1
|
Moreno RJ, Amara R, Ashwood P. Toward a better understanding of T cell dysregulation in autism: An integrative review. Brain Behav Immun 2024:S0889-1591(24)00649-4. [PMID: 39378971 DOI: 10.1016/j.bbi.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/28/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024] Open
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous disorder characterized by impairments in social, communicative, and restrictive behaviors. Over the past 20 years, research has highlighted the role of the immune system in regulating neurodevelopment and behavior. In ASD, immune abnormalities are frequently observed, such as elevations in pro-inflammatory cytokines, alterations in immune cell frequencies, and dysregulated mechanisms of immune suppression. The adaptive immune system - the branch of the immune system conferring cellular immunity - may be involved in the etiology of ASD. Specifically, dysregulated T cell activity, characterized by altered cellular function and increased cytokine release, presence of inflammatory phenotypes and altered cellular signaling, has been consistently observed in several studies across multiple laboratories and geographic regions. Similarly, mechanisms regulating their activation are also disrupted. T cells at homeostasis coordinate the healthy development of the central nervous system (CNS) during early prenatal and postnatal development, and aid in CNS maintenance into adulthood. Thus, T cell dysregulation may play a role in neurodevelopment and the behavioral and cognitive manifestations observed in ASD. Outside of the CNS, aberrant T cell activity may also be responsible for the increased frequency of immune based conditions in the ASD population, such as allergies, gut inflammation and autoimmunity. In this review, we will discuss the current understanding of T cell biology in ASD and speculate on mechanisms behind their dysregulation. This review also evaluates how aberrant T cell biology affects gastrointestinal issues and behavior in the context of ASD.
Collapse
Affiliation(s)
- R J Moreno
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA
| | - R Amara
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA
| | - P Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA.
| |
Collapse
|
2
|
Hofsink N, Groenink L, Plösch T. The fetal programming effect of maternal immune activation (MIA) on the offspring's immune system. Semin Immunopathol 2024; 46:14. [PMID: 39212791 PMCID: PMC11364800 DOI: 10.1007/s00281-024-01023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
The first 1000 days of life is a critical period of development in which adverse circumstances can have long-term consequences for the child's health. Maternal immune activation is associated with increased risk of neurodevelopmental disorders in the child. Aberrant immune responses have been reported in individuals with neurodevelopmental disorders. Moreover, lasting effects of maternal immune activation on the offspring's immune system have been reported. Taken together, this indicates that the effect of maternal immune activation is not limited to the central nervous system. Here, we explore the impact of maternal immune activation on the immune system of the offspring. We first describe the development of the immune system and provide an overview of reported alterations in the cytokine profiles, immune cell profiles, immune cell function, and immune induction in pre-clinical models. Additionally, we highlight recent research on the impact of maternal COVID-19 exposure on the neonatal immune system and the potential health consequences for the child. Our review shows that maternal immune activation alters the offspring's immune system under certain conditions, but the reported effects are conflicting and inconsistent. In general, epigenetic modifications are considered the mechanism for fetal programming. The available data was insufficient to identify specific pathways that may contribute to immune programming. As a consequence of the COVID-19 pandemic, more research now focuses on the possible health effects of maternal immune activation on the offspring. Future research addressing the offspring's immune response to maternal immune activation can elucidate specific pathways that contribute to fetal immune programming and the long-term health effects for the offspring.
Collapse
Affiliation(s)
- Naomi Hofsink
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Lucianne Groenink
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Carl von Ossietzky Universität Oldenburg School VI - School of Medicine and Health Sciences, Department of Paediatrics, Section of Neonatology, and Research Centre Neurosensory Science, Oldenburg, Germany
| |
Collapse
|
3
|
Hughes HK, Moreno RJ, Ashwood P. Innate Immune Dysfunction and Neuroinflammation in Autism Spectrum Disorder (ASD). FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2024; 22:229-241. [PMID: 38680981 PMCID: PMC11046725 DOI: 10.1176/appi.focus.24022004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder characterized by communication and social behavior deficits. The presence of restricted and repetitive behaviors often accompanies these deficits, and these characteristics can range from mild to severe. The past several decades have seen a significant rise in the prevalence of ASD. The etiology of ASD remains unknown; however, genetic and environmental risk factors play a role. Multiple hypotheses converge to suggest that neuroinflammation, or at least the interaction between immune and neural systems, may be involved in the etiology of some ASD cases or groups. Repeated evidence of innate immune dysfunction has been seen in ASD, often associated with worsening behaviors. This evidence includes data from circulating myeloid cells and brain resident macrophages/microglia in both human and animal models. This comprehensive review presents recent findings of innate immune dysfunction in ASD, including aberrant innate cellular function, evidence of neuroinflammation, and microglia activation. Appeared originally in Brain Behav Immun 2023; 108:245-254.
Collapse
Affiliation(s)
- H K Hughes
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| | - R J Moreno
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| | - P Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| |
Collapse
|
4
|
Jaswa EG, Cedars MI, Lindquist KJ, Bishop SL, Kim YS, Kaing A, Prahl M, Gaw SL, Corley J, Hoskin E, Cho YJ, Rogers E, Huddleston HG. In Utero Exposure to Maternal COVID-19 Vaccination and Offspring Neurodevelopment at 12 and 18 Months. JAMA Pediatr 2024; 178:258-265. [PMID: 38252445 PMCID: PMC10804280 DOI: 10.1001/jamapediatrics.2023.5743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 01/23/2024]
Abstract
Importance Uptake of COVID-19 vaccines among pregnant individuals was hampered by safety concerns around potential risks to unborn children. Data clarifying early neurodevelopmental outcomes of offspring exposed to COVID-19 vaccination in utero are lacking. Objective To determine whether in utero exposure to maternal COVID-19 vaccination was associated with differences in scores on the Ages and Stages Questionnaire, third edition (ASQ-3), at 12 and 18 months of age. Design, Setting, and Participants This prospective cohort study, Assessing the Safety of Pregnancy During the Coronavirus Pandemic (ASPIRE), enrolled pregnant participants from May 2020 to August 2021; follow-up of children from these pregnancies is ongoing. Participants, which included pregnant individuals and their offspring from all 50 states, self-enrolled online. Study activities were performed remotely. Exposure In utero exposure of the fetus to maternal COVID-19 vaccination during pregnancy was compared with those unexposed. Main Outcomes and Measures Neurodevelopmental scores on validated ASQ-3, completed by birth mothers at 12 and 18 months. A score below the established cutoff in any of 5 subdomains (communication, gross motor, fine motor, problem solving, social skills) constituted an abnormal screen for developmental delay. Results A total of 2487 pregnant individuals (mean [SD] age, 33.3 [4.2] years) enrolled at less than 10 weeks' gestation and completed research activities, yielding a total of 2261 and 1940 infants aged 12 and 18 months, respectively, with neurodevelopmental assessments. In crude analyses, 471 of 1541 exposed infants (30.6%) screened abnormally for developmental delay at 12 months vs 203 of 720 unexposed infants (28.2%; χ2 = 1.32; P = .25); the corresponding prevalences at 18 months were 262 of 1301 (20.1%) vs 148 of 639 (23.2%), respectively (χ2 = 2.35; P = .13). In multivariable mixed-effects logistic regression models adjusting for maternal age, race, ethnicity, education, income, maternal depression, and anxiety, no difference in risk for abnormal ASQ-3 screens was observed at either time point (12 months: adjusted risk ratio [aRR], 1.14; 95% CI, 0.97-1.33; 18 months: aRR, 0.88; 95% CI, 0.72-1.07). Further adjustment for preterm birth and infant sex did not affect results (12 months: aRR, 1.16; 95% CI, 0.98-1.36; 18 months: aRR, 0.87; 95% CI, 0.71-1.07). Conclusions and Relevance Results of this cohort study suggest that COVID-19 vaccination was safe during pregnancy from the perspective of infant neurodevelopment to 18 months of age. Additional longer-term research should be conducted to corroborate these findings and buttress clinical guidance with a strong evidence base.
Collapse
Affiliation(s)
- Eleni G. Jaswa
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Marcelle I. Cedars
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Karla J. Lindquist
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco
| | - Somer L. Bishop
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Young-Shin Kim
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Amy Kaing
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Mary Prahl
- Department of Pediatrics, Division of Pediatric Infectious Disease and Global Health, University of California, San Francisco, San Francisco
| | - Stephanie L. Gaw
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal Fetal Medicine, University of California, San Francisco, San Francisco
| | - Jamie Corley
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Elena Hoskin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Yoon Jae Cho
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Elizabeth Rogers
- Department of Pediatrics, Division of Neonatology, University of California, San Francisco, San Francisco
| | - Heather G. Huddleston
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| |
Collapse
|
5
|
Tamayo JM, Osman HC, Schwartzer JJ, Ashwood P. The influence of asthma on neuroinflammation and neurodevelopment: From epidemiology to basic models. Brain Behav Immun 2024; 116:218-228. [PMID: 38070621 DOI: 10.1016/j.bbi.2023.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/08/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
Asthma is a highly heterogeneous inflammatory disease that can have a significant effect on both the respiratory system and central nervous system. Population based studies and animal models have found asthma to be comorbid with a number of neurological conditions, including depression, anxiety, and neurodevelopmental disorders. In addition, maternal asthma during pregnancy has been associated with neurodevelopmental disorders in the offspring, such as autism spectrum disorders and attention deficit hyperactivity disorder. In this article, we review the most current epidemiological studies of asthma that identify links to neurological conditions, both as it relates to individuals that suffer from asthma and the impacts asthma during pregnancy may have on offspring neurodevelopment. We also discuss the relevant animal models investigating these links, address the gaps in knowledge, and explore the potential future directions in this field.
Collapse
Affiliation(s)
- Juan M Tamayo
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Hadley C Osman
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Jared J Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA.
| |
Collapse
|
6
|
Ashwood P. Though shall not pass: Blocking lymphocytes from recirculating to the gut as a potential therapeutic approach for autism. Brain Behav Immun 2024; 116:402-403. [PMID: 38157947 DOI: 10.1016/j.bbi.2023.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Affiliation(s)
- Paul Ashwood
- Department of Microbiology and Immunology, and the MIND Institute, 2805, 50th Street, Sacramento, CA 95817, United States.
| |
Collapse
|
7
|
Bryan EE, Bode NM, Chen X, Burris ES, Johnson DC, Dilger RN, Dilger AC. The effect of chronic, non-pathogenic maternal immune activation on offspring postnatal muscle and immune outcomes. J Anim Sci 2024; 102:skad424. [PMID: 38189595 PMCID: PMC10794819 DOI: 10.1093/jas/skad424] [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: 08/29/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024] Open
Abstract
The objective was to determine the effects of maternal inflammation on offspring muscle development and postnatal innate immune response. Sixteen first-parity gilts were randomly allotted to repeated intravenous injections with lipopolysaccharide (LPS; n = 8, treatment code INFLAM) or comparable volume of phosphate buffered saline (CON, n = 8). Injections took place every other day from gestational day (GD) 70 to GD 84 with an initial dose of 10 μg LPS/kg body weight (BW) increasing by 12% each time to prevent endotoxin tolerance. On GD 70, 76, and 84, blood was collected at 0 and 4 h postinjection via jugular or ear venipuncture to determine tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β concentrations. After farrowing, litter mortality was recorded, and the pig closest to litter BW average was used for dissection and muscle fiber characterization. On weaning (postnatal day [PND] 21), pigs were weighed individually and 2 barrows closest to litter BW average were selected for another study. The third barrow closest to litter BW average was selected for the postnatal LPS challenge. On PND 52, pigs were given 5 μg LPS/kg BW via intraperitoneal injection, and blood was collected at 0, 4, and 8 h postinjection to determine TNF-α concentration. INFLAM gilt TNF-α concentration increased (P < 0.01) 4 h postinjection compared to 0 h postinjection, while CON gilt TNF-α concentration did not differ between time points. INFLAM gilt IL-6 and IL-1β concentrations increased (P = 0.03) 4 h postinjection compared to 0 h postinjection on GD 70, but did not differ between time points on GD 76 and 84. There were no differences between INFLAM and CON gilts litter mortality outcomes (P ≥ 0.13), but INFLAM pigs were smaller (P = 0.04) at birth and tended (P = 0.09) to be smaller at weaning. Muscle and organ weights did not differ (P ≥ 0.17) between treatments, with the exception of semitendinosus, which was smaller (P < 0.01) in INFLAM pigs. INFLAM pigs tended (P = 0.06) to have larger type I fibers. INFLAM pig TNF-α concentration did not differ across time, while CON pig TNF-α concentration peaked (P = 0.01) 4 h postinjection. TNF-α concentration did not differ between treatments at 0 and 8 h postinjection, but CON pigs had increased (P = 0.01) TNF-α compared to INFLAM pigs 4 h postinjection. Overall, maternal immune activation did not alter pig muscle development, but resulted in suppressed innate immune activation.
