transduction
- Temporal Lobe/growth & development
- Temporal Lobe/metabolism
Collapse
Affiliation(s)
- Kushal Gandhi
- Department of Obstetrics and GynecologyTexas Tech University Health sciences Center at the Permian BasinOdessaTexas
| | | | - Stacy Martinez
- Department of Obstetrics and GynecologyTexas Tech University Health sciences Center at the Permian BasinOdessaTexas
| | - Samuel David
- Department of ChemistryUniversity of Texas at the Permian BasinOdessaTexas
| | - Bobby Jain
- Department of PsychiatryTexas Tech University Health Sciences Center at the Permian BasinOdessaTexas
| | - Grace Shim
- Department of Obstetrics and GynecologyTexas Tech University Health sciences Center at the Permian BasinOdessaTexas
| | - Cun Li
- University of WyomingLaramieWyoming
- Texas Biomedical Research InstituteSan AntonioTexas
| | - Susan Jenkins
- University of WyomingLaramieWyoming
- Texas Biomedical Research InstituteSan AntonioTexas
| | - Peter Nathanielsz
- University of WyomingLaramieWyoming
- Texas Biomedical Research InstituteSan AntonioTexas
| | - Natalia Schlabritz‐Loutsevitch
- Department of Obstetrics and GynecologyTexas Tech University Health sciences Center at the Permian BasinOdessaTexas
- Department of BiologyUniversity of Texas at the Permian BasinOdessaTexas
- Department of Neurobiology and PharmacologyTexas Tech University Health Sciences CenterLubbockTexas
| |
Collapse
|
47
|
Jahagirdar OB, Mittal AM, Song-Naba WL, Jha R, Kiven SB, Thompson ST, Connett JE, Gupta K. Diet and gender influence survival of transgenic Berkley sickle cell mice. Haematologica 2019; 104:e331-e334. [PMID: 30765475 DOI: 10.3324/haematol.2018.208322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Om B Jahagirdar
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Aditya M Mittal
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Waogwende L Song-Naba
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Ritu Jha
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Stacy B Kiven
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Susan T Thompson
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - John E Connett
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Kalpna Gupta
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| |
Collapse
|
48
|
Adverse neuropsychiatric development following perinatal brain injury: from a preclinical perspective. Pediatr Res 2019; 85:198-215. [PMID: 30367160 DOI: 10.1038/s41390-018-0222-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/11/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023]
Abstract
Perinatal brain injury is a leading cause of death and disability in young children. Recent advances in obstetrics, reproductive medicine and neonatal intensive care have resulted in significantly higher survival rates of preterm or sick born neonates, at the price of increased prevalence of neurological, behavioural and psychiatric problems in later life. Therefore, the current focus of experimental research shifts from immediate injury processes to the consequences for brain function in later life. The aetiology of perinatal brain injury is multi-factorial involving maternal and also labour-associated factors, including not only placental insufficiency and hypoxia-ischaemia but also exposure to high oxygen concentrations, maternal infection yielding excess inflammation, genetic factors and stress as important players, all of them associated with adverse long-term neurological outcome. Several animal models addressing these noxious stimuli have been established in the past to unravel the underlying molecular and cellular mechanisms of altered brain development. In spite of substantial efforts to investigate short-term consequences, preclinical evaluation of the long-term sequelae for the development of cognitive and neuropsychiatric disorders have rarely been addressed. This review will summarise and discuss not only current evidence but also requirements for experimental research providing a causal link between insults to the developing brain and long-lasting neurodevelopmental disorders.
