1
|
Davis LK, Ince LM, Gullapalli S, Fonken LK. Neuroimmune and behavioral changes elicited by maternal immune activation in mice are ameliorated by early postnatal immune stimulation. Brain Behav Immun 2025; 127:375-386. [PMID: 40081778 DOI: 10.1016/j.bbi.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Though the etiology of autism spectrum disorder (ASD) is complex and not fully understood, it is believed that genetic risk factors, coupled with early life inflammation may predispose individuals to develop ASD. Maternal immune activation (MIA) is associated with increased incidence of ASD in offspring; however, not all mothers who experience inflammation during pregnancy have children with autism, suggesting that MIA may act as a disease primer that results in ASD pathology when paired with additional inflammatory insults. Here, we tested the hypothesis that MIA is a disease primer by using a two-hit model that combined MIA with a secondary immune stimulation in early life. C57BL/6J mouse dams were treated with polyinosinic-polycytidylic acid (Poly(I:C)) at embyronic day 12.5, and a subset of litters were then treated with the endotoxin lipopolysaccharide (LPS) four days after birth. Offspring were assessed in young adulthood for changes in behavior including sociability, repetitive-like behaviors, and anxiety-like behaviors. Flow cytometry was performed in adulthood to assess changes in immune cell populations in the periphery and in the brain. MIA increased repetitive-like behaviors in male mice and decreased sociability in both sexes. Unexpectedly, the secondary immune stimulation with LPS did not exacerbate changes in social and repetitive-like behaviors in either sex. MIA also altered distribution of cytotoxic CD8 + T cell populations in the periphery and brain of both sexes: CD8 + T cells were elevated in thymus but reduced in spleen, lymph, and brain. Additionally, MIA altered microglia activity in a region-specific manner in male mice, which was also not exacerbated but rather ameliorated when combined with LPS. Our results demonstrate that changes in repetitive-like and social behaviors that are induced by MIA in male mice are not exacerbated by subsequent inflammatory challenge and highlights the importance of considering the timing of stressors in the appearance of developmental pathology.
Collapse
Affiliation(s)
- Lourdes K Davis
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Interdisciplinary Neuroscience Program, University of Texas at Austin, Austin, TX 78712, USA.
| | - Louise M Ince
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Sriya Gullapalli
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Laura K Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Interdisciplinary Neuroscience Program, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
2
|
Delorme TC, Arcego DM, Penichet D, O'Toole N, Huebener N, Silveira PP, Srivastava LK, Cermakian N. Large-scale effects of prenatal inflammation and early life circadian disruption in mice: Implications for neurodevelopmental disorders. Brain Behav Immun 2025; 127:409-422. [PMID: 40118225 DOI: 10.1016/j.bbi.2025.03.023] [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: 11/17/2024] [Revised: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Around 80 % of individuals with neurodevelopmental disorders such as schizophrenia and autism spectrum disorders experience disruptions in sleep/circadian rhythms. We explored whether environmental circadian disruption interacts with prenatal infection, a risk factor for neurodevelopmental disorders, to induce sex-specific deficits in mice. A maternal immune activation (MIA) protocol was used by injecting pregnant mice with viral mimic poly IC or saline at E9.5. Juvenile/adolescent male and female offspring (3-7 weeks old) were then subjected to a standard light:dark cycle (12:12LD) or to constant light (LL). Significant interactions between treatment (MIA, control) and lighting (12:12LD, LL) were evident in behaviors related to cognition, anxiety, and sociability. This pattern persisted in our RNA sequencing analysis of the dorsal hippocampus, where poly IC exposure resulted in numerous differentially expressed genes (DEGs) in males, while exposure to both poly IC and LL led to a marked reduction in DEGs. Through WGCNA analysis, many significant gene modules were found to be positively associated with poly IC (vs. saline) and LL (vs. LD) in males (fewer in females). Many of the identified hub-bottleneck genes were homologous to human genes associated with sleep/circadian rhythms and neurodevelopmental disorders as revealed by GWA studies. The MIA- and LL-associated modules were enriched in microglia gene signatures, which was paralleled by trends of effects of each of the factors on microglia morphology. In conclusion, in a mouse model of prenatal infection, circadian disruption induced by LL during adolescence acts as a modulator of the effects of MIA at behavioral, cellular, and molecular levels.
Collapse
Affiliation(s)
- Tara C Delorme
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Danusa M Arcego
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Danae Penichet
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Nicholas O'Toole
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada
| | - Nikki Huebener
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada
| | - Patrícia P Silveira
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Lalit K Srivastava
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada.
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada.
| |
Collapse
|
3
|
Gardner RM, Brynge M, Sjöqvist H, Dalman C, Karlsson H. Maternal Immune Activation and Autism in Offspring: What Is the Evidence for Causation? Biol Psychiatry 2025; 97:1127-1138. [PMID: 39581290 DOI: 10.1016/j.biopsych.2024.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/21/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
The maternal immune activation hypothesis has gained attention over the past 2 decades as a potential contributor to the etiology of autism. This hypothesis posits that maternal conditions associated with inflammation during pregnancy may increase the risk of autism in offspring. Autism is highly heritable, and causal environmental contributors to autism largely remain elusive. We review studies on maternal conditions during pregnancy, all associated with some degree of systemic inflammation, namely maternal infections, autoimmunity, and high body mass index. We also review studies of inflammatory markers in biological samples collected from mothers during pregnancy or from neonates and their relationship with autism assessed in children later in life. Recent reports indicate familial clustering of autism, autoimmunity, and infections, as well as genetic correlations between autism and aspects of immune function. Given this literature, there is an apparent risk of confounding of the reported associations between inflammatory exposures and autism by familial genetic factors in both clinical and epidemiological cohort studies. We highlight recent studies that have attempted to address potential confounding to assess evidence of causal effects of inflammation during early life in autism.
Collapse
Affiliation(s)
- Renee M Gardner
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Martin Brynge
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Hugo Sjöqvist
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Christina Dalman
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Kwak-Kim J, Maier CC, Villano CM, Bowman CJ, Brennan FR, Stanislaus D, Hillegas A, Krayer J, Prell RA, Papenfuss TL, Cauvin A, Gamse J, Dahlman A, Enright B, Leshin L, Rao GK, Helms W, Fuller CL, Yang X, Chen C, Mitchell-Ryan S. Assessing the impact and risk of immunomodulatory compounds on pregnancy. J Reprod Immunol 2025; 169:104453. [PMID: 39999662 DOI: 10.1016/j.jri.2025.104453] [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: 10/16/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
There have been remarkable advancements in understanding the complex and dynamic immune biological processes engaged during all stages of pregnancy. Exquisite control of immune processes is critical to successful outcome in all stages of pregnancy from ovulation to birth. There are many immunomodulatory therapeutics that may offer beneficial treatment options for a variety of diseases (e.g., inflammation/autoimmunity, cancer) to patients that are or desire to become pregnant. It is important to understand the potential for these immunomodulatory therapeutics to alter the critical immune processes in pregnancy to inform clinical risk relative to successful pregnancy. The Health and Environmental Sciences Institute-Developmental and Reproductive Toxicology/Immuno-safety Technical Committee (HESI DART/ITC) conducted a survey on approaches to assess adverse pregnancy outcomes with immunomodulators. HESI DART/ITC also organized a workshop for an extended discussion on immune mechanisms during pregnancy, the adequacy of current tools/methodologies to identify concerns for potential pregnancy hazards from immunomodulatory therapies, ways to identify and address scientific gaps, and global regulatory considerations across various immunomodulatory modalities and indications. In this manuscript we summarize learnings from these efforts to characterize risk within this patient population, promote more informed treatment decisions, and enable safer pharmacological interventions during pregnancy.
Collapse
Affiliation(s)
- Joanne Kwak-Kim
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Vernon Hills, IL, USA
| | | | - Caren M Villano
- Boehringer Ingelheim, Nonclinical Drug Safety, Ridgefield, CT, USA.