Collapse
Affiliation(s)
- Erin E Bryan
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Nick M Bode
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Xuenan Chen
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Elli S Burris
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Danielle C Johnson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Anna C Dilger
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| |
Collapse
|
8
|
Croen LA, Ames JL, Qian Y, Alexeeff S, Ashwood P, Gunderson EP, Wu YW, Boghossian AS, Yolken R, Van de Water J, Weiss LA. Inflammatory Conditions During Pregnancy and Risk of Autism and Other Neurodevelopmental Disorders. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:39-50. [PMID: 38045769 PMCID: PMC10689278 DOI: 10.1016/j.bpsgos.2023.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 12/05/2023] Open
Abstract
Background Maternal inflammation can result from immune dysregulation and metabolic perturbations during pregnancy. Whether conditions associated with inflammation during pregnancy increase the likelihood of autism spectrum disorder (ASD) or other neurodevelopmental disorders (DDs) is not well understood. Methods We conducted a case-control study among children born in California from 2011 to 2016 to investigate maternal immune-mediated and cardiometabolic conditions during pregnancy and risk of ASD (n = 311) and DDs (n = 1291) compared with children from the general population (n = 967). Data on maternal conditions and covariates were retrieved from electronic health records. Maternal genetic data were used to assess a causal relationship. Results Using multivariable logistic regression, we found that mothers with asthma were more likely to deliver infants later diagnosed with ASD (odds ratio [OR] = 1.62, 95% CI: 1.15-2.29) or DDs (OR = 1.30, 95% CI: 1.02-1.64). Maternal obesity was also associated with child ASD (OR = 1.51, 95% CI: 1.07-2.13). Mothers with both asthma and extreme obesity had the greatest odds of delivering an infant later diagnosed with ASD (OR = 16.9, 95% CI: 5.13-55.71). These increased ASD odds were observed among female children only. Polygenic risk scores for obesity, asthma, and their combination showed no association with ASD risk. Mendelian randomization did not support a causal relationship between maternal conditions and ASD. Conclusions Inflammatory conditions during pregnancy are associated with risk for neurodevelopmental disorders in children. These risks do not seem to be due to shared genetic risk; rather, inflammatory conditions may share nongenetic risk factors with neurodevelopmental disorders. Children whose mothers have both asthma and obesity during pregnancy may benefit from earlier screening and intervention.
Collapse
Affiliation(s)
- Lisa A. Croen
- Division of Research, Kaiser Permanente Northern California, Oakland, California
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California
| | - Jennifer L. Ames
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Yinge Qian
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Stacey Alexeeff
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology and the MIND Institute, University of California, Davis, Davis, California
| | - Erica P. Gunderson
- Division of Research, Kaiser Permanente Northern California, Oakland, California
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California
| | - Yvonne W. Wu
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, California
| | - Andrew S. Boghossian
- Institute for Human Genetics, Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| | - Robert Yolken
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Judy Van de Water
- Division of Rheumatology/Allergy/Clinical Immunology, Department of Internal Medicine, University of California at Davis, Davis, California
| | - Lauren A. Weiss
- Institute for Human Genetics, Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| |
Collapse
|
9
|
Mostallino R, Santoni M, Sagheddu C, Serra V, Orrù V, Pistis M, Castelli MP. The PPARα agonist fenofibrate reduces the cytokine imbalance in a maternal immune activation model of schizophrenia. Eur J Pharmacol 2023; 961:176172. [PMID: 37939988 DOI: 10.1016/j.ejphar.2023.176172] [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: 07/28/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Maternal infections during pregnancy may increase the risk of psychiatric disorders in offspring. We recently demonstrated that activation of peroxisome proliferator-activate receptor-α (PPARα), with the clinically available agonist fenofibrate (FEN), attenuates the neurodevelopmental disturbances induced by maternal immune activation (MIA) in rat offspring. We hypothesized that fenofibrate might reduce MIA-induced cytokine imbalance using a MIA model based on the viral mimetic polyriboinosinic-polyribocytidilic acid [poly (I:C)]. By using the Bio-Plex Multiplex-Immunoassay-System, we measured cytokine/chemokine/growth factor levels in maternal serum and in the fetal brain of rats treated with fenofibrate, at 6 and 24 h after poly (I:C). We found that MIA induced time-dependent changes in the levels of several cytokines/chemokines/colony-stimulating factors (CSFs). Specifically, the maternal serum of the poly (I:C)/control (CTRL) group showed increased levels of (i) proinflammatory chemokine macrophage inflammatory protein 1-alpha (MIP-1α), (ii) tumor necrosis factor-alpha (TNF-α), the monocyte chemoattractant protein-1 (MCP-1), the macrophage (M-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Conversely, in the fetal brain of the poly (I:C)/CTRL group, interleukin 12p70 and MIP-1α levels were lower than in vehicle (veh)/CTRL group. Notably, MIP-1α, TNF-α, keratinocyte derived chemokine (GRO/KC), GM-CSF, and M-CSF levels were lower in the poly (I:C)/FEN than in poly (I:C)/CTRL rats, suggesting the protective role of the PPARα agonist. PPARα might represent a therapeutic target to attenuate MIA-induced inflammation.
Collapse
Affiliation(s)
- Rafaela Mostallino
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Michele Santoni
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Valentina Serra
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; Neuroscience Institute, National Research Council of Italy, Section of Cagliari, Italy; Unit of Clinical Pharmacology, University Hospital, Cagliari, Italy.
| | - M Paola Castelli
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy.
| |
Collapse
|
10
|
Sharova V, Ignatiuk V, Izvolskaia M, Zakharova L. Disruption of Intranasal GnRH Neuronal Migration Route into the Brain Induced by Proinflammatory Cytokine IL-6: Ex Vivo and In Vivo Rodent Models. Int J Mol Sci 2023; 24:15983. [PMID: 37958965 PMCID: PMC10648422 DOI: 10.3390/ijms242115983] [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: 10/11/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Maternal immune activation results in altered levels of cytokines in the maternal-fetal system, which has a negative impact on fetal development, including the gonadotropin-releasing hormone (GnRH) system, which is crucial for the reproduction. Suppression of GnRH-neuron migration may be associated with cytokine imbalances, and primarily with proinflammatory cytokine interleukin (IL)-6. This study aimed to determine the effects of IL-6 and monoclonal antibody to IL-6 or IL-6R or polyclonal IgG on the formation of migration route of GnRH-neurons in ex vivo and in vivo rodent models on day 11.5 of embryonic development. The increased level of IL-6 in mouse nasal explants suppressed peripherin-positive fiber outgrowth, while this led to an increase in the number of GnRH-neurons in the nose and olfactory bulbs and a decrease in their number in the fetal brain. This effect is likely to be realized via IL-6 receptors along the olfactory nerves. The suppressive effect of IL-6 was diminished by monoclonal antibodies to IL-6 or its receptors and by IgG.
Collapse
Affiliation(s)
- Viktoria Sharova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov Street, 26, 119334 Moscow, Russia
| | | | | | | |
Collapse
|
11
|
Chen R, Routh BN, Gaudet AD, Fonken LK. Circadian Regulation of the Neuroimmune Environment Across the Lifespan: From Brain Development to Aging. J Biol Rhythms 2023; 38:419-446. [PMID: 37357738 PMCID: PMC10475217 DOI: 10.1177/07487304231178950] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Circadian clocks confer 24-h periodicity to biological systems, to ultimately maximize energy efficiency and promote survival in a world with regular environmental light cycles. In mammals, circadian rhythms regulate myriad physiological functions, including the immune, endocrine, and central nervous systems. Within the central nervous system, specialized glial cells such as astrocytes and microglia survey and maintain the neuroimmune environment. The contributions of these neuroimmune cells to both homeostatic and pathogenic demands vary greatly across the day. Moreover, the function of these cells changes across the lifespan. In this review, we discuss circadian regulation of the neuroimmune environment across the lifespan, with a focus on microglia and astrocytes. Circadian rhythms emerge in early life concurrent with neuroimmune sculpting of brain circuits and wane late in life alongside increasing immunosenescence and neurodegeneration. Importantly, circadian dysregulation can alter immune function, which may contribute to susceptibility to neurodevelopmental and neurodegenerative diseases. In this review, we highlight circadian neuroimmune interactions across the lifespan and share evidence that circadian dysregulation within the neuroimmune system may be a critical component in human neurodevelopmental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruizhuo Chen
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Brandy N. Routh
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Andrew D. Gaudet
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
- Department of Psychology, The University of Texas at Austin, Austin, Texas
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Laura K. Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
12
|
Lesh TA, Iosif AM, Tanase C, Vlasova RM, Ryan AM, Bennett J, Hogrefe CE, Maddock RJ, Geschwind DH, Van de Water J, McAllister AK, Styner MA, Bauman MD, Carter CS. Extracellular free water elevations are associated with brain volume and maternal cytokine response in a longitudinal nonhuman primate maternal immune activation model. Mol Psychiatry 2023; 28:4185-4194. [PMID: 37582858 PMCID: PMC10867284 DOI: 10.1038/s41380-023-02213-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/21/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
Maternal infection has emerged as an important environmental risk factor for neurodevelopmental disorders, including schizophrenia and autism spectrum disorders. Animal model systems of maternal immune activation (MIA) suggest that the maternal immune response plays a significant role in the offspring's neurodevelopment and behavioral outcomes. Extracellular free water is a measure of freely diffusing water in the brain that may be associated with neuroinflammation and impacted by MIA. The present study evaluates the brain diffusion characteristics of male rhesus monkeys (Macaca mulatta) born to MIA-exposed dams (n = 14) treated with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the end of the first trimester (n = 10) or were untreated (n = 4). Offspring underwent diffusion MRI scans at 6, 12, 24, 36, and 45 months. Offspring born to MIA-exposed dams showed significantly increased extracellular free water in cingulate cortex gray matter starting as early as 6 months of age and persisting through 45 months. In addition, offspring gray matter free water in this region was significantly correlated with the magnitude of the maternal IL-6 response in the MIA-exposed dams. Significant correlations between brain volume and extracellular free water in the MIA-exposed offspring also indicate converging, multimodal evidence of the impact of MIA on brain development. These findings provide strong evidence for the construct validity of the nonhuman primate MIA model as a system of relevance for investigating the pathophysiology of human neurodevelopmental psychiatric disorders. Elevated free water in individuals exposed to immune activation in utero could represent an early marker of a perturbed or vulnerable neurodevelopmental trajectory.