Collapse
|
49
|
Tulina NM, Brown AG, Barila GO, Elovitz MA. The Absence of TLR4 Prevents Fetal Brain Injury in the Setting of Intrauterine Inflammation. Reprod Sci 2018; 26:1082-1093. [PMID: 30463495 DOI: 10.1177/1933719118805859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Exposure to intrauterine inflammation during pregnancy is linked to brain injury and neurobehavioral disorders in affected children. Innate immunity, specifically Toll-like receptor (TLR) signaling pathways are present throughout the reproductive tract as well as in the placenta, fetal membranes, and fetus. The TLR pathways are mechanistically involved in host responses to foreign pathogens and may lead to brain injury associated with prenatal inflammation. OBJECTIVE We aimed to determine whether the activation of the TLR4 signaling pathway, in the mother and fetus, is critical to fetal brain injury in the setting of intrauterine inflammation. METHODS A mini-laparotomy was performed on time pregnant C57B6 mice and 2 knockout mouse strains lacking the function of the Tlr4 and Myd88 genes on embryonic day 15. Intrauterine injections of Escherichia coli lipopolysaccharide or saline were administered as described previously. Dams were killed 6 hours postsurgery, and placental, amniotic fluid, and fetal brain tissue were collected. To assess brain injury, quantitative polymerase chain reaction (qPCR) analysis was performed on multiple components of the NOTCH signaling pathway, including Hes genes. Interleukin (IL) IL6, IL1β, and CCL5 expression was assessed using qPCR and enzyme-linked immunosorbent assay. RESULTS Using an established mouse model of intrauterine inflammation, we demonstrate that the abrogation of TLR4 signaling eliminates the cytokine response in mother and fetus and prevents brain injury associated with increased expression of transcriptional effectors of the NOTCH signaling pathway, Hes1 and Hes5. CONCLUSIONS These data show that the activation of the TLR4 signaling pathway is necessary for the development of fetal brain injury in response to intrauterine inflammation.
Collapse
Affiliation(s)
- Natalia M Tulina
- 1 Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy G Brown
- 1 Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guillermo O Barila
- 1 Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michal A Elovitz
- 1 Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
50
|
Gustafsson HC, Sullivan EL, Nousen EK, Sullivan CA, Huang E, Rincon M, Nigg JT, Loftis JM. Maternal prenatal depression predicts infant negative affect via maternal inflammatory cytokine levels. Brain Behav Immun 2018; 73:470-481. [PMID: 29920327 PMCID: PMC6129422 DOI: 10.1016/j.bbi.2018.06.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 12/29/2022] Open
Abstract
Maternal depressive symptoms during pregnancy are associated with risk for offspring emotional and behavioral problems, but the mechanisms by which this association occurs are not known. Infant elevated negative affect (increased crying, irritability, fearfulness, etc.) is a key risk factor for future psychopathology, so understanding its determinants has prevention and early intervention potential. An understudied yet promising hypothesis is that maternal mood affects infant mood via maternal prenatal inflammatory mechanisms, but this has not been prospectively examined in humans. Using data from a pilot study of women followed from the second trimester of pregnancy through six months postpartum (N = 68) our goal was to initiate a prospective study as to whether maternal inflammatory cytokines mediate the association between maternal depressive symptoms and infant offspring negative affect. The study sample was designed to examine a broad range of likely self-regulation and mood-regulation problems in offspring; to that end we over-selected women with a family history or their own history of elevated symptoms of attention-deficit/hyperactivity disorder. Results supported the hypothesis: maternal pro-inflammatory cytokines during the third trimester (indexed using a latent variable that included plasma interleukin-6, tumor necrosis factor-alpha and monocyte chemoattractant protein-1 concentrations as indicators) mediated the effect, such that higher maternal depressive symptoms were associated with higher maternal inflammation, and this mediated the effect on maternal report of infant negative affect (controlling for maternal affect during the infant period). This is the first human study to demonstrate that maternal inflammatory cytokines mediate the association between prenatal depression and infant outcomes, and the first to demonstrate a biological mechanism through which depressive symptoms impact infant temperament.
Collapse
Affiliation(s)
- Hanna C Gustafsson
- Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA.
| | - Elinor L Sullivan
- Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA; Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR, USA; University of Oregon, 1585 E 13th Ave, Eugene, OR, USA.
| | - Elizabeth K Nousen
- Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA.
| | - Ceri A Sullivan
- Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA.
| | - Elaine Huang
- Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, USA.
| | - Monica Rincon
- Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA.
| | - Joel T Nigg
- Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA.
| | - Jennifer M Loftis
- Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, USA.
| |
Collapse
|