| | | | - Frank R Brennan
- Novartis Institute of BioMedical Research, Preclinical Safety (PCS), Basel, Switzerland
| | | | | | - John Krayer
- Johnson and Johnson, Non-clinical Safety, Springhouse, PA, USA
| | - Rodney A Prell
- Genentech, Inc., Department of Safety Assessment, South San Francisco, CA, USA
| | | | - Annick Cauvin
- UCB Biopharma SRL, Nonclinical Safety Evaluation, Brussels, Belgium
| | - Joshua Gamse
- Genmab, Non-Clinical Safety & Toxicology, Plainsboro, NJ, USA
| | - Anna Dahlman
- Genmab, Non-Clinical Safety & Toxicology, Copenhagen, Denmark
| | - Brian Enright
- AbbVie Inc., Preclinical Safety, North Chicago, IL, USA
| | - Lawrence Leshin
- United States Food and Drug Administration, CDER-OND-OII-DRTM, Silver Spring, MD, USA
| | - Gautham K Rao
- Genentech, Inc., Department of Safety Assessment, South San Francisco, CA, USA
| | | | | | - Xiuhua Yang
- The First Hospital of China Medical University, Department of Obstetrics and Gynecology, Shenyang, Liaoning, PR China
| | - Connie Chen
- The Health and Environmental Sciences Institute, Washington, DC, USA
| | | |
Collapse
|
5
|
Nguyen LT, Nguyen PM, Nguyen HP, Bui HT, Dao LTM, Van Pham M, Hoang CK, Nguyen PT, Nguyen TTP, Nguyen ATP, Hoang VT, Bui HTP, Vuong NK, Van Ngo D. Outcomes of autologous bone marrow mononuclear cell administration combined with educational intervention in the treatment of autism spectrum disorder: a randomized, open-label, controlled phase II clinical trial. Stem Cell Res Ther 2025; 16:268. [PMID: 40442857 PMCID: PMC12123819 DOI: 10.1186/s13287-025-04404-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 05/19/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND This study evaluated the effectiveness of intrathecal autologous bone marrow mononuclear cell (BMMNC) therapy combined with education compared with education alone for the treatment of autism spectrum disorder (ASD). METHODS Fifty-four children with ASD, aged three to seven years, were randomly assigned to two groups. Fifty patients completed the study (25 patients per group). The cell therapy (CT) group received two BMMNC infusions six months apart along with an educational intervention, while the control group received education only. Efficacy outcomes were assessed at baseline, two, six, and 12 months, based on: (1) changes in ASD severity evaluated through the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5), the Childhood Autism Rating Scale (CARS), and the Clinical Global Impression-Severity (CGI-S) scale scores and (2) improvements in social interaction, adaptive behavior, and daily living skills measured by the Vineland Adaptive Behavior Scales (VABS-II) and Clinical Global Impression-Improvement (CGI-I) scale scores. RESULTS At 12 months, the CT group presented a 48.0% reduction in individuals classified at the most severe DSM-5 level compared with 8.0% in the control group (p = 0.004). The CARS scores were significantly lower in the CT group (-5.9 points) than in the control group (-1.5 points) (p < 0.0001). Similarly, the CT group exhibited greater improvement in CGI-S scores (-1.5 points) than did the control group (-0.1 points) (p < 0.0001). The VABS-II scores increased by 8.5 points in the CT group versus 1.4 points in the control group (p < 0.0001). Finally, the CGI-I scores improved from 2.8 to 2.0 in the CT group but worsened from 3.0 to 3.5 in the control group (p < 0.0001). CONCLUSIONS Intrathecal BMMNC combined with an educational intervention improved disease severity and adaptability more than education alone in children with ASD. TRIAL REGISTRATION clinicaltrials.gov, NCT05307536. Date registered 12 February 2022. http://clinicaltrials.gov/study/NCT05307536 .
Collapse
Affiliation(s)
- Liem Thanh Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), College of Health Sciences, VinUniversity, Vinhomes Ocean Park, Gia Lam, Hanoi, 100000, Vietnam.
- Vinmec Times City International Hospital, Vinmec Health Care System, 458 Minh Khai Street, Hai Ba Trung, Hanoi, 100000, Vietnam.
| | - Phuong Mai Nguyen
- Department of Pediatrics, Faculty of Medicine Haiphong, Hai Phong University of Medicine and Pharmacy, Hanoi, 100000, Vietnam
| | - Hoang-Phuong Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), College of Health Sciences, VinUniversity, Vinhomes Ocean Park, Gia Lam, Hanoi, 100000, Vietnam
| | - Hau Thi Bui
- Vinmec Times City International Hospital, Vinmec Health Care System, 458 Minh Khai Street, Hai Ba Trung, Hanoi, 100000, Vietnam
| | - Lan Thi Mai Dao
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), College of Health Sciences, VinUniversity, Vinhomes Ocean Park, Gia Lam, Hanoi, 100000, Vietnam
| | - Minh Van Pham
- Rehabilitation Department, Ha Noi Medical University, 1 Ton That Tung Road, Dong Da, Hanoi, 100000, Vietnam
- Ha Noi Rehabilitation Hospital, 35 Le Van Thiem, Thanh Xuan, Hanoi, 100000, Vietnam
| | - Chi Khanh Hoang
- Ha Noi Rehabilitation Hospital, 35 Le Van Thiem, Thanh Xuan, Hanoi, 100000, Vietnam
| | - Phuong Thi Nguyen
- Child Integration Education Center, 52/2 Yen Lac, Hai Ba Trung, Hanoi, 100000, Vietnam
| | - Thao Thi Phuong Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), College of Health Sciences, VinUniversity, Vinhomes Ocean Park, Gia Lam, Hanoi, 100000, Vietnam
| | - Anh Thi Phuong Nguyen
- Vinmec Times City International Hospital, Vinmec Health Care System, 458 Minh Khai Street, Hai Ba Trung, Hanoi, 100000, Vietnam
| | - Van Thi Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), College of Health Sciences, VinUniversity, Vinhomes Ocean Park, Gia Lam, Hanoi, 100000, Vietnam
| | - Hoa Thi Phuong Bui
- Vinmec Times City International Hospital, Vinmec Health Care System, 458 Minh Khai Street, Hai Ba Trung, Hanoi, 100000, Vietnam
| | - Ngan Kim Vuong
- Vinmec Times City International Hospital, Vinmec Health Care System, 458 Minh Khai Street, Hai Ba Trung, Hanoi, 100000, Vietnam
| | - Doan Van Ngo
- Vinmec Times City International Hospital, Vinmec Health Care System, 458 Minh Khai Street, Hai Ba Trung, Hanoi, 100000, Vietnam
| |
Collapse
|
6
|
McEwan F, Kambara C, Lorusso JM, Harte MK, Glazier JD, Hager R. Association between redox dysregulation and vulnerability to cognitive deficits induced by maternal immune activation. Transl Psychiatry 2025; 15:184. [PMID: 40419496 DOI: 10.1038/s41398-025-03398-0] [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: 09/22/2024] [Revised: 05/01/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Exposure to maternal immune activation (MIA) in utero is a major risk factor for neurodevelopmental disorders, including schizophrenia. However, a proportion of individuals are resilient to developing schizophrenia following exposure to MIA, which has also been reported in animal models of MIA. The molecular mechanisms leading to resilient and vulnerable behavioural phenotypes remain poorly understood, and we currently lack reliable blood biomarkers that predict resilience or vulnerability. Redox dysregulation, caused by an imbalance between oxidative stress and antioxidant defence mechanisms, has recently been predicted to be central to the pathogenesis of schizophrenia. Here, we use a poly(I:C)-induced MIA model of schizophrenia to investigate mechanisms underlying cognitive dysfunction and redox dysregulation in resilient and vulnerable individuals. We show that activity of the antioxidant enzyme superoxide dismutase (SOD) was reduced in the plasma of poly(I:C) offspring with a cognitive deficit, in contrast to individuals with typical cognition during both adolescence and adulthood. However, SOD activity in the hippocampus was not significantly different between vulnerable and resilient offspring. In addition, the lipid peroxidation marker malondialdehyde (MDA) and the pro-inflammatory cytokine IL-6 were not differentially expressed within the hippocampus or plasma of vulnerable poly(I:C) offspring. Our results suggest that reduced plasma SOD activity may be a potential blood biomarker to identify resilience or vulnerability to MIA-induced cognitive deficits. Further research is necessary to determine if reduced antioxidant capacity is present in plasma prior to symptom presentation and to understand if this predicts redox dysregulation in the brain.
Collapse
Affiliation(s)
- Francesca McEwan
- Division of Evolution, Infection, and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom.
| | - Chiho Kambara
- Division of Evolution, Infection, and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Jarred M Lorusso
- Division of Evolution, Infection, and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom
- School of Humanities and Social Science, University of Brighton, Brighton, BN2 4AT, United Kingdom
| | - Michael K Harte
- Division of Pharmacy & Optometry, School of Health Sciences, Geoffrey Jefferson Brain Research Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Jocelyn D Glazier
- Division of Evolution, Infection, and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Reinmar Hager
- Division of Evolution, Infection, and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| |
Collapse
|
7
|
Villarin JM, Kellendonk C. An ace in the hole? Opportunities and limits of using mice to understand schizophrenia neurobiology. Mol Psychiatry 2025:10.1038/s41380-025-03060-7. [PMID: 40405017 DOI: 10.1038/s41380-025-03060-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 05/02/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025]
Abstract
In applying model organisms to study the neurobiology of mental disorders, rodents offer unique potential for probing, with high spatiotemporal resolution, the neural and molecular mechanisms underlying behavior in a mammalian system. Furthermore, investigators can wield exceptional power to manipulate genes, molecules, and circuits in mice to pin down causal relationships. While these advantages have allowed us to understand much more deeply than ever before the brain mechanisms regulating complex behaviors, the impact of rodent models on developing therapeutic strategies for psychiatric disorders has remained thus far limited. Herein, we will discuss the opportunities and limits of using mouse models in the context of schizophrenia, a complex psychiatric disorder with strong genetic basis that poses various unmet clinical needs calling out for basic science research. We review approaches for employing behavioral, genetic, and circuit-based methods in rodents to inform schizophrenia symptomatology, pathophysiology, and, ultimately, treatment.
Collapse
Affiliation(s)
- Joseph M Villarin
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA.
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA.
| | - Christoph Kellendonk
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA.
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA.