Collapse
Affiliation(s)
- Tyler A Lesh
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Ana-Maria Iosif
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Costin Tanase
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Roza M Vlasova
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- California National Primate Research Center, Davis, CA, USA
| | - Jeffrey Bennett
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | | | - Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Judy Van de Water
- MIND Institute, University of California, Davis, CA, USA
- Rheumatology/Allergy and Clinical Immunology, University of California, Davis, CA, USA
| | - A Kimberley McAllister
- MIND Institute, University of California, Davis, CA, USA
- Center for Neuroscience, University of California, Davis, CA, USA
| | - Martin A Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- California National Primate Research Center, Davis, CA, USA
| | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA.
| |
Collapse
|
13
|
Sager REH, Walker AK, Middleton FA, Robinson K, Webster MJ, Gentile K, Wong ML, Shannon Weickert C. Changes in cytokine and cytokine receptor levels during postnatal development of the human dorsolateral prefrontal cortex. Brain Behav Immun 2023; 111:186-201. [PMID: 36958512 DOI: 10.1016/j.bbi.2023.03.015] [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: 09/30/2022] [Revised: 03/09/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023] Open
Abstract
In addition to their traditional roles in immune cell communication, cytokines regulate brain development. Cytokines are known to influence neural cell generation, differentiation, maturation, and survival. However, most work on the role of cytokines in brain development investigates rodents or focuses on prenatal events. Here, we investigate how mRNA and protein levels of key cytokines and cytokine receptors change during postnatal development of the human prefrontal cortex. We find that most cytokine transcripts investigated (IL1B, IL18, IL6, TNF, IL13) are lowest at birth and increase between 1.5 and 5 years old. After 5 years old, transcriptional patterns proceeded in one of two directions: decreased expression in teens and young adults (IL1B, p = 0.002; and IL18, p = 0.004) or increased mean expression with maturation, particularly in teenagers (IL6, p = 0.004; TNF, p = 0.002; IL13, p < 0.001). In contrast, cytokine proteins tended to remain elevated after peaking significantly around 3 years of age (IL1B, p = 0.012; IL18, p = 0.026; IL6, p = 0.039; TNF, p < 0.001), with TNF protein being highest in teenagers. An mRNA-only analysis of cytokine receptor transcripts found that early developmental increases in cytokines were paralleled by increases in their ligand-binding receptor subunits, such as IL1R1 (p = 0.033) and IL6R (p < 0.001) transcripts. In contrast, cytokine receptor-associated signaling subunits, IL1RAP and IL6ST, did not change significantly between age groups. Of the two TNF receptors, the 'pro-death' TNFRSF1A and 'pro-survival' TNFRSF1B, only TNFRSF1B was significantly changed (p = 0.028), increasing first in toddlers and again in young adults. Finally, the cytokine inhibitor, IL13, was elevated first in toddlers (p = 0.006) and again in young adults (p = 0.053). While the mean expression of interleukin-1 receptor antagonist (IL1RN) was highest in toddlers, this increase was not statistically significant. The fluctuations in cytokine expression reported here support a role for increases in specific cytokines at two different stages of human cortical development. The first is during the toddler/preschool period (IL1B, IL18, and IL13), and the other occurs at adolescence/young adult maturation (IL6, TNF and IL13).
Collapse
Affiliation(s)
- Rachel E H Sager
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Adam K Walker
- Laboratory of Immunopsychiatry, Neuroscience Research Australia, Sydney, NSW, Australia; Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia; Monash Institute of Pharmaceutical Science, Monash University, Parkville, VIC, Australia
| | - Frank A Middleton
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Kate Robinson
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
| | | | - Karen Gentile
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Ma-Li Wong
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA; Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Cynthia Shannon Weickert
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA; Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia.
| |
Collapse
|
14
|
Li Q, Li S, Yao Y, Ma Z, Huang C. MIA mice exhibit enteric nerve defects and are more susceptible to dextran sulfate sodium-induced colitis. Brain Behav Immun 2023:S0889-1591(23)00158-7. [PMID: 37315701 DOI: 10.1016/j.bbi.2023.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/15/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023] Open
Abstract
Maternal immune activation (MIA) during pregnancy impairs the development of the central nervous system as well as the peripheral nervous system. Emerging evidence indicates that individuals with MIA suffer more from gastrointestinal disorders. The present study aims to test the hypothesis that MIA-induced susceptibility to inflammatory bowel disease is due to defects in the innervation of mucosal sensory nerves. Acute dextran sulfate sodium (DSS) colitis was induced in MIA and control adult mice. Body weight loss, disease activity index and colonic histological changes were measured during colitis. The study found that MIA mice were hypersusceptible to DSS-induced colitis and that macrophage infiltration and cytokine production were elevated in the colon of MIA mice. In vitro experiments also demonstrated that colonic macrophages from MIA mice presented hyperinflammatory responses to LPS stimulation. Sensory nerve-secreted calcitonin gene-related peptide (CGRP) is an important neuropeptide in modulating enteric inflammation. Intriguingly, we found that CGRP-positive nerves were sparsely distributed in the colon of MIA mice regardless of DSS treatment. And the protein level of CGRP was significantly reduced in colon of MIA mice. However, there was no decrease in the number of CGRP-positive cell bodies in either the DRG or vagal ganglion, suggesting that innervation defects of CGRP mucosal sensory nerves exist in the colon of MIA mice. Critically, administration of recombinant CGRP to MIA mice during DSS colitis significantly reversed their hyperinflammatory pathology. Additionally, the hyperinflammatory phenotype of colonic macrophages of MIA mice could also be reversed by CGRP treatment in vitro. Collectively, these findings suggested that the sensor nerve innervation defect-induced CGRP deficiency in MIA mice participates in their increased susceptibility to colitis. Thus, sensor nerve-secreted CGRP may be a new therapeutic target for autism combined with inflammatory bowel disease.
Collapse
Affiliation(s)
- Qian Li
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Shuang Li
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yiwei Yao
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhongxiang Ma
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chutian Huang
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
15
|
Smucny J, Vlasova RM, Lesh TA, Rowland DJ, Wang G, Chaudhari AJ, Chen S, Iosif AM, Hogrefe CE, Bennett JL, Shumann CM, Van de Water JA, Maddock RJ, Styner MA, Geschwind DH, McAllister AK, Bauman MD, Carter CS. Increased Striatal Presynaptic Dopamine in a Nonhuman Primate Model of Maternal Immune Activation: A Longitudinal Neurodevelopmental Positron Emission Tomography Study With Implications for Schizophrenia. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2023; 8:505-513. [PMID: 36805246 PMCID: PMC10164700 DOI: 10.1016/j.bpsc.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Epidemiological studies suggest that maternal immune activation (MIA) is a significant risk factor for future neurodevelopmental disorders, including schizophrenia (SZ), in offspring. Consistent with findings in SZ research and work in rodent systems, preliminary cross-sectional findings in nonhuman primates suggest that MIA is associated with dopaminergic hyperfunction in young adult offspring. METHODS In this unique prospective longitudinal study, we used [18F]fluoro-l-m-tyrosine positron emission tomography to examine the developmental time course of striatal presynaptic dopamine synthesis in male rhesus monkeys born to dams (n = 13) injected with a modified form of the inflammatory viral mimic, polyinosinic:polycytidylic acid [poly(I:C)], in the late first trimester. Striatal (caudate, putamen, and nucleus accumbens) dopamine from these animals was compared with that of control offspring born to dams that received saline (n = 10) or no injection (n = 4). Dopamine was measured at 15, 26, 38, and 48 months of age. Prior work with this cohort found decreased prefrontal gray matter volume in MIA offspring versus controls between 6 and 45 months of age. Based on theories of the etiology and development of SZ-related pathology, we hypothesized that there would be a delayed (relative to the gray matter decrease) increase in striatal fluoro-l-m-tyrosine signal in the MIA group versus controls. RESULTS [18F]fluoro-l-m-tyrosine signal showed developmental increases in both groups in the caudate and putamen. Group comparisons revealed significantly greater caudate dopaminergic signal in the MIA group at 26 months. CONCLUSIONS These findings are highly relevant to the known pathophysiology of SZ and highlight the translational relevance of the MIA model in understanding mechanisms by which MIA during pregnancy increases risk for later illness in offspring.
Collapse
Affiliation(s)
- Jason Smucny
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California.
| | - Roza M Vlasova
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Tyler A Lesh
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California; Center for Neuroscience, University of California, Davis, California
| | - Douglas J Rowland
- Center for Genomic and Molecular Imaging, University of California, Davis, California
| | - Guobao Wang
- Department of Radiology, University of California, Davis, California
| | - Abhijit J Chaudhari
- Center for Genomic and Molecular Imaging, University of California, Davis, California; Department of Radiology, University of California, Davis, California
| | - Shuai Chen
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, California
| | - Ana-Maria Iosif
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, California
| | - Casey E Hogrefe
- California National Primate Research Center, University of California, Davis, California
| | - Jeffrey L Bennett
- Department of Psychology, University of California, Davis, California
| | - Cynthia M Shumann
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California
| | - Judy A Van de Water
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, California
| | - Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California
| | - Martin A Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina; Department of Computer Science, University of North Carolina, Chapel Hill, North Carolina
| | - Daniel H Geschwind
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
| | | | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California; California National Primate Research Center, University of California, Davis, California
| | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California; Center for Neuroscience, University of California, Davis, California.
| |
Collapse
|
16
|
Hanson KL, Weir RK, Iosif AM, Van de Water J, Carter CS, McAllister AK, Bauman MD, Schumann CM. Altered dendritic morphology in dorsolateral prefrontal cortex of nonhuman primates prenatally exposed to maternal immune activation. Brain Behav Immun 2023; 109:92-101. [PMID: 36610487 PMCID: PMC10023379 DOI: 10.1016/j.bbi.2023.01.003] [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: 08/23/2022] [Revised: 12/06/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Women who contract a viral or bacterial infection during pregnancy have an increased risk of giving birth to a child with a neurodevelopmental or psychiatric disorder. The effects of maternal infection are likely mediated by the maternal immune response, as preclinical animal models have confirmed that maternal immune activation (MIA) leads to long lasting changes in offspring brain and behavior development. The present study sought to determine the impact of MIA-exposure during the first or second trimester on neuronal morphology in dorsolateral prefrontal cortex (DLPFC) and hippocampus from brain tissue obtained from MIA-exposed and control male rhesus monkey (Macaca mulatta) during late adolescence. MIA-exposed offspring display increased neuronal dendritic branching in pyramidal cells in DLPFC infra- and supragranular layers relative to controls, with no significant differences observed between offspring exposed to maternal infection in the first and second trimester. In addition, the diameter of apical dendrites in DLPFC infragranular layer is significantly decreased in MIA-exposed offspring relative to controls, irrespective of trimester exposure. In contrast, alterations in hippocampal neuronal morphology of MIA-exposed offspring were not evident. These findings demonstrate that a maternal immune challenge during pregnancy has long-term consequences for primate offspring dendritic structure, selectively in a brain region vital for socioemotional and cognitive development.
Collapse
Affiliation(s)
- Kari L Hanson
- Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, United States; MIND Institute, University of California, Davis, United States
| | - Ruth K Weir
- Innovation & Enterprise Department, University College London, United Kingdom
| | - Ana-Maria Iosif
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, United States
| | - Judy Van de Water
- MIND Institute, University of California, Davis, United States; Rheumatology/Allergy and Clinical Immunology, University of California, Davis, United States
| | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, United States; Center for Neuroscience, University of California, Davis, United States
| | | | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, United States; MIND Institute, University of California, Davis, United States; California National Primate Research Center, University of California, Davis, United States.
| | - Cynthia M Schumann
- Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, United States; MIND Institute, University of California, Davis, United States.
| |
Collapse
|
17
|
Influence of Immune System Abnormalities Caused by Maternal Immune Activation in the Postnatal Period. Cells 2023; 12:cells12050741. [PMID: 36899877 PMCID: PMC10001371 DOI: 10.3390/cells12050741] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
The developmental origins of health and disease (DOHaD) indicate that fetal tissues and organs in critical and sensitive periods of development are susceptible to structural and functional changes due to the adverse environment in utero. Maternal immune activation (MIA) is one of the phenomena in DOHaD. Exposure to maternal immune activation is a risk factor for neurodevelopmental disorders, psychosis, cardiovascular diseases, metabolic diseases, and human immune disorders. It has been associated with increased levels of proinflammatory cytokines transferred from mother to fetus in the prenatal period. Abnormal immunity induced by MIA includes immune overreaction or immune response failure in offspring. Immune overreaction is a hypersensitivity response of the immune system to pathogens or allergic factor. Immune response failure could not properly fight off various pathogens. The clinical features in offspring depend on the gestation period, inflammatory magnitude, inflammatory type of MIA in the prenatal period, and exposure to prenatal inflammatory stimulation, which might induce epigenetic modifications in the immune system. An analysis of epigenetic modifications caused by adverse intrauterine environments might allow clinicians to predict the onset of diseases and disorders before or after birth.
Collapse
|
18
|
Innate immune dysfunction and neuroinflammation in autism spectrum disorder (ASD). Brain Behav Immun 2023; 108:245-254. [PMID: 36494048 DOI: 10.1016/j.bbi.2022.12.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/21/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder characterized by communication and social behavior deficits. The presence of restricted and repetitive behaviors often accompanies these deficits, and these characteristics can range from mild to severe. The past several decades have seen a significant rise in the prevalence of ASD. The etiology of ASD remains unknown; however, genetic and environmental risk factors play a role. Multiple hypotheses converge to suggest that neuroinflammation, or at least the interaction between immune and neural systems, may be involved in the etiology of some ASD cases or groups. Repeated evidence of innate immune dysfunction has been seen in ASD, often associated with worsening behaviors. This evidence includes data from circulating myeloid cells and brain resident macrophages/microglia in both human and animal models. This comprehensive review presents recent findings of innate immune dysfunction in ASD, including aberrant innate cellular function, evidence of neuroinflammation, and microglia activation.
Collapse
|
19
|
Lampiasi N, Bonaventura R, Deidda I, Zito F, Russo R. Inflammation and the Potential Implication of Macrophage-Microglia Polarization in Human ASD: An Overview. Int J Mol Sci 2023; 24:2703. [PMID: 36769026 PMCID: PMC9916462 DOI: 10.3390/ijms24032703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous collection of neurodevelopmental disorders, difficult to diagnose and currently lacking treatment options. The possibility of finding reliable biomarkers useful for early identification would offer the opportunity to intervene with treatment strategies to improve the life quality of ASD patients. To date, there are many recognized risk factors for the development of ASD, both genetic and non-genetic. Although genetic and epigenetic factors may play a critical role, the extent of their contribution to ASD risk is still under study. On the other hand, non-genetic risk factors include pollution, nutrition, infection, psychological states, and lifestyle, all together known as the exposome, which impacts the mother's and fetus's life, especially during pregnancy. Pathogenic and non-pathogenic maternal immune activation (MIA) and autoimmune diseases can cause various alterations in the fetal environment, also contributing to the etiology of ASD in offspring. Activation of monocytes, macrophages, mast cells and microglia and high production of pro-inflammatory cytokines are indeed the cause of neuroinflammation, and the latter is involved in ASD's onset and development. In this review, we focused on non-genetic risk factors, especially on the connection between inflammation, macrophage polarization and ASD syndrome, MIA, and the involvement of microglia.
Collapse
Affiliation(s)
- Nadia Lampiasi
- Istituto per la Ricerca e l’Innovazione Biomedica IRIB, Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | | | | | | | | |
Collapse
|
20
|
Boktor JC, Adame MD, Rose DR, Schumann CM, Murray KD, Bauman MD, Careaga M, Mazmanian SK, Ashwood P, Needham BD. Global metabolic profiles in a non-human primate model of maternal immune activation: implications for neurodevelopmental disorders. Mol Psychiatry 2022; 27:4959-4973. [PMID: 36028571 PMCID: PMC9772216 DOI: 10.1038/s41380-022-01752-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 01/14/2023]
Abstract
Epidemiological evidence implicates severe maternal infections as risk factors for neurodevelopmental disorders, such as ASD and schizophrenia. Accordingly, animal models mimicking infection during pregnancy, including the maternal immune activation (MIA) model, result in offspring with neurobiological, behavioral, and metabolic phenotypes relevant to human neurodevelopmental disorders. Most of these studies have been performed in rodents. We sought to better understand the molecular signatures characterizing the MIA model in an organism more closely related to humans, rhesus monkeys (Macaca mulatta), by evaluating changes in global metabolic profiles in MIA-exposed offspring. Herein, we present the global metabolome in six peripheral tissues (plasma, cerebrospinal fluid, three regions of intestinal mucosa scrapings, and feces) from 13 MIA and 10 control offspring that were confirmed to display atypical neurodevelopment, elevated immune profiles, and neuropathology. Differences in lipid, amino acid, and nucleotide metabolism discriminated these MIA and control samples, with correlations of specific metabolites to behavior scores as well as to cytokine levels in plasma, intestinal, and brain tissues. We also observed modest changes in fecal and intestinal microbial profiles, and identify differential metabolomic profiles within males and females. These findings support a connection between maternal immune activation and the metabolism, microbiota, and behavioral traits of offspring, and may further the translational applications of the MIA model and the advancement of biomarkers for neurodevelopmental disorders such as ASD or schizophrenia.
Collapse
Affiliation(s)
- Joseph C Boktor
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Mark D Adame
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Destanie R Rose
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Cynthia M Schumann
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Karl D Murray
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Melissa D Bauman
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Sarkis K Mazmanian
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA.
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA.
| | - Brittany D Needham
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
21
|
Ni MZ, Zhang YM, Li Y, Wu QT, Zhang ZZ, Chen J, Luo BL, Li XW, Chen GH. Environmental enrichment improves declined cognition induced by prenatal inflammatory exposure in aged CD-1 mice: Role of NGPF2 and PSD-95. Front Aging Neurosci 2022; 14:1021237. [PMID: 36479357 PMCID: PMC9720164 DOI: 10.3389/fnagi.2022.1021237] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/03/2022] [Indexed: 12/08/2023] Open
Abstract
INTRODUCTION Research suggests that prenatal inflammatory exposure could accelerate age-related cognitive decline that may be resulted from neuroinflammation and synaptic dysfunction during aging. Environmental enrichment (EE) may mitigate the cognitive and synaptic deficits. Neurite growth-promoting factor 2 (NGPF2) and postsynaptic density protein 95 (PSD-95) play critical roles in neuroinflammation and synaptic function, respectively. METHODS We examined whether this adversity and EE exposure can cause alterations in Ngpf2 and Psd-95 expression. In this study, CD-1 mice received intraperitoneal injection of lipopolysaccharide (50 μg/kg) or normal saline from gestational days 15-17. After weaning, half of the male offspring under each treatment were exposed to EE. The Morris water maze was used to assess spatial learning and memory at 3 and 15 months of age, whereas quantitative real-time polymerase chain reaction and Western blotting were used to measure hippocampal mRNA and protein levels of NGPF2 and PSD-95, respectively. Meanwhile, serum levels of IL-6, IL-1β, and TNF-α were determined by enzyme-linked immunosorbent assay. RESULTS The results showed that aged mice exhibited poor spatial learning and memory ability, elevated NGPF2 mRNA and protein levels, and decreased PSD-95 mRNA and protein levels relative to their young counterparts during natural aging. Embryonic inflammatory exposure accelerated age-related changes in spatial cognition, and in Ngpf2 and Psd-95 expression. Additionally, the levels of Ngpf2 and Psd-95 products were significantly positively and negatively correlated with cognitive dysfunction, respectively, particularly in prenatal inflammation-exposed aged mice. Changes in serum levels of IL-6, IL-1β, and TNF-α reflective of systemic inflammation and their correlation with cognitive decline during accelerated aging were similar to those of hippocampal NGPF2. EE exposure could partially restore the accelerated decline in age-related cognitive function and in Psd-95 expression, especially in aged mice. DISCUSSION Overall, the aggravated cognitive disabilities in aged mice may be related to the alterations in Ngpf2 and Psd-95 expression and in systemic state of inflammation due to prenatal inflammatory exposure, and long-term EE exposure may ameliorate this cognitive impairment by upregulating Psd-95 expression.
Collapse
Affiliation(s)
- Ming-Zhu Ni
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Yue-Ming Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Yun Li
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Qi-Tao Wu
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Zhe-Zhe Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Jing Chen
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Bao-Ling Luo
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Xue-Wei Li
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
22
|
The role of maternal immune activation in the immunological and neurological pathogenesis of autism. JOURNAL OF NEURORESTORATOLOGY 2022. [DOI: 10.1016/j.jnrt.2022.100030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
23
|
Guedes JR, Ferreira PA, Costa JM, Cardoso AL, Peça J. Microglia-dependent remodeling of neuronal circuits. J Neurochem 2022; 163:74-93. [PMID: 35950924 PMCID: PMC9826178 DOI: 10.1111/jnc.15689] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 01/11/2023]
Abstract
Microglia are tissue-resident macrophages responsible for the surveillance, neuronal support, and immune defense of the brain parenchyma. Recently, the role played by microglia in the formation and function of neuronal circuits has garnered substantial attention. During development, microglia have been shown to engulf neuronal precursors and participate in pruning mechanisms while, in the mature brain, they influence synaptic signaling, provide trophic support and shape synaptic plasticity. Recently, studies have unveiled different microglial characteristics associated with specific brain regions. This emerging view suggests that the maturation and function of distinct neuronal circuits may be potentially associated with the molecular identity microglia adopts across the brain. Here, we review and summarize the known role of these cells in the thalamus, hippocampus, cortex, and cerebellum. We focus on in vivo studies to highlight the characteristics of microglia that may be important in the remodeling of these neuronal circuits and in relation to neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Joana R. Guedes
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Institute of Interdisciplinary Research (IIIUC), University of CoimbraCoimbraPortugal
| | - Pedro A. Ferreira
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Department of Life SciencesUniversity of CoimbraCoimbraPortugal
| | - Jéssica M. Costa
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Institute of Interdisciplinary Research (IIIUC), University of CoimbraCoimbraPortugal
| | - Ana L. Cardoso
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Institute of Interdisciplinary Research (IIIUC), University of CoimbraCoimbraPortugal
| | - João Peça
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Department of Life SciencesUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
24
|
Nani JV, Almeida PGC, Noto C, Bressan RA, Brietzke E, Hayashi MAF. Unraveiling the correlation among neurodevelopmental and inflammatory biomarkers in patients with chronic schizophrenia. Nord J Psychiatry 2022; 76:559-564. [PMID: 36189960 DOI: 10.1080/08039488.2021.2023217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Nuclear distribution element like-1 (Ndel1) is a cytosolic oligopeptidase, which was suggested as a potential biomarker of aberrant neurodevelopment and early stage of schizophrenia (SCZ). The involvement of Ndel1 in neurite outgrowth, neuronal migration and neurodevelopment was demonstrated. Moreover, Ndel1 cleaves neuropeptides, including the endogenous antipsychotic peptide neurotensin, and lower Ndel1 activity was reported in SCZ patients compared with healthy controls (HCs). Changes in brain-derived neurotrophic factor (BDNF) and inflammatory cytokines levels were also implicated in SCZ. OBJECTIVE This preliminary study aimed to investigate the interactions between these immune and neurodevelopmental/neurotrophic biomarkers, namely BDNF and the recently identified SCZ biomarker Ndel1. RESULTS We observed lower Ndel1 activity and IL-4 levels, and higher BDNF levels, in plasma of SCZ (N = 23) compared with HCs (N = 29). Interestingly, significant correlation between Ndel1 activity and IL-4 levels was observed in SCZ, while no correlation with any other evaluated interleukins (namely IL-2, IL-8, IL-10 and IL-17A) or BDNF levels was noticed. CONCLUSION Although this hypothesis needs to be further explored for a better understanding of the mechanisms by which these altered pathways are associated to each other in SCZ, we suggest that Ndel1 and the inflammatory marker IL-4 are directly correlated.
Collapse
Affiliation(s)
- João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Sao Paulo, Brazil
| | - Priscila G C Almeida
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Cristiano Noto
- Department of Psychiatry, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Rodrigo A Bressan
- Department of Psychiatry, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Elisa Brietzke
- Department of Psychiatry, Queen's University School of Medicine, Kingston, Canada
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Sao Paulo, Brazil
| |
Collapse
|
25
|
Hanson KL, Grant SE, Funk LH, Schumann CM, Bauman MD. Impact of Maternal Immune Activation on Nonhuman Primate Prefrontal Cortex Development: Insights for Schizophrenia. Biol Psychiatry 2022; 92:460-469. [PMID: 35773097 PMCID: PMC9888668 DOI: 10.1016/j.biopsych.2022.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/30/2022] [Accepted: 04/13/2022] [Indexed: 02/02/2023]
Abstract
Late adolescence is a period of dynamic change in the brain as humans learn to navigate increasingly complex environments. In particular, prefrontal cortical (PFC) regions undergo extensive remodeling as the brain is fine-tuned to orchestrate cognitive control over attention, reasoning, and emotions. Late adolescence also presents a uniquely vulnerable period as neurodevelopmental illnesses, such as schizophrenia, become evident and worsen into young adulthood. Challenges in early development, including prenatal exposure to infection, may set the stage for a cascade of maladaptive events that ultimately result in aberrant PFC connectivity and function before symptoms emerge. A growing body of research suggests that activation of the mother's immune system during pregnancy may act as a disease primer, in combination with other environmental and genetic factors, contributing to an increased risk of neurodevelopmental disorders, including schizophrenia. Animal models provide an invaluable opportunity to examine the course of brain and behavioral changes in offspring exposed to maternal immune activation (MIA). Although the vast majority of MIA research has been carried out in rodents, here we highlight the translational utility of the nonhuman primate (NHP) as a model species more closely related to humans in PFC structure and function. In this review, we consider the protracted period of brain and behavioral maturation in the NHP, describe emerging findings from MIA NHP offspring in the context of rodent preclinical models, and lastly explore the translational relevance of the NHP MIA model to expand understanding of the etiology and developmental course of PFC pathology in schizophrenia.