- Department of Molecular Pharmacology & Therapeutics, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
8
|
Wu X, Cao T, Ye J, Shi R, Bao X, Ge Y, Li D, Hao S, Liu F, Liu X. Supplementation of 2'-Fucosyllactose during the Growth Period Improves Neurodevelopmental Disorders in Offspring Mice Induced by Maternal Immune Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12292-12307. [PMID: 40350763 DOI: 10.1021/acs.jafc.5c01184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Autism spectrum disorder is a serious neurodevelopmental disorder whose early onset significantly affects an individual's social interactions and cognitive function. Recent research suggests that modulating the gut microbiota could be a potential intervention strategy for autism spectrum disorder symptoms. 2'-Fucosyllactose has been identified as a regulator of gut microbiota homeostasis, however, its effectiveness in addressing autism spectrum disorder remains unclear. In this study, the effects of daily supplementation of 2'-FL in 3-week-old male offspring mice for 5 weeks were examined. The results showed that 2'-fucosyllactose significantly improved autism spectrum disorder-like behavioral deficits. Furthermore, supplementation with 2'-fucosyllactose restored intestinal barrier integrity and increased relative abundance of beneficial gut bacteria, particularly Akkermansia and Bifidobacterium that are closely related to bile acid metabolism. Notably, 2'-fucosyllactose treatment elevated the content of bile acids and upregulated the expression of bile acid receptors in the brain. Co-housing experiments further confirmed the crucial role of gut microbiota in mediating the beneficial effects of 2'-fucosyllactose. Overall, this study suggests that 2'-fucosyllactose could alleviate maternal immune activation-induced behavioral deficits and neuroinflammation through the regulation of the gut-brain axis, offering potential therapeutic value.
Collapse
Affiliation(s)
- Xiaoning Wu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Tengzheng Cao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiaowei Bao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yunshu Ge
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Dongning Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Shijin Hao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Feitong Liu
- H&H Research, China Research and Innovation Center, H&H Group, Guangzhou 510700, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
9
|
Li X, Ma Y, Hu F. Decoding the neuro-immune crosstalk a path to early intervention for neurodevelopmental disorders. Pediatr Res 2025:10.1038/s41390-025-04135-5. [PMID: 40382468 DOI: 10.1038/s41390-025-04135-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/20/2025]
Affiliation(s)
- Xin Li
- School of medicine, Wuhan University of Science and Technology, Wuhan City, Hubei Province, China
| | - Yuan Ma
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei Province, China
| | - Feifei Hu
- School of medicine, Wuhan University of Science and Technology, Wuhan City, Hubei Province, China.
| |
Collapse
|
10
|
Kim K, Abramishvili D, Du S, Papadopoulos Z, Cao J, Herz J, Smirnov I, Thomas JL, Colonna M, Kipnis J. Meningeal lymphatics-microglia axis regulates synaptic physiology. Cell 2025; 188:2705-2719.e23. [PMID: 40120575 DOI: 10.1016/j.cell.2025.02.022] [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: 06/14/2024] [Revised: 12/30/2024] [Accepted: 02/21/2025] [Indexed: 03/25/2025]
Abstract
Meningeal lymphatics serve as an outlet for cerebrospinal fluid, and their dysfunction is associated with various neurodegenerative conditions. Previous studies have demonstrated that dysfunctional meningeal lymphatics evoke behavioral changes, but the neural mechanisms underlying these changes have remained elusive. Here, we show that prolonged impairment of meningeal lymphatics alters the balance of cortical excitatory and inhibitory synaptic inputs, accompanied by deficits in memory tasks. These synaptic and behavioral alterations induced by lymphatic dysfunction are mediated by microglia, leading to increased expression of the interleukin 6 gene (Il6). IL-6 drives inhibitory synapse phenotypes via a combination of trans- and classical IL-6 signaling. Restoring meningeal lymphatic function in aged mice reverses age-associated synaptic and behavioral alterations. Our findings suggest that dysfunctional meningeal lymphatics adversely impact cortical circuitry through an IL-6-dependent mechanism and identify a potential target for treating aging-associated cognitive decline.
Collapse
Affiliation(s)
- Kyungdeok Kim
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| | - Daviti Abramishvili
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Siling Du
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Zachary Papadopoulos
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA; Neuroscience Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jay Cao
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jasmin Herz
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Igor Smirnov
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Paris Brain Institute, Université Pierre et Marie Curie Paris 06, UMRS1127, Sorbonne Université, Paris, France
| | - Marco Colonna
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
11
|
Velloso FJ, Zaritsky R, Houbeika RY, Rios N, Levison SW. Interleukin-6 produces behavioral deficits in pre-pubescent mice independent of neuroinflammation. Brain Behav Immun 2025; 126:275-288. [PMID: 39984136 DOI: 10.1016/j.bbi.2025.02.009] [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: 05/09/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025] Open
Abstract
Maternal inflammation during pregnancy increases the offspring's risk of developing autism, ADHD, schizophrenia, and depression. Epidemiologic studies have demonstrated that maternal infections stimulate the production of interleukin-6 (IL-6), which can cross the placenta and fetal blood-brain barrier to alter brain development with functional and behavioral consequences. To model the effects of increased IL-6 between weeks 24-30 of human gestation, we injected male and female mice with 75 ng IL-6 twice daily, from P3 to P6. Our published studies have shown that this increases circulating IL-6 two-fold, alters post-pubescent ultrasonic vocalization patterns, reduces sociability, and increases self-grooming. However, most neurodevelopmental disorders in humans manifest in children as young as 2 years of age. Hence, a critical unexplored question is whether behavioral changes in immune activation models can be detected in pre-pubescent mice. Therefore, we evaluated early communication, sociability, and repetitive behaviors in pre-pubescent mice following the IL-6 treatment. A second open question is whether the cellular and behavioral changes are secondary to systemic or neuroinflammation. To address this question, we profiled 18 cytokines and chemokines in the circulation and CNS and evaluated eight immune cell types in P7 male and female brains following systemic IL-6 administration. We found an increase in ultrasonic vocalizations with simpler morphologies produced by the IL-6-injected male pups and a decrease in frequency in the female vocalizations upon removal from the nest at P7. The IL-6-treated male pups also socially interacted less when introduced to a novel mouse vs. controls as juveniles and spent almost twice as much time grooming themselves, a phenotype not present in the females. Tactile sensitivity was also increased, but only in the IL-6-treated female mice. The IL-6-treated mice had increased circulating IL-6 and IL-7 and reduced IL-13 at P7 that were no longer elevated at P14. There were no changes in brain levels of IL-6, IL-10, IL-13 or IL-17A mRNAs at P7. Altogether, these studies show that changes in the three core behavioral domains associated with several psychiatric disorders can be detected early in pre-pubescent mice following a transient developmental increase in IL-6. Yet, these behavioral alterations do not require neuroinflammation.
Collapse
Affiliation(s)
- Fernando Janczur Velloso
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| | - Rebecca Zaritsky
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| | - Rouba Y Houbeika
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| | - Nicolas Rios
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| | - Steven W Levison
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| |
Collapse
|
12
|
Goldstein JM, Konishi K, Aroner S, Lee H, Remington A, Chitnis T, Buka SL, Hornig M, Tobet SA. Prenatal immune origins of brain aging differ by sex. Mol Psychiatry 2025; 30:1887-1896. [PMID: 39567743 PMCID: PMC12014477 DOI: 10.1038/s41380-024-02798-w] [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: 08/07/2023] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024]
Abstract
With an increasing aging population and Alzheimer's disease tsunami, it is critical to identify early antecedents of brain aging to target for intervention and prevention. Women and men develop and age differently, thus using a sex differences lens can contribute to identification of early risk biomarkers and resilience. There is growing evidence for fetal antecedents to adult memory impairments, potentially through disruption of maternal prenatal immune pathways. Here, we hypothesized that in utero exposure to maternal pro-inflammatory cytokines will have sex-dependent effects on specific brain circuitry regulating offspring's memory and immune function that will be retained across the lifespan. Using a unique prenatal cohort, we tested this in 204 adult offspring, equally divided by sex, who were exposed/unexposed to an adverse in utero maternal immune environment and followed into early midlife (~age 50). Functional magnetic resonance imaging results showed exposure to pro-inflammatory cytokines in utero (i.e., higher maternal IL-6 and TNF-α levels) was significantly associated with sex differences in brain activity and connectivity underlying memory circuitry and performance and with a hyperimmune state, 50 years later. In contrast, the anti-inflammatory cytokine, IL-10 alone, was not significantly associated with memory circuitry in midlife. Predictive validity of prenatal exposure was underscored by significant associations with age 7 academic achievement, also associated with age 50 memory performance. Results uniquely demonstrated that adverse levels of maternal in utero pro-inflammatory cytokines during a critical period of the sexual differentiation of the brain produced long-lasting effects on immune function and memory circuitry/function from childhood to midlife that were sex-dependent, brain region-specific, and, within women, reproductive stage-dependent.