Collapse
Affiliation(s)
- Kari L Hanson
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California
| | - Simone E Grant
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California
| | - Lucy H Funk
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California
| | - Cynthia M Schumann
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California.
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California; California National Primate Research Center, University of California, Davis, Davis, California.
| |
Collapse
|
26
|
Desale H, Buekens P, Alger J, Cafferata ML, Harville EW, Herrera C, Truyens C, Dumonteil E. Epigenetic signature of exposure to maternal Trypanosoma cruzi infection in cord blood cells from uninfected newborns. Epigenomics 2022; 14:913-927. [PMID: 36039408 PMCID: PMC9475499 DOI: 10.2217/epi-2022-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To assess the epigenetic effects of in utero exposure to maternal Trypanosoma cruzi infection. Methods: We performed an epigenome-wide association study to compare the DNA methylation patterns of umbilical cord blood cells from uninfected babies from chagasic and uninfected mothers. DNA methylation was measured using Infinium EPIC arrays. Results: We identified a differential DNA methylation signature of fetal exposure to maternal T. cruzi infection, in the absence of parasite transmission, with 12 differentially methylated sites in B cells and CD4+ T cells, including eight protein-coding genes. Conclusion: These genes participate in hematopoietic cell differentiation and the immune response and may be involved in immune disorders. They also have been associated with several developmental disorders and syndromes.
Collapse
Affiliation(s)
- Hans Desale
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| | - Pierre Buekens
- Department of Epidemiology, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA 70112, USA
| | - Jackeline Alger
- Instituto de Enfermedades Infecciosas y Parasitologia Antonio Vidal, Tegucigalpa, Honduras.,Ministry of Health, Hospital Escuela, Tegucigalpa, Honduras
| | - Maria Luisa Cafferata
- Unidad de Investigación Clínica y Epidemiológica Montevideo (UNICEM), Hospital de Clínicas, Montevideo, 11600, Uruguay
| | - Emily Wheeler Harville
- Department of Epidemiology, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA 70112, USA
| | - Claudia Herrera
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, & ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles, Brussels, Belgium
| | - Eric Dumonteil
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| |
Collapse
|
27
|
Ryan AM, Bauman MD. Primate Models as a Translational Tool for Understanding Prenatal Origins of Neurodevelopmental Disorders Associated With Maternal Infection. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:510-523. [PMID: 35276404 PMCID: PMC8902899 DOI: 10.1016/j.bpsc.2022.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/13/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023]
Abstract
Pregnant women represent a uniquely vulnerable population during an infectious disease outbreak, such as the COVID-19 pandemic. Although we are at the early stages of understanding the specific impact of SARS-CoV-2 exposure during pregnancy, mounting epidemiological evidence strongly supports a link between exposure to a variety of maternal infections and an increased risk for offspring neurodevelopmental disorders. Inflammatory biomarkers identified from archived or prospectively collected maternal biospecimens suggest that the maternal immune response is the critical link between infection during pregnancy and altered offspring neurodevelopment. This maternal immune activation (MIA) hypothesis has been tested in animal models by artificially activating the immune system during pregnancy and evaluating the neurodevelopmental consequences in MIA-exposed offspring. Although the vast majority of MIA model research is carried out in rodents, the nonhuman primate model has emerged in recent years as an important translational tool. In this review, we briefly summarize human epidemiological studies that have prompted the development of translationally relevant MIA models. We then highlight notable similarities between humans and nonhuman primates, including placental structure, pregnancy physiology, gestational timelines, and offspring neurodevelopmental stages, that provide an opportunity to explore the MIA hypothesis in species more closely related to humans. Finally, we provide a comprehensive review of neurodevelopmental alterations reported in current nonhuman primate models of maternal infection and discuss future directions for this promising area of research.
Collapse
Affiliation(s)
- Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis, Davis, California; California National Primate Research Center, University of California Davis, Davis, California
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis, Davis, California; California National Primate Research Center, University of California Davis, Davis, California.
| |
Collapse
|
28
|
Bryan EE, Chen X, Smith BS, Dilger RN, Dilger AC. Maternal Immune Activation and Dietary Soy Isoflavone Supplementation Influence Pig Immune Function but not Muscle Fiber Formation. J Anim Sci 2022; 100:6568979. [PMID: 35426431 PMCID: PMC9155173 DOI: 10.1093/jas/skac134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
The goals of this study were to determine the impact of maternal PRRSV infection on offspring muscle and immune development and the potential of dietary soy isoflavones to mitigate those effects. Thirteen first-parity gilts (“gilts”) were randomly allotted into one of three treatments: not infected and fed a diet devoid of isoflavones (CON), infected with porcine reproductive and respiratory syndrome virus (PRRSV) and fed the control diet (POS) or that supplemented with 1,500 mg/kg soy-derived isoflavones (ISF). Gilts were inoculated with PRRSV intranasally on gestational day (GD) 70. After farrowing (GD 114 ± 2), 1-2 offspring (“pigs”) closest to the average litter weight were selected either at birth (3 ± 2 d of age) or weaning (21 ±2 d of age) to determine body, muscle, and organ weights as well as muscle cell number and size. Four weaned pigs of average body weight within each litter were selected for postnatal immune challenge. At PND 52, pigs were injected with 5 µg/kg BW lipopolysaccharide (LPS) intraperitoneally. Serum was collected at 0, 4, and 8 h following LPS administration to analyze tumor necrosis factor alpha (TNF-α). At PND 59, pigs were administered a novel vaccine to elicit an adaptive immune response. At PND 59, 66, and 73, peripheral blood mononuclear cells were isolated and T-cell populations determined by flow cytometry. Both POS and ISF pigs exhibited persistent PRRSV infections throughout the study (PND 1-73). At PND 3, whole body, muscle, and organ weights were not different (P > 0.22) between groups, with the exception of relative liver weight, which was increased (P < 0.05) in POS compared with CON pigs. At PND 21, ISF pigs had reduced (P ≤ 0.05) whole body and muscle weights, but greater (P < 0.05) kidney weight compared with CON, and greater (P < 0.05) relative liver weight compared with CON and POS. Muscle fiber number and size were not different (P > 0.39) between groups at birth or weaning. After LPS administration, TNF-α was greatest in ISF pigs (P < 0.05) at both 0 and 8 h post-challenge. At the peak time-point of 4 h post-challenge, ISF pigs had the greatest concentration of TNF-α and CON pigs had the lowest, with POS pigs being intermediate (P = 0.01). After vaccination, ISF offspring had shifts in T-cell populations indicating an impaired immune response. These data indicate that maternal PRRSV infection may impact offspring organ growth and immune function, particularly when the dam is supplemented with isoflavones.
Collapse
Affiliation(s)
- E E Bryan
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, USA
| | - X Chen
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, USA
| | - B S Smith
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, USA
| | - R N Dilger
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, USA
| | - A C Dilger
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, USA
| |
Collapse
|
29
|
Massrali A, Adhya D, Srivastava DP, Baron-Cohen S, Kotter MR. Virus-Induced Maternal Immune Activation as an Environmental Factor in the Etiology of Autism and Schizophrenia. Front Neurosci 2022; 16:834058. [PMID: 35495047 PMCID: PMC9039720 DOI: 10.3389/fnins.2022.834058] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/01/2022] [Indexed: 12/22/2022] Open
Abstract
Maternal immune activation (MIA) is mediated by activation of inflammatory pathways resulting in increased levels of cytokines and chemokines that cross the placental and blood-brain barriers altering fetal neural development. Maternal viral infection is one of the most well-known causes for immune activation in pregnant women. MIA and immune abnormalities are key players in the etiology of developmental conditions such as autism, schizophrenia, ADHD, and depression. Experimental evidence implicating MIA in with different effects in the offspring is complex. For decades, scientists have relied on either MIA models or human epidemiological data or a combination of both. MIA models are generated using infection/pathogenic agents to induce an immunological reaction in rodents and monitor the effects. Human epidemiological studies investigate a link between maternal infection and/or high levels of cytokines in pregnant mothers and the likelihood of developing conditions. In this review, we discuss the importance of understanding the relationship between virus-mediated MIA and neurodevelopmental conditions, focusing on autism and schizophrenia. We further discuss the different methods of studying MIA and their limitations and focus on the different factors contributing to MIA heterogeneity.
Collapse
Affiliation(s)
- Aïcha Massrali
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Dwaipayan Adhya
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, King’s College London, London, United Kingdom
| | - Simon Baron-Cohen
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Mark R. Kotter
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
30
|
Abstract
Many patients under treatment for mood disorders, in particular patients with bipolar mood disorders, experience episodes of mood switching from one state to another. Various hypotheses have been proposed to explain the mechanism of mood switching, spontaneously or induced by drug treatment. Animal models have also been used to test the role of psychotropic drugs in the switching of mood states. We examine the possible relationship between the pharmacology of psychotropic drugs and their reported incidents of induced mood switching, with reference to the various hypotheses of mechanisms of mood switching.
Collapse
|
31
|
Tian J, Gao X, Yang L. Repetitive Restricted Behaviors in Autism Spectrum Disorder: From Mechanism to Development of Therapeutics. Front Neurosci 2022; 16:780407. [PMID: 35310097 PMCID: PMC8924045 DOI: 10.3389/fnins.2022.780407] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 02/09/2022] [Indexed: 01/28/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by deficits in social communication, social interaction, and repetitive restricted behaviors (RRBs). It is usually detected in early childhood. RRBs are behavioral patterns characterized by repetition, inflexibility, invariance, inappropriateness, and frequent lack of obvious function or specific purpose. To date, the classification of RRBs is contentious. Understanding the potential mechanisms of RRBs in children with ASD, such as neural connectivity disorders and abnormal immune functions, will contribute to finding new therapeutic targets. Although behavioral intervention remains the most effective and safe strategy for RRBs treatment, some promising drugs and new treatment options (e.g., supplementary and cell therapy) have shown positive effects on RRBs in recent studies. In this review, we summarize the latest advances of RRBs from mechanistic to therapeutic approaches and propose potential future directions in research on RRBs.
Collapse
Affiliation(s)
| | | | - Li Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| |
Collapse
|
32
|
Kwon HK, Choi GB, Huh JR. Maternal inflammation and its ramifications on fetal neurodevelopment. Trends Immunol 2022; 43:230-244. [PMID: 35131181 PMCID: PMC9005201 DOI: 10.1016/j.it.2022.01.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
Exposure to heightened inflammation in pregnancy caused by infections or other inflammatory insults has been associated with the onset of neurodevelopmental and psychiatric disorders in children. Rodent models have provided unique insights into how this maternal immune activation (MIA) disrupts brain development. Here, we discuss the key immune factors involved, highlight recent advances in determining the molecular and cellular pathways of MIA, and review how the maternal immune system affects fetal development. We also examine the roles of microbiomes in shaping maternal immune function and the development of autism-like phenotypes. A comprehensive understanding of the gut bacteria-immune-neuro interaction in MIA is essential for developing diagnostic and therapeutic measures for high-risk pregnant women and identifying targets for treating inflammation-induced neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ho-Keun Kwon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea; Pohang University of Science and Technology, Pohang, Korea.
| | - Gloria B. Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jun R. Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA.,Correspondence: Ho-Keun Kwon () and Jun R. Huh ()
| |
Collapse
|
33
|
Increased Monocyte Production of IL-6 after Toll-like Receptor Activation in Children with Autism Spectrum Disorder (ASD) Is Associated with Repetitive and Restricted Behaviors. Brain Sci 2022; 12:brainsci12020220. [PMID: 35203983 PMCID: PMC8870658 DOI: 10.3390/brainsci12020220] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 01/27/2023] Open
Abstract
The prevalence of autism spectrum disorder (ASD) has starkly increased, instigating research into risk factors for ASD. This research has identified immune risk factors for ASD, along with evidence of immune dysfunction and excess inflammation frequently experienced by autistic individuals. Increased innate inflammatory cytokines, including interleukin (IL)-6, are seen repeatedly in ASD; however, the origin of excess IL-6 in ASD has not been identified. Here we explore specific responses of circulating monocytes from autistic children. We isolated CD14+ monocytes from whole blood and stimulated them for 24 h under three conditions: media alone, lipoteichoic acid to activate TLR2, and lipopolysaccharide to activate TLR4. We then measured secreted cytokine concentrations in cellular supernatant using a human multiplex bead immunoassay. We found that after TLR4 activation, CD14+ monocytes from autistic children produce increased IL-6 compared to monocytes from children with typical development. IL-6 concentration also correlated with worsening restrictive and repetitive behaviors. These findings suggest dysfunctional activation of myeloid cells, and may indicate that other cells of this lineage, including macrophages, and microglia in the brain, might have a similar dysfunction. Further research on myeloid cells in ASD is warranted.
Collapse
|
34
|
Moreno R, Ashwood P. The Issue of Monocyte Activation in ASD: Troubles with Translation. JOURNAL OF CELLULAR IMMUNOLOGY 2022; 4:167-170. [PMID: 36688057 PMCID: PMC9853954 DOI: 10.33696/immunology.4.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD) prevalence has increased year on year for the past two decades and currently affects 1 in 44 individuals in the US. An increasing number of studies have pointed to increased immune activation as both an etiological agent and also involved in the ongoing pathological process of ASD. Both adaptive and innate immune responses have been implicated. Evidence of innate dysregulation has so far included increased production of innate inflammatory cytokines, increased cell numbers, and altered activation in monocytes in the blood and microglia in the brain. Suggesting an orchestrated innate immune response may be involved in ASD. Hughes et al. (2022) recently assessed transcriptome differences that could underlie altered activation of monocytes using next-generation bulk-RNA sequencing on isolated CD14+ monocytes at baseline and after activation with different Toll-like receptor agonists. Circulating CD14+ monocyte from children with autistic disorder (AD) and children diagnosed with perverse developmental disorder not otherwise specified (PDD-NOS) were found to differ in a number of activation pathways after gene enrichment analysis compared to typically developing children. There was an overall upregulation in translational machinery in both neurodevelopmental disorder groups, whereas typically developing children were downregulated, indicating an issue with monocyte activation. Several identified differentially expressed genes in monocytes were also identified as ASD at-risk genes, according to the Simons Foundation Autism Research Initiative (SFARI), and genes involved in inflammatory bowel diseases. This work implicates altered monocyte activation with a lack of regulation as a potential mechanistic issue in ASD. Future work is warranted to evaluate how monocyte regulatory mechanisms differ in ASD individuals.
Collapse
Affiliation(s)
- R.J. Moreno
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA,M.I.N.D. Institute, University of California at Davis, CA, USA
| | - P. Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA,M.I.N.D. Institute, University of California at Davis, CA, USA,Correspondence should be addressed to Paul Ashwood,
| |
Collapse
|
35
|
Vlasova RM, Iosif AM, Ryan AM, Funk LH, Murai T, Chen S, Lesh TA, Rowland DJ, Bennett J, Hogrefe CE, Maddock RJ, Gandal MJ, Geschwind DH, Schumann CM, Van de Water J, McAllister AK, Carter CS, Styner MA, Amaral DG, Bauman MD. Maternal Immune Activation during Pregnancy Alters Postnatal Brain Growth and Cognitive Development in Nonhuman Primate Offspring. J Neurosci 2021; 41:9971-9987. [PMID: 34607967 PMCID: PMC8638691 DOI: 10.1523/jneurosci.0378-21.2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/28/2021] [Accepted: 09/06/2021] [Indexed: 11/21/2022] Open
Abstract
Human epidemiological studies implicate exposure to infection during gestation in the etiology of neurodevelopmental disorders. Animal models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant offspring brain and behavior development. Here we evaluate neurodevelopment of male rhesus monkeys (Macaca mulatta) born to MIA-treated dams (n = 14) injected with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the same gestational time points (n = 10) or were untreated (n = 4). MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature, and inflammatory cytokines. Although offspring born to control or MIA-treated dams did not differ on measures of physical growth and early developmental milestones, the MIA-treated animals exhibited subtle changes in cognitive development and deviated from species-typical brain growth trajectories. Longitudinal MRI revealed significant gray matter volume reductions in the prefrontal and frontal cortices of MIA-treated offspring at 6 months that persisted through the final time point at 45 months along with smaller frontal white matter volumes in MIA-treated animals at 36 and 45 months. These findings provide the first evidence of early postnatal changes in brain development in MIA-exposed nonhuman primates and establish a translationally relevant model system to explore the neurodevelopmental trajectory of risk associated with prenatal immune challenge from birth through late adolescence.SIGNIFICANCE STATEMENT Women exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder. Preclinical maternal immune activation (MIA) models have demonstrated that the effects of maternal infection on fetal brain development are mediated by maternal immune response. Since the majority of MIA models are conducted in rodents, the nonhuman primate provides a unique system to evaluate the MIA hypothesis in a species closely related to humans. Here we report the first longitudinal study conducted in a nonhuman primate MIA model. MIA-exposed offspring demonstrate subtle changes in cognitive development paired with marked reductions in frontal gray and white matter, further supporting the association between prenatal immune challenge and alterations in offspring neurodevelopment.
Collapse
Affiliation(s)
- Roza M Vlasova
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, 27514
| | - Ana-Maria Iosif
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- California National Primate Research Center, University of California, Davis, California, 95616
| | - Lucy H Funk
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Takeshi Murai
- California National Primate Research Center, University of California, Davis, California, 95616
| | - Shuai Chen
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Tyler A Lesh
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Douglas J Rowland
- Center for Genomic and Molecular Imaging, University of California, Davis, California, 95616
| | - Jeffrey Bennett
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Casey E Hogrefe
- California National Primate Research Center, University of California, Davis, California, 95616
| | - Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Michael J Gandal
- Neurogenetics Program, Department of Neurology, University of California, Los Angeles, California, 90095
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, University of California, Los Angeles, California, 90095
| | - Cynthia M Schumann
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Judy Van de Water
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- Rheumatology/Allergy and Clinical Immunology, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - A Kimberley McAllister
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- Center for Neuroscience, University of California, Davis, California, 95618
| | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Martin A Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, 27514
- Department of Computer Science, University of North Carolina, Chapel Hill, North Carolina, 27599
| | - David G Amaral
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- California National Primate Research Center, University of California, Davis, California, 95616
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- California National Primate Research Center, University of California, Davis, California, 95616
| |
Collapse
|
36
|
O'Connor TG, Ciesla AA. Maternal Immune Activation Hypotheses for Human Neurodevelopment: Some Outstanding Questions. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:471-479. [PMID: 34688920 PMCID: PMC9021321 DOI: 10.1016/j.bpsc.2021.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
The Maternal Immune Activation (MIA) hypothesis is a leading model for understanding prenatal influences on individual differences in, and clinical syndromes of, neurodevelopment. Experimental animal and human research has proliferated in recent years, and there is now a sizable research base. Several meta-analyses demonstrate general support for an association between prenatal immune activation and neurodevelopment in human research. However, questions remain about the nature of the immune activation, the network of underlying mechanisms involved, and the breadth of impact across behavioral phenotypes. Complementing recent reviews of results, the current review places particular emphasis on how advances in understanding mechanisms may be improved with greater attention to addressing the methodological variation and limitations of existing studies, and identifies areas for further clinical research.
Collapse
Affiliation(s)
- Thomas G O'Connor
- Department of Psychiatry, University of Rochester; Department of Psycholog, University of Rochestery; Department of Neuroscience, University of Rochester; Department of Obstetrics and Gynecology, University of Rochester; Wynne Center for Family Research, University of Rochester.
| | | |
Collapse
|
37
|
Hameete BC, Fernández-Calleja JM, de Groot MW, Oppewal TR, Tiemessen MM, Hogenkamp A, de Vries RB, Groenink L. The poly(I:C)-induced maternal immune activation model; a systematic review and meta-analysis of cytokine levels in the offspring. Brain Behav Immun Health 2021; 11:100192. [PMID: 34589729 PMCID: PMC8474626 DOI: 10.1016/j.bbih.2020.100192] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022] Open
Abstract
The maternal polyinosinic:polycytidylic acid (poly(I:C)) animal model is frequently used to study how maternal immune activation may impact neuro development in the offspring. Here, we present the first systematic review and meta-analysis on the effects of maternal poly(I:C) injection on immune mediators in the offspring and provide an openly accessible systematic map of the data including methodological characteristics. Pubmed and EMBASE were searched for relevant publications, yielding 45 unique papers that met inclusion criteria. We extracted data on immune outcomes and methodological characteristics, and assessed the risk of bias. The descriptive summary showed that most studies reported an absence of effect, with an equal number of studies reporting an increase or decrease in the immune mediator being studied. Meta-analysis showed increased IL-6 concentrations in the offspring of poly(I:C) exposed mothers. This effect appeared larger prenatally than post-weaning. Furthermore, poly(I:C) administration during mid-gestation was associated with higher IL-6 concentrations in the offspring. Maternal poly(I:C) induced changes in IL-1β, Il-10 and TNF-α concentrations were small and could not be associated with age of offspring, gestational period or sampling location. Finally, quality of reporting of potential measures to minimize bias was low, which stresses the importance of adherence to publication guidelines. Since neurodevelopmental disorders in humans tend to be associated with lifelong changes in cytokine concentrations, the absence of these effects as identified in this systematic review may suggest that combining the model with other etiological factors in future studies may provide further insight in the mechanisms through which maternal immune activation affects neurodevelopment. Long-term effects of maternal poly(I:C) on immune mediators in the offspring appear limited. Prenatal measurements and mid gestation poly(I:C) injection are associated with increases in IL-6 concentrations. Variety in methodological conduct hampers identification of key elements that affect cytokine concentrations. The quality of reporting of potential measures to minimize bias is poor.
Collapse
Affiliation(s)
- Bart C. Hameete
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - José M.S. Fernández-Calleja
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Martje W.G.D.M. de Groot
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Titia Rixt Oppewal
- University College Utrecht (UCU), Campusplein 1, Utrecht, 3584 ED, the Netherlands
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, the Netherlands
| | - Machteld M. Tiemessen
- Research & Innovation, GCoE Immunology, Danone Nutricia Research, Uppsalalaan 12, Utrecht, 3584 CT, the Netherlands
| | - Astrid Hogenkamp
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Rob B.M. de Vries
- SYstematic Review Center for Laboratory (Animal) Experimentation, Department for Health Evidence, Radboud University Medical Center, Geert Grooteplein zuid 10, Nijmegen, 6525 GA, the Netherlands
| | - Lucianne Groenink
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
- Corresponding author.
| |
Collapse
|
38
|
Maternal effects in mammals: Broadening our understanding of offspring programming. Front Neuroendocrinol 2021; 62:100924. [PMID: 33992652 DOI: 10.1016/j.yfrne.2021.100924] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/18/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
The perinatal period is a sensitive time in mammalian development that can have long-lasting consequences on offspring phenotype via maternal effects. Maternal effects have been most intensively studied with respect to two major conditions: maternal diet and maternal stress. In this review, we shift the focus by discussing five major additional maternal cues and their influence on offspring phenotype: maternal androgen levels, photoperiod (melatonin), microbiome, immune regulation, and milk composition. We present the key findings for each of these topics in mammals, their mechanisms of action, and how they interact with each other and with the maternal influences of diet and stress. We explore their impacts in the contexts of both predictive adaptive responses and the developmental origins of disease, identify knowledge gaps and research opportunities in the field, and place a particular emphasis on the application and consideration of these effects in non-model species and natural ecological systems.