Collapse
Affiliation(s)
- Jill M Goldstein
- Department of Psychiatry, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Kyoko Konishi
- Department of Psychiatry, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, MA, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sarah Aroner
- Department of Psychiatry, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, MA, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hang Lee
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Biostatistics, Massachusetts General Hospital, Boston, MA, USA
| | - Anne Remington
- Department of Psychiatry, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, MA, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tanuja Chitnis
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and MGH, Harvard Medical School, Boston, MA, USA
| | - Stephen L Buka
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Epidemiology and Population Health, Brown University, Providence, RI, USA
| | - Mady Hornig
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Stuart A Tobet
- Department of Psychiatry, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, MA, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Biomedical Sciences and School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
13
|
Hill RA, Gibbons A, Suwakulsiri W, Taseska A, Darke H, Malhotra A, Yee H, Fahey M, Hunt RW, Lim I, Palmer K, Sundram S. Investigating the impact of severe maternal SARS-CoV-2 infection on infant DNA methylation and neurodevelopment. Mol Psychiatry 2025; 30:1976-1984. [PMID: 39478169 DOI: 10.1038/s41380-024-02808-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 04/24/2025]
Abstract
Maternal infections during pregnancy can increase the risk to offspring of developing a neurodevelopmental disorder. Given the global prevalence and severity of infection with Severe Acute Respiratory Syndrome related Coronavirus 2 (SARS-CoV-2), the objective of this study was to determine if in utero exposure to severe maternal SARS-CoV-2 infection alters infant neurodevelopmental outcomes at 12 months and to identify potential biological markers of adverse infant outcomes. Mother-infant dyads exposed to severe SARS-CoV-2 infection (requiring hospitalization) during pregnancy and age and sociodemographic matched control dyads were recruited from Monash Medical Centre, Australia in 2021/22 and prospectively assessed over 12 months. Maternal serum cytokine levels and Edinburgh Postnatal Depression Scale (EPDS) scores were assessed at birth. DNA methylation was assessed from infant buccal swabs at birth (Illumina EPIC BeadChip). Infant neurodevelopmental outcomes at 12 months were assessed using the Ages and Stages Questionnaire (ASQ-3). Mothers exposed to severe SARS-CoV-2 exhibited elevated serum IL-6 and IL-17A and higher EPDS scores than controls at birth. Infants exposed to severe SARS-CoV-2 in utero demonstrated over 3000 significant differentially methylated sites within their genomes compared to non-exposed (adjusted p-value < 0.05), including genes highly relevant to ASD and synaptic pathways. At 12 months, severe SARS-CoV-2 exposed infants scored lower on the ASQ-3 than non-exposed infants, and communication and problem-solving scores negatively correlated with maternal IL-6 levels at birth. DNA methylation changes therefore unveil potential mechanisms linking infection exposure to delayed neurodevelopment and maternal serum IL-6 levels may be a potential biomarker of child developmental delay. Mothers exposed to severe SARS-CoV-2 infections show elevated pro-inflammatory cytokines. Infants exposed in utero to severe SARS-CoV-2 infection show altered DNA methylation at birth and delayed development at 12 months of age. Created in Biorender.com.
Collapse
Affiliation(s)
- Rachel A Hill
- Department of Psychiatry, Monash University, Clayton, Vic, Australia.
| | - Andrew Gibbons
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | | | - Angela Taseska
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | - Hayley Darke
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Clayton, Vic, Australia
- Monash Children's Hospital, Clayton, Vic, Australia
| | - Hnin Yee
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | - Michael Fahey
- Department of Paediatrics, Monash University, Clayton, Vic, Australia
- Monash Children's Hospital, Clayton, Vic, Australia
| | - Rod W Hunt
- Department of Paediatrics, Monash University, Clayton, Vic, Australia
- Monash Children's Hospital, Clayton, Vic, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Parkville, Vic, Australia
| | - Izaak Lim
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | - Kirsten Palmer
- Monash Women's, Monash Health, Clayton, Vic, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Vic, Australia
| | - Suresh Sundram
- Department of Psychiatry, Monash University, Clayton, Vic, Australia.
- Mental Health Program, Monash Health, Clayton, Vic, Australia.
| |
Collapse
|
14
|
Mohammadzadeh P, Jepsen JRM, Lemvigh CK, Rosenberg JB, Hernández-Lorca M, Sevelsted A, Vinding R, Vahman N, Horner D, Sørensen ME, Aagaard K, Pedersen CET, Brix S, Fagerlund B, Schoos AMM, Stokholm J, Chawes B, Pantelis C, Glenthøj BY, Bønnelykke K, Ebdrup BH. Maternal interleukin 6 in pregnancy is associated with everyday, but not test-based executive functioning in 10-year-old children. Psychol Med 2025; 55:e112. [PMID: 40211088 DOI: 10.1017/s0033291725000674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
BACKGROUND Elevated maternal interleukin 6 (IL-6) during pregnancy has been associated with adverse fetal brain development and neurodevelopmental disorders, which often involve executive functioning (EF) impairments. However, the association between maternal IL-6 levels during pregnancy and EF remains largely unexplored. METHODS The COPSYCH study is based on the prospective COPSAC2010 birth cohort of 700 mother-child pairs, recruited during pregnancy. The children's executive functioning was assessed at age 10 using: (i) the Behavior Rating Inventory of Executive Function, Second Edition (BRIEF-2) parental questionnaire, and (ii) a comprehensive neuropsychological test battery. Maternal blood levels of IL-6 and hs-CRP were measured at gestational week 24. Associations between IL-6 (main analysis) and hs-CRP (secondary analysis) and EF in children at age 10 were investigated with regression models with extensive confounder adjustment. RESULTS Six hundred and four children (86% of the cohort) completed the 10-year follow-up. Higher maternal IL-6 levels were significantly associated with less efficient parental-rated executive functioning in the children: BRIEF-2 Global Executive Composite score (p = 0.003), Behavior Regulation Index (p = 0.005), Emotion Regulation Index (p=0.04), and Cognitive Regulation Index (p=0.007). Interaction analysis with sex was significant (p-value=0.01) and exploratory analyses showed that IL-6 associations to BRIEF-2 were solely driven by boys. Associations between IL-6 and neuropsychological tests, as well as associations between hs-CRP and EF outcomes, were non-significant. CONCLUSION IL-6 during pregnancy was associated with less efficient everyday EF in children at age 10. If replicated, preventive strategies targeting inflammation in pregnancy may ameliorate adverse cognitive outcomes in offspring.
Collapse
Affiliation(s)
- Parisa Mohammadzadeh
- Center for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jens Richardt Møllegaard Jepsen
- Center for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Child and Adolescent Mental Health Center, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark
| | - Cecilie K Lemvigh
- Center for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Julie B Rosenberg
- Center for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - María Hernández-Lorca
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Astrid Sevelsted
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Rebecca Vinding
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Nilo Vahman
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - David Horner
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Mikkel E Sørensen
- Center for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Kristina Aagaard
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Casper-Emil T Pedersen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Birgitte Fagerlund
- Center for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Child and Adolescent Mental Health Center, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Marie M Schoos
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
| | - Birte Y Glenthøj
- Center for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Bjørn H Ebdrup
- Center for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Shimamura T, Kitashiba M, Nishizawa K, Hattori Y. Physiological roles of embryonic microglia and their perturbation by maternal inflammation. Front Cell Neurosci 2025; 19:1552241. [PMID: 40260079 PMCID: PMC12009865 DOI: 10.3389/fncel.2025.1552241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
The interplay between the nervous and immune systems is well documented in the context of adult physiology and disease. Recent advances in understanding immune cell development have highlighted a significant interaction between neural lineage cells and microglia, the resident brain macrophages, during developmental stages. Throughout development, particularly from the embryonic to postnatal stages, diverse neural lineage cells are sequentially generated, undergo fate determination, migrate dynamically to their appropriate locations while maturing, and establish connections with their surroundings to form neural circuits. Previous studies have demonstrated that microglia contribute to this highly orchestrated process, ensuring the proper organization of brain structure. These findings underscore the need to further investigate how microglia behave and function within a broader framework of neurodevelopment. Importantly, recent epidemiological studies have suggested that maternal immune activation (MIA), triggered by various factors, such as viral or bacterial infections, environmental stressors, or other external influences, can affect neurogenesis and neural circuit formation, increasing the risk of neurodevelopmental disorders (NDDs) in offspring. Notably, many studies have revealed that fetal microglia undergo significant changes in response to MIA. Given their essential roles in neurogenesis and vascular development, inappropriate activation or disruption of microglial function may impair these critical processes, potentially leading to abnormal neurodevelopment. This review highlights recent advances in rodent models and human studies that have shed light on the behaviors and multifaceted roles of microglia during brain development, with a particular focus on the embryonic stage. Furthermore, drawing on insights from rodent MIA models, this review explores how MIA disrupts microglial function and how such disturbances may impair brain development, ultimately contributing to the onset of NDDs.
Collapse
Affiliation(s)
| | | | | | - Yuki Hattori
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
16
|
AlSharari SD, Mahmood HM, Alasmari AF, AlDhalaan HM, Alasmari F, Khan MR, Ahmad SF, Aljasham AT, Damaj IM, Alshammari MA. Nicotine Attenuates Molecular Signalings in the BTBR T + Itpr3 tf/J Mouse Model of Autism. Mol Neurobiol 2025:10.1007/s12035-025-04894-6. [PMID: 40172818 DOI: 10.1007/s12035-025-04894-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
Accumulating evidence indicates that nicotinic receptor subtypes are altered in the brains of autistic individuals, and nicotinic acetylcholine receptors (nAChRs) play essential roles in autistic profiles in BTBR T+ Itpr3tf/J mice. This study aimed to elucidate the roles of nicotine on systemic inflammatory cytokine levels and expression patterns of nicotinic receptor subtypes in the prefrontal cortex in BTBR T+ Itpr3tf/J mice. This research project characterized the effect of chronic treatment with nicotine at a dose (100 mcg/ml; po) administrated orally in drinking water over a period of fourteen days in BTBR T+ Itpr3tf/J mice, while C57BL/6 J mice were served as the controls. Following the nicotine treatment, the levels of tumor necrosis factor (TNF)-α, interferon (IFN)-γ interleukin (IL)-1β, and granulocyte-macrophage colony-stimulating factor (GM-CSF) were assessed in the serum; the levels of pro-inflammatory cytokines [interleukin (IL)-17 and interferon (IFN)-γ], on CD4+ and CD8+ T cells were evaluated in the blood. Moreover, the expression of α7, α4, and β2-nAChRs in the prefrontal cortex in BTBR T+ Itpr3tf/J mice was examined. Biochemical analysis showed that nicotine had significantly decreased the concentration of inflammatory cytokines, including TNF-α, IFN-γ, IL-1β, and GM-CSF in the serum, and reduced the expression levels of intracellular pro-inflammatory cytokines (IL-17 & IFN-γ) on CD4+ and CD8+ T cells in the blood while mecamylamine reversed the effect of IL-17+ CD4+ T cells. Nicotine administration up-regulated the expressions of α7, α4, and β2 nAChRs in the prefrontal cortex in BTBR T+ Itpr3tf/J mice. The current results indicate that nAChRs play a significant role, at least in part, in ASD and might serve as a crucial target for therapeutic interventions in ASD.