Collapse
|
39
|
Carta A, Zarbo IR, Scoppola C, Pisuttu G, Conti M, Melis MC, Martino FD, Serra A, Biancu MA, Guerini FR, Bazzardi R, Sotgiu S. Maternal multiple sclerosis is not a risk factor for neurodevelopmental disorders in offspring. Mult Scler J Exp Transl Clin 2021; 7:20552173211017301. [PMID: 34104473 PMCID: PMC8165841 DOI: 10.1177/20552173211017301] [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: 03/01/2021] [Accepted: 04/25/2021] [Indexed: 11/24/2022] Open
Abstract
Background Childhood neurodevelopmental disorders (NDDs), including specific learning disorders (SLD), attention deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD), are pathogenically linked to familial autoimmunity and maternal immune-mediated diseases during pregnancy. Objective We studied maternal MS as a potential risk factor for NDDs occurrence in offspring. Methods MS and control mothers were subjected to questionnaires to ascertain NDD diagnosis in their progeny and the occurrence of both autoimmune and neurodevelopment disorders in their families. Suspected NDD cases were evaluated to confirm or rule out the diagnosis. Results Of the 322 MS women, 206 (64%) have 361 children; of these, 27 (7.5%) were diagnosed with NDD (11% ADHD; 22% ASD; 67% SLD). NDD-risk in offspring was associated to family history of autoimmunity and to NDDs both in MS and non-MS mother families (r = 0.75; p = 0.005) whereas it was not associated to maternal MS. Conclusions For the first time, we demonstrate that maternal MS does not predispose children to higher risk for NDD. On a mechanistic view, we suggest that the intrinsic organ-specific nature of MS does not impair the mother–child cross-talk in decidua nor does it influence fetal neurodevelopment.
Collapse
Affiliation(s)
- Alessandra Carta
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Ignazio R Zarbo
- Unit of Clinical Neurology, Department of Medical, Surgical and Experimental Sciences, Multiple Sclerosis Centre, University of Sassari, Sassari, Italy
| | - Chiara Scoppola
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giulia Pisuttu
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Marta Conti
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Maria C Melis
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Federica De Martino
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Antonella Serra
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Maria A Biancu
- Unit of Clinical Neurology, Department of Medical, Surgical and Experimental Sciences, Multiple Sclerosis Centre, University of Sassari, Sassari, Italy
| | | | - Riccardo Bazzardi
- Struttura Complessa Controllo Microbiologico e Ispezione degli Alimenti, Istituto Zooprofilattico Sperimentale della Sardegna "G. Pegreffi", Sassari, Italy
| | - Stefano Sotgiu
- Unit of Child Neuropsychiatry, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
40
|
Mycobacterium tuberculosis-Induced Maternal Immune Activation Promotes Autism-Like Phenotype in Infected Mice Offspring. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18094513. [PMID: 33922864 PMCID: PMC8122996 DOI: 10.3390/ijerph18094513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/30/2021] [Accepted: 04/03/2021] [Indexed: 12/25/2022]
Abstract
The maternal system’s exposure to pathogens during pregnancy influences fetal brain development causing a persistent inflammation characterized by elevated pro-inflammatory cytokine levels in offspring. Mycobacterium tuberculosis (Mtb) is a global pathogen that causes tuberculosis, a pandemic responsible for health and economic burdens. Although it is known that maternal infections increase the risk of autism spectrum disorder (ASD), it is not known whether Mtb infection is sufficient to induce ASD associated behaviors, immune dysregulation and altered expression of synaptic regulatory genes. The current study infected pregnant Balb/c mice with Mtb H37Rv and valproic acid (VPA) individually and in combination. Plasma cytokine profiles were measured in offspring using the Bio-plex Th17 pro mouse cytokine panel. Mtb infection increased plasma interleukin (IL)-6 and IL-17A, while tumor necrosis factor alpha (TNF-α), interferon (IFN)-γ and IL-1β were reduced when compared with saline. Mtb-induced maternal immune activation (MIA) offspring displayed increased grooming behavior. The study also revealed dysregulation in gene expression of synaptic molecules in the cerebellum. MIA rescued the VPA-induced effects on self-grooming and social interaction behaviors. Our finding therefore highlights a potential role of Mtb as a MIA agent that can potentially contribute to ASD.
Collapse
|
41
|
Shimizu Y, Tsukada T, Sakata-Haga H, Sakai D, Shoji H, Saikawa Y, Hatta T. Exposure to Maternal Immune Activation Causes Congenital Unfolded Protein Response Defects and Increases the Susceptibility to Postnatal Inflammatory Stimulation in Offspring. J Inflamm Res 2021; 14:355-365. [PMID: 33603435 PMCID: PMC7886242 DOI: 10.2147/jir.s294238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Background A number of childhood diseases have been identified, such as severe infection or autoinflammatory disease, in which immune overreaction against inflammation is a possible underlying mechanism. Previous reports have demonstrated that fetal cells exposed to maternal immune activation (MIA) induced by polyriboinosinic-polyribocytidylic acid [poly(I:C)] exhibited hypersensitivity to inflammation in vitro. However, the details of this mechanism remain unclear. Therefore, this study aimed to reveal the reaction to inflammation in offspring exposed to MIA in the prenatal period, as well as its molecular mechanism, using a viral infection mouse model. Materials and Methods Pregnant mice at 12.5, 14.5, and 16.5 days post coitum were injected intraperitoneally with poly(I:C) 20 mg/kg body weight (BW) or saline. Offspring aged 3-4 weeks received the second injection of 20 mg/kg BW or 4 mg/kg BW poly(I:C) or saline. Serum and tissues were collected at 2, 24, 48, and 72 h after the postnatal injection. The cytokine profile, histopathology of organs, and unfolded protein response (UPR) in offspring were examined. Results The serum levels of interleukin (IL)-6, IL-17, and interferon-γ were significantly higher in the MIA group, and acute liver necrosis was detected. Moreover, failure in UPR was observed in the MIA group compared with that in the control group. Conclusion Overall, MIA exposure in utero caused failure in UPR as well as immune overreaction to the second attack of inflammation in offspring. Our results suggested that prenatal exposure to MIA might contribute to the congenital inflammatory constitution after birth.
Collapse
Affiliation(s)
- Yo Shimizu
- Department of Pediatrics, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Tsuyoshi Tsukada
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Hiromi Sakata-Haga
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Daisuke Sakai
- Department of Biology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Hiroki Shoji
- Department of Biology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Yutaka Saikawa
- Department of Pediatrics, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| |
Collapse
|
42
|
Mueller FS, Scarborough J, Schalbetter SM, Richetto J, Kim E, Couch A, Yee Y, Lerch JP, Vernon AC, Weber-Stadlbauer U, Meyer U. Behavioral, neuroanatomical, and molecular correlates of resilience and susceptibility to maternal immune activation. Mol Psychiatry 2021; 26:396-410. [PMID: 33230204 PMCID: PMC7850974 DOI: 10.1038/s41380-020-00952-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/24/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
Infectious or noninfectious maternal immune activation (MIA) is an environmental risk factor for psychiatric and neurological disorders with neurodevelopmental etiologies. Whilst there is increasing evidence for significant health consequences, the effects of MIA on the offspring appear to be variable. Here, we aimed to identify and characterize subgroups of isogenic mouse offspring exposed to identical MIA, which was induced in C57BL6/N mice by administration of the viral mimetic, poly(I:C), on gestation day 12. Cluster analysis of behavioral data obtained from a first cohort containing >150 MIA and control offspring revealed that MIA offspring could be stratified into distinct subgroups that were characterized by the presence or absence of multiple behavioral dysfunctions. The two subgroups also differed in terms of their transcriptional profiles in cortical and subcortical brain regions and brain networks of structural covariance, as measured by ex vivo structural magnetic resonance imaging (MRI). In a second, independent cohort containing 50 MIA and control offspring, we identified a subgroup of MIA offspring that displayed elevated peripheral production of innate inflammatory cytokines, including IL-1β, IL-6, and TNF-α, in adulthood. This subgroup also showed significant impairments in social approach behavior and sensorimotor gating, whereas MIA offspring with a low inflammatory cytokine status did not. Taken together, our results highlight the existence of subgroups of MIA-exposed offspring that show dissociable behavioral, transcriptional, brain network, and immunological profiles even under conditions of genetic homogeneity. These data have relevance for advancing our understanding of the variable neurodevelopmental effects induced by MIA and for biomarker-guided approaches in preclinical psychiatric research.
Collapse
Affiliation(s)
- Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Joseph Scarborough
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Sina M Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Eugene Kim
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Amalie Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Yohan Yee
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
43
|
Ketharanathan T, Pereira A, Reets U, Walker D, Sundram S. Brain changes in NF-κB1 and epidermal growth factor system markers at peri-pubescence in the spiny mouse following maternal immune activation. Psychiatry Res 2021; 295:113564. [PMID: 33229121 DOI: 10.1016/j.psychres.2020.113564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/05/2020] [Indexed: 10/23/2022]
Abstract
Environmental risk factors that operate at foetal or neonatal levels increase the vulnerability to schizophrenia, plausibly via stress-immune activation that perturbs the epidermal growth factor (EGF) system, a system critical for neurodevelopment. We investigated potential associations between environmental insults and immune and EGF system changes through a maternal immune activation (MIA) model, using the precocial spiny mice (Acomys cahirinus). After mid-gestation MIA prepubescent offspring showed elevated NF-κB1 protein in nucleus accumbens, decreased EGFR in caudate putamen and a trend for increased PI3K-110δ in ventral hippocampus. Thus, prenatal stress may cause a heightened NF-κB1-mediated immune attenuation of EGF system signalling.
Collapse
Affiliation(s)
- Tharini Ketharanathan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia.
| | - Avril Pereira
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia; Department of Psychiatry, University of Melbourne, Parkville, VIC 3052, Australia
| | - Udani Reets
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| | - David Walker
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| | - Suresh Sundram
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
44
|
Santana-Coelho D, Layne-Colon D, Valdespino R, Ross CC, Tardif SD, O'Connor JC. Advancing Autism Research From Mice to Marmosets: Behavioral Development of Offspring Following Prenatal Maternal Immune Activation. Front Psychiatry 2021; 12:705554. [PMID: 34421684 PMCID: PMC8377364 DOI: 10.3389/fpsyt.2021.705554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/07/2021] [Indexed: 12/31/2022] Open
Abstract
Understanding the mechanism(s) by which maternal immune activation (MIA) during gestation may disrupt neurodevelopment and increase the susceptibility for disorders such as autism spectrum disorder (ASD) or schizophrenia is a critical step in the development of better treatments and preventive measures. A large body of literature has investigated the pathophysiology of MIA in rodents. However, a translatability gap plagues pre-clinical research of complex behavioral/developmental diseases and those diseases requiring clinical diagnosis, such as ASD. While ideal for their genetic flexibility, vast reagent toolkit, and practicality, rodent models often lack important elements of ethological validity. Hence, our study aimed to develop and characterize the prenatal MIA model in marmosets. Here, we adapted the well-characterized murine maternal immune activation model. Pregnant dams were administered 5 mg/kg poly-L-lysine stabilized polyinosinic-polycytidylic acid (Poly ICLC) subcutaneously three times during gestation (gestational day 63, 65, and 67). Dams were allowed to deliver naturally with no further experimental treatments. After parturition, offspring were screened for general health and vigor, and individual assessment of communication development and social behavior was measured during neonatal or adolescent periods. Similar to rodent models, offspring subjected to MIA exhibited a disruption in patterns of communication during early development. Assessment of social behavior in a marmoset-modified 3-chamber test at 3 and 9 months of age revealed alterations in social behavior that, in some instances, was sex-dependent. Together, our data indicate that marmosets are an excellent non-human primate model for investigating the neurodevelopmental and behavioral consequences of exposure to prenatal challenges, like MIA. Additional studies are necessary to more completely characterize the effect of prenatal inflammation on marmoset development and explore therapeutic intervention strategies that may be applicable in a clinical setting.
Collapse
Affiliation(s)
- Danielle Santana-Coelho
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Donna Layne-Colon
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Roslyn Valdespino
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Corinna C Ross
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Suzette D Tardif
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jason C O'Connor
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Audie L. Murphy Veterans Affairs, South Texas Veterans Health System, San Antonio, TX, United States
| |
Collapse
|
45
|
Volk HE, Park B, Hollingue C, Jones KL, Ashwood P, Windham GC, Lurman F, Alexeeff SE, Kharrazi M, Pearl M, Van de Water J, Croen LA. Maternal immune response and air pollution exposure during pregnancy: insights from the Early Markers for Autism (EMA) study. J Neurodev Disord 2020; 12:42. [PMID: 33327930 PMCID: PMC7745402 DOI: 10.1186/s11689-020-09343-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 11/13/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Perinatal exposure to air pollution and immune system dysregulation are two factors consistently associated with autism spectrum disorders (ASD) and other neurodevelopmental outcomes. However, little is known about how air pollution may influence maternal immune function during pregnancy. OBJECTIVES To assess the relationship between mid-gestational circulating levels of maternal cytokines/chemokines and previous month air pollution exposure across neurodevelopmental groups, and to assess whether cytokines/chemokines mediate the relationship between air pollution exposures and risk of ASD and/or intellectual disability (ID) in the Early Markers for Autism (EMA) study. METHODS EMA is a population-based, nested case-control study which linked archived maternal serum samples collected during weeks 15-19 of gestation for routine prenatal screening, birth records, and Department of Developmental Services (DDS) records. Children receiving DDS services for ASD without intellectual disability (ASD without ID; n = 199), ASD with ID (ASD with ID; n = 180), ID without ASD (ID; n = 164), and children from the general population (GP; n = 414) with no DDS services were included in this analysis. Serum samples were quantified for 22 cytokines/chemokines using Luminex multiplex analysis technology. Air pollution exposure for the month prior to maternal serum collection was assigned based on the Environmental Protection Agency's Air Quality System data using the maternal residential address reported during the prenatal screening visit. RESULTS Previous month air pollution exposure and mid-gestational maternal cytokine and chemokine levels were significantly correlated, though weak in magnitude (ranging from - 0.16 to 0.13). Ten pairs of mid-pregnancy immune markers and previous month air pollutants were significantly associated within one of the child neurodevelopmental groups, adjusted for covariates (p < 0.001). Mid-pregnancy air pollution was not associated with any neurodevelopmental outcome. IL-6 remained associated with ASD with ID even after adjusting for air pollution exposure. CONCLUSION This study suggests that maternal immune activation is associated with risk for neurodevelopmental disorders. Furthermore, that prenatal air pollution exposure is associated with small, but perhaps biologically relevant, effects on maternal immune system function during pregnancy. Additional studies are needed to better evaluate how prenatal exposure to air pollution affects the trajectory of maternal immune activation during pregnancy, if windows of heightened susceptibility can be identified, and how these factors influence neurodevelopment of the offspring.
Collapse
Affiliation(s)
- Heather E Volk
- Department of Mental Health, Wendy Klag Center for Autism and Developmental Disabilities, Bloomberg School of Public Health, Johns Hopkins University, Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center, 624 N. Broadway, HH833, Baltimore, MD, 21205, USA.
| | - Bo Park
- Department of Public Health, California State University, Fullerton, CA, USA
| | - Calliope Hollingue
- Department of Mental Health, Wendy Klag Center for Autism and Developmental Disabilities, Bloomberg School of Public Health, Johns Hopkins University, Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center, 624 N. Broadway, HH833, Baltimore, MD, 21205, USA
| | - Karen L Jones
- UC Davis MIND Institute, University of California Davis, Davis, CA, USA
| | - Paul Ashwood
- UC Davis MIND Institute, University of California Davis, Davis, CA, USA
| | - Gayle C Windham
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, CA, USA
| | | | - Stacey E Alexeeff
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA, USA
| | - Martin Kharrazi
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, CA, USA
| | - Michelle Pearl
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, CA, USA
| | - Judy Van de Water
- UC Davis MIND Institute, University of California Davis, Davis, CA, USA
| | - Lisa A Croen
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA, USA
| |
Collapse
|
46
|
Sotgiu S, Manca S, Gagliano A, Minutolo A, Melis MC, Pisuttu G, Scoppola C, Bolognesi E, Clerici M, Guerini FR, Carta A. Immune regulation of neurodevelopment at the mother-foetus interface: the case of autism. Clin Transl Immunology 2020; 9:e1211. [PMID: 33209302 PMCID: PMC7662086 DOI: 10.1002/cti2.1211] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder defined by deficits in social communication and stereotypical behaviours. ASD’s aetiology remains mostly unclear, because of a complex interaction between genetic and environmental factors. Recently, a strong consensus has developed around ASD’s immune‐mediated pathophysiology, which is the subject of this review. For many years, neuroimmunological studies tried to understand ASD as a prototypical antibody‐ or cell‐mediated disease. Other findings indicated the importance of autoimmune mechanisms such as familial and individual autoimmunity, adaptive immune abnormalities and the influence of infections during gestation. However, recent studies have challenged the idea that autism may be a classical autoimmune disease. Modern neurodevelopmental immunology shows the double‐edged nature of many immune effectors, which can be either beneficial or detrimental depending on tissue homeostasis, stressors, neurodevelopmental stage, inherited and de novo gene mutations and other variables. Nowadays, mother–child interactions in the prenatal environment appear to be crucial for the occurrence of ASD. Studies of animal maternal–foetal immune interaction are being fruitfully carried out using different combinations of type and timing of infection, of maternal immune response and foetal vulnerability and of resilience factors to hostile events. The derailed neuroimmune crosstalk through the placenta initiates and maintains a chronic foetal neuroglial activation, eventually causing the alteration of neurogenesis, migration, synapse formation and pruning. The importance of pregnancy can also allow early immune interventions, which can significantly reduce the increasing risk of ASD and its heavy social burden.
Collapse
Affiliation(s)
- Stefano Sotgiu
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| | - Salvatorica Manca
- Unità Operativa di Neuropsichiatria Infanzia e Adolescenza (UONPIA) ASSL Sassari Sassari Italy
| | - Antonella Gagliano
- Child & Adolescent Neuropsychiatry Unit Department of Biomedical Sciences University of Cagliari Cagliari Italy
| | - Alessandra Minutolo
- Child & Adolescent Neuropsychiatry Unit Department of Biomedical Sciences University of Cagliari Cagliari Italy
| | - Maria Clotilde Melis
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| | - Giulia Pisuttu
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| | - Chiara Scoppola
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| | | | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi - ONLUS Milan Italy.,Department of Pathophysiology and Transplantation University of Milano Milan Italy
| | | | - Alessandra Carta
- Unit of Child Neuropsychiatry Department of Medical Surgical and Experimental Sciences University of Sassari Sassari Italy
| |
Collapse
|
47
|
Fitzgerald E, Hor K, Drake AJ. Maternal influences on fetal brain development: The role of nutrition, infection and stress, and the potential for intergenerational consequences. Early Hum Dev 2020; 150:105190. [PMID: 32948364 PMCID: PMC7481314 DOI: 10.1016/j.earlhumdev.2020.105190] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
An optimal early life environment is crucial for ensuring ideal neurodevelopmental outcomes. Brain development consists of a finely tuned series of spatially and temporally constrained events, which may be affected by exposure to a sub-optimal intra-uterine environment. Evidence suggests brain development may be particularly vulnerable to factors such as maternal nutrition, infection and stress during pregnancy. In this review, we discuss how maternal factors such as these can affect brain development and outcome in offspring, and we also identify evidence which suggests that the outcome can, in many cases, be stratified by socio-economic status (SES), with individuals in lower brackets typically having a worse outcome. We consider the relevant epidemiological evidence and draw parallels to mechanisms suggested by preclinical work where appropriate. We also discuss possible transgenerational effects of these maternal factors and the potential mechanisms involved. We conclude that modifiable factors such as maternal nutrition, infection and stress are important contributors to atypical brain development and that SES also likely has a key role.
Collapse
Affiliation(s)
- Eamon Fitzgerald
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Kahyee Hor
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Amanda J Drake
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
48
|
Maternal Immunity in Autism Spectrum Disorders: Questions of Causality, Validity, and Specificity. J Clin Med 2020; 9:jcm9082590. [PMID: 32785127 PMCID: PMC7464885 DOI: 10.3390/jcm9082590] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders with unknown heterogeneous aetiologies. Epidemiological studies have found an association between maternal infection and development of ASD in the offspring, and clinical findings reveal a state of immune dysregulation in the pre- and postnatal period of affected subjects. Maternal immune activation (MIA) has been proposed to mediate this association by altering fetal neurodevelopment and leading to autism. Although animal models have supported a causal link between MIA and development of ASD, their validity needs to be explored. Moreover, considering that only a small proportion of affected offspring develop autism, and that MIA has been implicated in related diseases such as schizophrenia, a key unsolved question is how disease specificity and phenotypic outcome are determined. Here, we have integrated preclinical and clinical evidence, including the use of animal models for establishing causality, to explore the role of maternal infections in ASD. A proposed priming/multi-hit model may offer insights into the clinical heterogeneity of ASD, its convergence with related disorders, and therapeutic strategies.
Collapse
|
49
|
Sala R, Amet L, Blagojevic-Stokic N, Shattock P, Whiteley P. Bridging the Gap Between Physical Health and Autism Spectrum Disorder. Neuropsychiatr Dis Treat 2020; 16:1605-1618. [PMID: 32636630 PMCID: PMC7335278 DOI: 10.2147/ndt.s251394] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is a highly complex and heterogeneous developmental disorder that affects how individuals communicate with other people and relate to the world around them. Research and clinical focus on the behavioural and cognitive manifestations of ASD, whilst important, have obscured the recognition that ASD is also commonly associated with a range of physical and mental health conditions. Many physical conditions appear with greater frequency in individuals with ASD compared to non-ASD populations. These can contribute to a worsening of social communication and behaviour, lower quality of life, higher morbidity and premature mortality. We highlight some of the key physical comorbidities affecting the immune and the gastrointestinal systems, metabolism and brain function in ASD. We discuss how healthcare professionals working with individuals with ASD and parents/carers have a duty to recognise their needs in order to improve their overall health and wellbeing, deliver equality in their healthcare experiences and reduce the likelihood of morbidity and early mortality associated with the condition.
Collapse
Affiliation(s)
- Regina Sala
- Centre for Psychiatry, Wolfson Institute, Barts & The London School of Medicine & Dentistry Queen Mary University of London, London, UK
| | | | | | - Paul Shattock
- Education & Services for People with Autism, Sunderland, UK
| | - Paul Whiteley
- Education & Services for People with Autism Research, Sunderland, UK
| |
Collapse
|
50
|
Baines KJ, Hillier DM, Haddad FL, Rajakumar N, Schmid S, Renaud SJ. Maternal Immune Activation Alters Fetal Brain Development and Enhances Proliferation of Neural Precursor Cells in Rats. Front Immunol 2020; 11:1145. [PMID: 32582210 PMCID: PMC7295982 DOI: 10.3389/fimmu.2020.01145] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Maternal immune activation (MIA) caused by exposure to pathogens or inflammation during critical periods of neurodevelopment is a major risk factor for behavioral deficits and psychiatric illness in offspring. A spectrum of behavioral abnormalities can be recapitulated in rodents by inducing MIA using the viral mimetic, PolyI:C. Many studies have focused on long-term changes in brain structure and behavioral outcomes in offspring following maternal PolyI:C exposure, but acute changes in prenatal development are not well-characterized. Using RNA-Sequencing, we profiled acute transcriptomic changes in rat conceptuses (decidua along with nascent embryo and placenta) after maternal PolyI:C exposure during early gestation, which enabled us to capture gene expression changes provoked by MIA inclusive to the embryonic milieu. We identified a robust increase in expression of genes related to antiviral inflammation following maternal PolyI:C exposure, and a corresponding decrease in transcripts associated with nervous system development. At mid-gestation, regions of the developing cortex were thicker in fetuses prenatally challenged with PolyI:C, with females displaying a thicker ventricular zone and males a thicker cortical mantle. Along these lines, neural precursor cells (NPCs) isolated from fetal brains prenatally challenged with PolyI:C exhibited a higher rate of self-renewal. Expression of Notch1 and the Notch ligand, delta-like ligand 1, which are both highly implicated in maintenance of NPCs and nervous system development, was increased following PolyI:C exposure. These results suggest that MIA elicits rapid gene expression changes within the conceptus, including repression of neurodevelopmental pathways, resulting in profound alterations in fetal brain development.
Collapse
Affiliation(s)
- Kelly J Baines
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Dendra M Hillier
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Faraj L Haddad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Nagalingam Rajakumar
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, University of Western Ontario, London, ON, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|