Collapse
Affiliation(s)
- Shakir D AlSharari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Hafiz M Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hesham M AlDhalaan
- Department of Neuroscience, Center for Autism Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad R Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Alanoud T Aljasham
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Imad M Damaj
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Howard CE, Cheenath M, Crouch E. The promise of cerebral organoids for neonatology. Curr Opin Pediatr 2025; 37:182-190. [PMID: 40013913 PMCID: PMC11902893 DOI: 10.1097/mop.0000000000001446] [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] [Indexed: 02/28/2025]
Abstract
PURPOSE OF REVIEW Applying discoveries from basic research to patients in the neonatal intensive care unit (NICU) is challenging given the difficulty of modeling this population in animal models, lack of translational relevance from animal models to humans, and scarcity of primary human tissue. Human cell-derived cerebral organoid models are an appealing way to address some of these gaps. In this review, we will touch on previous work to model neonatal conditions in cerebral organoids, some limitations of this approach, and recent strategies that have attempted to address these limitations. RECENT FINDINGS While modeling of neurodevelopmental disorders has been an application of cerebral organoids since their initial description, recent studies have dramatically expanded the types of brain regions and disease models available. Additionally, work to increase the complexity of organoid models by including immune and vascular cells, as well as modeling human heterogeneity with mixed donor organoids will provide new opportunities to model neonatal pathologies. SUMMARY Organoids are an attractive model to study human neurodevelopmental pathologies relevant to patients in the neonatal ICU. New technologies will broaden the applicability of these models to neonatal research and their usefulness as a drug screening platform.
Collapse
Affiliation(s)
- Clare E Howard
- Division of Newborn Medicine, Boston Children’s Hospital
| | - Manju Cheenath
- Department of Obstetrics and Gynecology, University of California, San Francisco
| | - Elizabeth Crouch
- Department of Pediatrics, University of California, San Francisco
| |
Collapse
|
18
|
Kirkpatrick M, Mandal G, Elhadidy I, Mariani N, Priestley K, Pariante CM, Borsini A. From placenta to the foetus: a systematic review of in vitro models of stress- and inflammation-induced depression in pregnancy. Mol Psychiatry 2025; 30:1689-1707. [PMID: 39639175 PMCID: PMC11919713 DOI: 10.1038/s41380-024-02866-1] [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: 04/23/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Depression in pregnancy can increase vulnerability for psychiatric disorders in the offspring, likely via the transfer of heightened maternal cortisol and cytokines to the in-utero environment. However, the precise cellular and molecular mechanisms, are largely unclear. Animal studies can represent this complex pathophysiology at a systemic level but are expensive and ethically challenging. While simpler, in vitro models offer high-throughput opportunities. Therefore, this systematic review integrates findings of in vitro models relevant to depression in pregnancy, to generate novel hypotheses and targets for intervention. METHODS The systematic analysis covered studies investigating glucocorticoid or cytokine challenges on placental or foetal neural progenitor cells (NPCs), with or without co-treatment with sex hormones. RESULTS Of the 50 included studies, 11 used placental cells and 39 NPCs; surprisingly, only one used a combination of oestrogen and cortisol, and no study combined placental cells and NPCs. In placental cells, cortisol or cytokines decreased nutrient transporter expression and steroidogenic enzyme activity, and increased cytokine production. NPCs exhibited decreases in proliferation and differentiation, via specific molecular pathways, namely, inhibition of hedgehog signalling and activation of kynurenine pathway. In these cells, studies also highlighted epigenetic priming of stress and inflammatory pathways. CONCLUSIONS Overall, results suggest that stress and inflammation not only detrimentally impact placental regulation of nutrients and hormones to the foetus, but also activate downstream pathways through increased inflammation in the placenta, ultimately eliciting adverse effects on foetal neurogenesis. Future research should investigate how sex hormones regulate these mechanisms, with the aim of developing targeted therapeutic approaches for depression in pregnancy.
Collapse
Affiliation(s)
- Madeline Kirkpatrick
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Gargi Mandal
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Ismail Elhadidy
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Nicole Mariani
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Kristi Priestley
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Alessandra Borsini
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK.
| |
Collapse
|
19
|
Li X, Cheng R, Naeem M, Nie X, Wang J, Zhao L, Liu X, Shi Z, Zhang J. Sevoflurane Inhibits the Proliferation of Neural Precursor Cells and Neural Migration of Mice by Inducing Iron Metabolism Disorders. CNS Neurosci Ther 2025; 31:e70369. [PMID: 40202153 PMCID: PMC11979790 DOI: 10.1111/cns.70369] [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: 12/19/2023] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Sevoflurane (Sev) is a volatile anesthetic and inhibits the proliferation of neural precursor cells (NPCs) and neuronal migration in the embryonic brain, thereby affecting offspring's cortical development and cognitive function. METHODS Pregnant mice were treated with 2.5% Sev. In utero, plasmids with GFP were electroporated into embryonic cortical neural precursor cells. Cell proliferation and neurite growth were detected by immunofluorescence of Ki67, pH 3, BrdU, Map2, and phalloidin labeling, respectively. Ferritin, transferrin receptor1 (TfR1), and confilin were detected by western blot. RESULTS Sev inhibited the proliferation of NPCs by down-regulating the expression of pH 3 and Ki67, and also delayed the radial migration of cortical neurons. Sev impaired the multipolar-to-bipolar transition of migrating neurons by affecting Golgi orientation. Furthermore, Sev down-regulated the expression of TfR1and increased the protein levels of ferritin heavy chain (FtH) and ferritin light chain (FtL) and caused the iron accumulation in the brain. Meanwhile, Sev induced the abnormal depolymerization and polymerization of microfilaments by increasing the ratio of p-Cofilin/Cofilin and decreasing the ratio of F-actin/G-actin. Meanwhile, Sev inhibited cortical development by decreasing the neurite growth and number of branches of neurites. DFO, an iron-chelating agent, could significantly ameliorate the inhibitory effect of Sev on the proliferation of NPCs and radial migration of projection neurons. CONCLUSIONS Sev inhibited the NPCs proliferation and neuronal migration by inducing iron metabolic dysfunction. Regulating iron homeostasis could protect the cortical development of the embryo against Sev exposure during pregnancy.
Collapse
Affiliation(s)
- Xincheng Li
- Laboratory of Molecular Iron MetabolismCollege of Life Science, Hebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Runjiao Cheng
- Laboratory of Molecular Iron MetabolismCollege of Life Science, Hebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Mahammad Naeem
- Laboratory of Molecular Iron MetabolismCollege of Life Science, Hebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Xiaoou Nie
- Laboratory of Molecular Iron MetabolismCollege of Life Science, Hebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Jiaqi Wang
- Laboratory of Molecular Iron MetabolismCollege of Life Science, Hebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Liqiang Zhao
- Laboratory of Molecular Iron MetabolismCollege of Life Science, Hebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Xiaopeng Liu
- The Second Affiliated Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Zhenhua Shi
- Laboratory of Molecular Iron MetabolismCollege of Life Science, Hebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Jianhua Zhang
- Laboratory of Molecular Iron MetabolismCollege of Life Science, Hebei Normal UniversityShijiazhuangHebei ProvinceChina
| |
Collapse
|
20
|
Mohyee RA, Elliott BL, Pike MR, Smith E, Kring AM, Olson IR, Breen EC, Cohn BA, Cirillo PM, Krigbaum NY, Olino TM, D’Esposito M, Cogan AB, Patwardan BP, Ellman LM. Decreased hippocampal neurite density in late middle-aged adults following prenatal exposure to higher levels of maternal inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.01.616156. [PMID: 40196686 PMCID: PMC11974695 DOI: 10.1101/2024.10.01.616156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
In animal models, exposure to heightened maternal inflammation in utero is associated with altered offspring hippocampal development, including reduced dendritic arborization and density. However, the effects of prenatal maternal inflammation (PNMI) on offspring hippocampal microstructure in humans remains unclear. Here, we examined the relationship between exposure to PNMI and neurite density in the hippocampus and its subfields among offspring during late middle age. Participants included 72 mother-offspring dyads from the Child Health and Development Studies (CHDS) cohort. Data for four inflammatory biomarkers (IL-6, IL-8, IL-1 receptor antagonist [IL-1RA], and soluble TNF receptor-II [sTNF-RII]) were available from first and second trimester maternal sera. Neurite density in the offspring hippocampus and its subfields was estimated using microstructural modeling of offsprings' diffusion-weighted Magnetic Resonance Imaging data (mean age of offspring at imaging = 59 years; 51% male). We estimated the relationship between each biomarker and region-of-interest's neurite density. Higher first trimester maternal IL-1RA and IL-6 levels were associated with lower offspring hippocampal neurite density. These relationships were specific to the CA3, CA4, dentate gyrus, and subiculum subfields. In addition, higher second trimester IL-6 was associated with lower subiculum neurite density. Our findings reveal that exposure to heightened prenatal levels of maternal inflammation is linked to altered offspring hippocampal microstructure in late middle age, which could have implications for memory decreases during this period and may be relevant for understanding risk of aging-related cognitive changes.
Collapse
Affiliation(s)
| | | | | | - Emma Smith
- Department of Psychology & Neuroscience, Temple University
| | - Ann M. Kring
- Department of Psychology, University of California, Berkeley
| | | | - Elizabeth C. Breen
- Cousins Center for Psychoneuroimmunology, University of California, Los Angeles
| | - Barbara A. Cohn
- Child Health and Development Studies, Public Health Institute
| | | | | | | | - Mark D’Esposito
- Department of Psychology, University of California, Berkeley
| | - Ashby B. Cogan
- Department of Psychology, University of California, Berkeley
| | | | | |
Collapse
|
21
|
Kılınç K, Türkoğlu S, Kocabaş R, Güler HA, Yılmaz Ç, Büyükateş A. What are the levels and interactions of neuroligin-1, neuroligin-3, and inflammatory cytokines (IL-6, IL-8) in children diagnosed with autism spectrum disorder? Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111275. [PMID: 39875012 DOI: 10.1016/j.pnpbp.2025.111275] [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: 10/29/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
Autism spectrum disorder (ASD) is characterized by deficits in social interaction, restricted interests, and repetitive behaviors. Several genes, including synaptic proteins and environmental risk factors, play a role in the etiology of autism. We aimed to evaluate the relationship between neuroligin-1 (NLGN-1) and neuroligin-3 (NLGN-3) levels, which are neuronal cell adhesion molecules (CAMs), and inflammatory cytokine (IL-6, IL-8) levels with disease severity and symptom clusters and with each other in children with ASD. Eighty children diagnosed with autism who met the inclusion criteria and sixty-five typically developing children matched for age and sex were included in the study. The children were evaluated psychiatrically through a semi-structured interview, DSM-5 criteria, the Childhood Autism Rating Scale (CARS), and the Social Communication Questionnaire (SCQ). IL-6, IL-8, NLGN-1, and NLGN-3 levels were analyzed in peripheral serum samples using human ELISA kits. IL-8 and NLGN-3 levels were higher in the autism group (p < 0.001, p < 0.001). IL-6 was positively related to CARS and SCQ total scores (p = 0.021, p = 0.040, respectively). IL-8, and NLGN-3 were positively associated with the all subtests of the SCQ and the SCQ total score (all p values <0.001). NLGN-1, NLGN-3, and inflammatory cytokine (IL-6, IL-8) levels were positively correlated (all p values <0.001). Neuroligins play a central role in the brain's ability to process information and maybe a key target in the pathogenesis of ASD. Further research is needed to determine whether, to what extent and how neuronal CAMs and immunity modulate each other and whether this contributes to ASD pathogenesis. Future studies should also be expanded to investigate the influence of variables such as oxidative stress, metalloproteases responsible for ectodomain shedding, or epigenetic regulation.
Collapse
Affiliation(s)
- Kübra Kılınç
- Department of Child and Adolescent Psychiatry, Konya City Hospital, 42020 Konya, Turkey.
| | - Serhat Türkoğlu
- Department of Child and Adolescent Psychiatry, Selcuk University Faculty of Medicine Hospital, 42130 Konya, Turkey
| | - Ramazan Kocabaş
- Department of Biochemistry, Selcuk University Faculty of Medicine Hospital, 42130 Konya, Turkey
| | - Hasan Ali Güler
- Department of Child and Adolescent Psychiatry, Selcuk University Faculty of Medicine Hospital, 42130 Konya, Turkey
| | - Çiğdem Yılmaz
- Department of Child and Adolescent Psychiatry, Selcuk University Faculty of Medicine Hospital, 42130 Konya, Turkey
| | - Ayşe Büyükateş
- Department of Child and Adolescent Psychiatry, Selcuk University Faculty of Medicine Hospital, 42130 Konya, Turkey
| |
Collapse
|
22
|
Harbi RA, Mouihate A. Maternal immune activation alters the GABAergic system in the prefrontal cortex of female rat offspring: Role of interleukin-6. Neuroscience 2025; 568:399-407. [PMID: 39884421 DOI: 10.1016/j.neuroscience.2025.01.055] [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: 09/05/2024] [Revised: 12/06/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
Maternal immune activation (MIA) induces long-term cognitive impairments by modulating the gamma-aminobutyric acid (GABA)ergic system. Experimental evidence suggests that maternal immune challenge with bacterial active ingredient lipopolysaccharide (LPS) reduces GABAergic tone in the offspring's prefrontal cortex. In this study, we aimed to assess whether interleukin-6 (IL-6) contributes to this reduced GABAergic system in the prefrontal cortex of juvenile offspring. Pregnant rats were given intraperitoneal injections of either LPS (100 µg/Kg) or a pyrogen-free saline solution in the absence or the presence of an IL-6 neutralizing antibody (IL-6Ab, 10 µg/Kg) on gestation day (GD) 15, GD17 and GD19. Parvalbumin and somatostatin GABAergic interneurons and the density of inhibitory synapses were monitored in 30-day-old male and female rat offspring using fluorescent immunohistochemistry. The expression levels of Cl- transporters (NKCC1 and KCC2) were assessed using western blotting. Prenatal LPS induced a significant reduction in the cell density of parvalbumin-containing interneurons in the prefrontal cortex of female but not male rat offspring. LPS-induced MIA led to a reduction in the expression levels of NKCC1 in the prefrontal cortices of both male and female offspring. These long-lasting impacts of the MIA were alleviated when the IL-6Ab was co-administered with LPS during pregnancy. This study shows that the GABAergic system in the prefrontal cortex of female rats is highly sensitive to prenatal immune challenges. These data pave the way for exploring the specific mechanism(s) underlying the sex-dependent effects of early-life immune challenges.
Collapse
Affiliation(s)
- Retaj Al Harbi
- Department of Physiology, College of Medicine, Health Sciences Centre, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait
| | - Abdeslam Mouihate
- Department of Physiology, College of Medicine, Health Sciences Centre, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| |
Collapse
|
23
|
Hikosaka M, Parvez MSA, Yamawaki Y, Oe S, Liang Y, Wada Y, Hirahara Y, Koike T, Imai H, Oishi N, Schalbetter SM, Kumagai A, Yoshida M, Sakurai T, Kitada M, Meyer U, Narumiya S, Ohtsuki G. Maternal immune activation followed by peripubertal stress combinedly produce reactive microglia and confine cerebellar cognition. Commun Biol 2025; 8:296. [PMID: 40033126 PMCID: PMC11876345 DOI: 10.1038/s42003-025-07566-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/15/2025] [Indexed: 03/05/2025] Open
Abstract
The functional alteration of microglia arises in brains exposed to external stress during early development. Pathophysiological findings of neurodevelopmental disorders such as schizophrenia and autism spectrum disorder suggest cerebellar functional deficits. However, the link between stress-induced microglia reactivity and cerebellar dysfunction is missing. Here, we investigate the developmental immune environment in translational mouse models that combine two risk factors: maternal infection and repeated social defeat stress (2HIT). We find the synergy of inflammatory stress insults, leading to microglial increase specifically in the cerebellum of both sexes. Microglial turnover correlates with the Purkinje neuron loss in 2HIT mice. Highly multiplexed imaging-mass-cytometry identifies a cell transition to TREM2(+) stress-associated microglia in the cerebellum. Single-cell-proteomic clustering reveals IL-6- and TGFβ-signaling association with microglial cell transitions. Reduced excitability of remaining Purkinje cells, cerebellum-involved brain-wide functional dysconnectivity, and behavioral abnormalities indicate cerebellar cognitive dysfunctions in 2HIT animals, which are ameliorated by both systemic and cerebellum-specific microglia replacement.
Collapse
Affiliation(s)
- Momoka Hikosaka
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Md Sorwer Alam Parvez
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yuki Yamawaki
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Souichi Oe
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Yuan Liang
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
- Institute of Basic Theory in Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yayoi Wada
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Yukie Hirahara
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Taro Koike
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Hirohiko Imai
- Department of Systems Science, Kyoto University Graduate School of Informatics, Yoshida-Honmachi, Kyoto, Japan
- Innovation Research Center for Quantum Medicine, Gifu University School of Medicine, Gifu, Japan
| | - Naoya Oishi
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sina M Schalbetter
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | | | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Takeshi Sakurai
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Masaaki Kitada
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Urs Meyer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Gen Ohtsuki
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
24
|
Díaz-Pons A, Castaño-Castaño S, Ortiz-García de la Foz V, Yorca-Ruiz Á, Martínez-Asensi C, Munarriz-Cuezva E, Ayesa-Arriola R. Understanding the potential impact of trimester-specific maternal immune activation due to SARS-CoV-2 on early human neurodevelopment and the role of cytokine balance. Brain Behav Immun Health 2025; 44:100956. [PMID: 39990281 PMCID: PMC11846590 DOI: 10.1016/j.bbih.2025.100956] [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: 09/30/2024] [Revised: 12/23/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
Purpose The COVID-19 pandemic presents significant future health challenges. Its impact on pregnant women and their newborn is a particular area of concern. This study aims to examine the potential role of maternal immune activation (MIA), due to SARS-CoV-2 infection, on early neurodevelopment. Methods We analysed 107 mother-infant dyads from the COGESTCOV-19 study in Cantabria, Spain, which included 59 SARS-CoV-2 exposed (cases) and 48 unexposed (controls) mothers, recruited between December 2020 and February 2022. Cytokine levels (IL-6 and IL-10) were obtained from maternal blood and cord blood. Neurodevelopment was assessed using the Neonatal Behavioral Assessment Scale (NBAS) at six weeks of age. Trimester of infection was considered in the main analyses. Results Results showed no significant overall delays in early neurodevelopment due to maternal SARS-CoV-2 infection. Control infants performed better in some NBAS items. However, cases infants showed trimester-specific differences. First-trimester exposure was related to motor and reflex delays, second-trimester to poorer performances in motor tasks and autonomic stability, and third-trimester to weaker state organization, regulation, and reflexes. Some correlations between cytokine levels and NBAS performance showed moderate associations. Conclusions These findings highlight the need for ongoing neurodevelopmental monitoring of infants born during the COVID-19 pandemic. The study enhances our understanding of MIA's impact on early development, emphasizing the importance of addressing homeostatic mechanisms in mothers and newborns.
Collapse
Affiliation(s)
- Alexandre Díaz-Pons
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
- Escuela de Doctorado de la Universidad de Cantabria (EDUC), Universidad de Cantabria (UC), 39005, Santander, Spain
- Departamento de Medicina y Ciencias de la Salud, Facultad de Medicina, Universidad de Cantabria (UC), 39011, Santander, Spain
- Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28015, Madrid, Spain
| | - Sergio Castaño-Castaño
- Departamento de Psicobiología, Facultad de Psicología, Universidad de Oviedo (UO), 33003, Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011, Oviedo, Spain
| | - Víctor Ortiz-García de la Foz
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Ángel Yorca-Ruiz
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
- Escuela de Doctorado de la Universidad de Cantabria (EDUC), Universidad de Cantabria (UC), 39005, Santander, Spain
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria (UC), 39011, Santander, Spain
| | - Carlos Martínez-Asensi
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
- Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28015, Madrid, Spain
| | - Eva Munarriz-Cuezva
- Departamento de Farmacología, Facultad de Medicina y Enfermería, Universidad del País Vasco/ Euskal Herriko Unibertsitatea (UPV/EHU), 48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Rosa Ayesa-Arriola
- Departamento de Investigación en Enfermedades Mentales, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
- Departamento de Medicina y Ciencias de la Salud, Facultad de Medicina, Universidad de Cantabria (UC), 39011, Santander, Spain
- Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28015, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
25
|
Chang G, Sebastian P, Virupakshaiah A, Schoeps VA, Cherbuin N, Casper TC, Gorman MP, Benson LA, Chitnis T, Rensel M, Abrams AW, Lotze T, Mar SS, Schreiner TL, Wheeler YS, Rose JW, Graves J, Krupp LB, Waldman AT, Lucas R, Waubant E. Association Between Sun Exposure and Risk of Relapse in Pediatric-Onset Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200375. [PMID: 39938013 PMCID: PMC11820808 DOI: 10.1212/nxi.0000000000200375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/28/2024] [Indexed: 02/14/2025]
Abstract
BACKGROUND AND OBJECTIVES Low sun and ultraviolet radiation (UVR) exposures have been associated with increased risk of developing pediatric-onset multiple sclerosis (MS); however, their effect on disease course has not been well characterized. We primarily investigated whether there was an association between time spent in the sun in early childhood and risk of relapse in pediatric MS. We secondarily investigated the effect of sun exposure during more recent periods on risk of relapse. METHODS We conducted a multicenter cohort study of participants with pediatric-onset MS recruited from 18 pediatric MS clinics across the United States between November 1, 2011, and July 1, 2017. Relapses were identified prospectively after study enrollment; relapses preceding study enrollment were entered retrospectively. Time spent in the sun at various periods of life was measured using a detailed environmental questionnaire, and ambient UVR exposure was determined using zip codes. Multivariable Cox regression models were used to assess the association between time spent in the sun and UVR dose at specific periods of life and the risk of relapse. Models were adjusted for demographic, clinical, and sun exposure-related characteristics. RESULTS In our cohort of 334 children with MS, 206 (62%) experienced at least one relapse from disease onset to the end of the follow-up period. After adjustment, ≥30 minutes of daily sun exposure during the first summer of life was associated with a lower risk of relapse compared with <30 minutes (adjusted hazard ratio [aHR] 0.67, CI 0.48-0.92, p = 0.01). Greater time spent in the sun during the second trimester of pregnancy was also associated with reduced risk of relapse (aHR 0.68, CI 0.48-0.97, p = 0.04). UVR dose and time spent in the sun later in life were not significantly associated with relapse risk. DISCUSSION In this large cohort study of children with MS, greater early childhood and prenatal sun exposure time was associated with lower risk of relapse. Further investigation of sun exposure at other periods is needed to better characterize its impact on disease course and guide potential future interventions.
Collapse
Affiliation(s)
- Gina Chang
- Division of Child Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Akash Virupakshaiah
- UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Vinicius A Schoeps
- UCSF Weill Institute for Neurosciences, University of California San Francisco
| | - Nicolas Cherbuin
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | | | | | | | - Tanuja Chitnis
- Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | | | | | - Tim Lotze
- Texas Children's Hospital, Houston, TX
| | - Soe S Mar
- Washington University in St. Louis, MO
| | - Teri L Schreiner
- Children's Hospital Colorado, Aurora
- University of Colorado, Aurora
| | | | - John W Rose
- George E. Wahlen Department of Veterans Affairs Medical Center, University of Utah, Salt Lake City
| | | | | | - Amy T Waldman
- Division of Child Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Robyn Lucas
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, Australia
| | - Emmanuelle Waubant
- UCSF Weill Institute for Neurosciences, University of California San Francisco
| |
Collapse
|
26
|
Nakai T, Fukui S, Ozawa H, Kitada A, Okada M, Kishimoto M. Management of pregnant with rheumatoid arthritis: Preconception care, pregnancy and lactation strategies, and maternal-fetal outcomes. Best Pract Res Clin Rheumatol 2025; 39:102022. [PMID: 39572276 DOI: 10.1016/j.berh.2024.102022] [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: 10/29/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 03/17/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disorder that can affect women of reproductive age. In recent decades, significant advances have been made in the development of new medications, including biologic disease-modifying anti-rheumatic drugs (DMARDs) and Janus kinase (JAK) inhibitors. Women with RA are prone to infertility, with 42% experiencing a time to pregnancy exceeding 12 months. High disease activity, as well as the use of high-dose glucocorticoids and Non-Steroidal Anti-Inflammatory Drugs (NSAIDs), are associated with infertility and adverse pregnancy outcomes. Additionally, some medications, such as methotrexate, are linked to teratogenicity, highlighting the importance of providing preconception care in everyday practice. Recent advancements in reproductive care have improved our ability to manage RA during pregnancy, leading to better pregnancy outcomes. In this review, we summarize key aspects of fertility care, pregnancy and lactation management, including medication strategies, neonatal vaccination, and long-term outcomes for offspring born to mothers with RA.
Collapse
Affiliation(s)
- Takehiro Nakai
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan.
| | - Sho Fukui
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Emergency and General Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Hiroki Ozawa
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Ayako Kitada
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan; Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masato Okada
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Mitsumasa Kishimoto
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan; Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Orchanian SB, Hsiao EY. The microbiome as a modulator of neurological health across the maternal-offspring interface. J Clin Invest 2025; 135:e184314. [PMID: 39959974 PMCID: PMC11827852 DOI: 10.1172/jci184314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
The maternal microbiome is emerging as an important factor that influences the neurological health of mothers and their children. Recent studies highlight how microbial communities in the maternal gut can shape early-life development in ways that inform long-term health trajectories. Research on the neurodevelopmental effects of maternal microbiomes is expanding our understanding of the microbiome-gut-brain axis to include signaling across the maternal-offspring unit during the perinatal period. In this Review, we synthesize existing literature on how the maternal microbiome modulates brain function and behavior in both mothers and their developing offspring. We present evidence from human and animal studies showing that the maternal microbiome interacts with environmental factors to impact risk for neurodevelopmental abnormalities. We further discuss molecular and cellular mechanisms that facilitate maternal-offspring crosstalk for neuromodulation. Finally, we consider how advancing understanding of these complex interactions could lead to microbiome-based interventions for promoting maternal and offspring health.
Collapse
Affiliation(s)
| | - Elaine Y. Hsiao
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California, USA
- UCLA Goodman-Luskin Microbiome Center, Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California, USA
| |
Collapse
|
28
|
Ignatiuk V, Sharova V, Zakharova L. Prenatal Inflammation Reprograms Hypothalamic-Pituitary-Gonadal Axis Development in Female Rats. Inflammation 2025:10.1007/s10753-025-02243-2. [PMID: 39909991 DOI: 10.1007/s10753-025-02243-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025]
Abstract
The hypothalamic-pituitary-gonadal (HPG) axis development during critical periods of ontogenesis can be disrupted by stress factors, including in particular maternal immune activation by infectious agents. Bacterial lipopolysaccharide (LPS, E.coli) exposure induces inflammation accompanied by proinflammatory cytokine release. The resulting elevated cytokine levels may lead to a disruption of epigenetic mechanisms regulating HPG axis development and to a reduced fertility in the offspring. This study focused on the long-term effects of prenatal LPS exposure on HPG axis development in female rats and the modulation of such effects by anti-inflammatory drugs: polyclonal IgG and monoclonal anti-IL6-receptor antibodies. LPS exposure on embryonic day 12 led to a decrease in the number of synaptic inputs on gonadotropin-releasing-hormone-producing neurons in the hypothalamus, high levels of follicular atresia, and suppressed steroidogenesis in the ovaries of adult female offspring. IgG treatment or IL6 receptor blockade by monoclonal antibodies 40 minutes after LPS exposure prevented these long-term negative effects of LPS. The data obtained suggest that IL6 is involved in the regulation of HPG axis development.
Collapse
Affiliation(s)
- Vasilina Ignatiuk
- Koltsov Institute of Developmental Biology, The Russian Academy of Sciences, Vavilov Street, 26, 119334, Moscow, Russia
| | - Viktoriya Sharova
- Koltsov Institute of Developmental Biology, The Russian Academy of Sciences, Vavilov Street, 26, 119334, Moscow, Russia.
| | - Liudmila Zakharova
- Koltsov Institute of Developmental Biology, The Russian Academy of Sciences, Vavilov Street, 26, 119334, Moscow, Russia
| |
Collapse
|
29
|
Martín-Guerrero SM, Martín-Estebané M, Lara Ordóñez AJ, Cánovas M, Martín-Oliva D, González-Maeso J, Cutillas PR, López-Giménez JF. Maternal immune activation imprints translational dysregulation and differential MAP2 phosphorylation in descendant neural stem cells. Mol Psychiatry 2025:10.1038/s41380-025-02905-5. [PMID: 39900676 DOI: 10.1038/s41380-025-02905-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/19/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025]
Abstract
Alterations induced by maternal immune activation (MIA) during gestation impact the subsequent neurodevelopment of progeny, a process that in humans, has been linked to the development of several neuropsychiatric conditions. To undertake a comprehensive examination of the molecular mechanisms governing MIA, we have devised an in vitro model based on neural stem cells (NSCs) sourced from fetuses carried by animals subjected to Poly I:C treatment. These neural progenitors demonstrate proliferative capacity and can be effectively differentiated into both neurons and glial cells. Transcriptomic, proteomic, and phosphoproteomic analyses conducted on these cellular models, in conjunction with counterparts from control treatments, revealed discernible shifts in the expression levels of a specific subset of proteins implicated in neuronal function. Furthermore, the phosphoproteomic data highlighted a discernible discrepancy in the basal phosphorylation of proteins between differentiated cells from both experimental groups, particularly within proteins associated with cytoskeletal architecture and synaptic functionality, notably those belonging to the MAP family. Observed alterations in MAP phosphorylation were found to potentially have functional consequences as they correlate with changes in neuronal plasticity and the establishment of neuronal synapses. Our data agrees with previous published observations and further underscore the importance of MAP2 phosphorylation state on its function and the impact that this protein has in neuronal structure and function.
Collapse
Affiliation(s)
- Sandra M Martín-Guerrero
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | | | | | - Miguel Cánovas
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), Granada, Spain
| | - David Martín-Oliva
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Pedro R Cutillas
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK.
| | - Juan F López-Giménez
- Instituto de Parasitología y Biomedicina "López-Neyra" (IPBLN-CSIC), Granada, Spain.
| |
Collapse
|
30
|
Clark DN, Brown SV, Xu L, Lee RL, Ragusa JV, Xu Z, Milner JD, Filiano AJ. Prolonged STAT1 signaling in neurons causes hyperactive behavior. Brain Behav Immun 2025; 124:1-8. [PMID: 39542073 PMCID: PMC11745914 DOI: 10.1016/j.bbi.2024.11.018] [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: 07/01/2024] [Revised: 10/29/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
The interferon (IFN)-induced STAT1 signaling pathway is a canonical immune pathway that has also been implicated in regulating neuronal activity. The pathway is enriched in brains of individuals with autism spectrum disorder (ASD) and schizophrenia (SZ). Over-activation of the STAT1 pathway causes pathological transcriptional responses, however it is unclear how these responses might translate into behavioral phenotypes. We hypothesized that prolonged STAT1 signaling in neurons would be sufficient to cause behavioral deficits associated with neurodevelopmental disorders. In this study, we developed a novel mouse model with the clinical STAT1 gain-of-function mutation, T385M, in neurons. These mice were hyperactive and displayed neural hypoactivity with less neuron counts in the caudate putamen. Driving the STAT1 gain-of-function mutation exclusively in dopaminergic neurons, which project to the caudate putamen of the dorsal striatum, mimicked some hyperactive behaviors without a reduction of neurons. Moreover, we demonstrated that this phenotype is neuron specific, as mice with prolonged STAT1 signaling in all excitatory or inhibitory neurons or in microglia were not hyperactive. Overall, these findings suggest that STAT1 signaling in neurons is a crucial player in regulating striatal neuron activity and aspects of motor behavior.
Collapse
Affiliation(s)
- Danielle N Clark
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA; Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Shelby V Brown
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Li Xu
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Rae-Ling Lee
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Joey V Ragusa
- Department of Pathology, Duke University, Durham, NC, USA
| | - Zhenghao Xu
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Joshua D Milner
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Anthony J Filiano
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA; Marcus Center for Cellular Cures, Duke University, Durham, NC, USA; Department of Pathology, Duke University, Durham, NC, USA; Department of Neurosurgery, Duke University, Durham, NC, USA.
| |
Collapse
|
31
|
Mukhtar I. Unravelling the critical role of neuroinflammation in epilepsy-associated neuropsychiatric comorbidities: A review. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111135. [PMID: 39237022 DOI: 10.1016/j.pnpbp.2024.111135] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 09/01/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Epilepsy is a complex neurological disorder characterized not only by seizures but also by significant neuropsychiatric comorbidities, affecting approximately one-third of those diagnosed. This review explores the intricate relationship between epilepsy and its associated psychiatric and cognitive disturbances, with a focus on the role of inflammation. Recent definitions of epilepsy emphasize its multifaceted nature, linking it to neurobiological, psychiatric, cognitive, and social deficits. Inflammation has emerged as a critical factor influencing both seizure activity and neuropsychiatric outcomes in epilepsy patients. This paper critically examines how dysregulated inflammatory pathways disrupt neurotransmitter transmission and contribute to depression, mood disorders, and anxiety prevalent among individuals with epilepsy. It also evaluates current therapeutic approaches and underscores the potential of anti-inflammatory therapies in managing epilepsy and related neuropsychiatric conditions. Additionally, the review highlights the importance of the anti-inflammatory effects of anti-seizure medications, antidepressants, and antipsychotics and their therapeutic implications for mood disorders. Also, the role of ketogenic diet in managing epilepsy and its psychiatric comorbidities is briefly presented. Furthermore, it briefly discusses the role of the gut-brain axis in maintaining neurological health and how its dysregulation is associated with epilepsy. The review concludes that inflammation plays a pivotal role in linking epilepsy with its neuropsychiatric comorbidities, suggesting that targeted anti-inflammatory interventions may offer promising therapeutic strategies. Future research should focus on longitudinal studies comparing outcomes between epileptic patients with and without neuropsychiatric comorbidities, the development of diagnostic tools, and the exploration of novel anti-inflammatory treatments to better manage these complex interactions.
Collapse
Affiliation(s)
- Iqra Mukhtar
- Faculty of Pharmacy, Iqra University, Karachi, Pakistan.
| |
Collapse
|
32
|
Cui J, Li H, Hu C, Zhang F, Li Y, Weng Y, Yang L, Li Y, Yao M, Li H, Luo X, Hao Y. Unraveling pathogenesis and potential biomarkers for autism spectrum disorder associated with HIF1A pathway based on machine learning and experiment validation. Neurobiol Dis 2025; 204:106763. [PMID: 39657846 DOI: 10.1016/j.nbd.2024.106763] [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: 08/04/2024] [Revised: 10/05/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a high social burden and limited treatments. Hypoxic condition of the brain is considered an important pathological mechanism of ASD. HIF1A is a key participant in brain hypoxia, but its contribution to the pathophysiological landscape of ASD remains unclear. METHODS ASD-related datasets were obtained from GEO database, and HIF1A-related genes from GeneCards. Co-expression module analysis identified module genes, which were intersected with HIF1A-related genes to identify common genes. Machine learning identified hub genes from intersection genes and PPI networks were constructed to explore relationships among hub and HIF1A. Single-cell RNA sequencing analyzed hub gene distribution across cell clusters. ASD mouse model was created by inducing maternal immune activation (MIA) with poly(I:C) injections, verified through behavioral tests. Validation of HIF1A pathway and hub genes was confirmed through Western Blot, qPCR, and immunofluorescence in ASD mice and microglia BV-2 cells. RESULTS Using CEMiTool and GeneCards, 45 genes associated with ASD and HIF1A pathway were identified. Machine learning identified CDKN1A, ETS2, LYN, and SLC16A3 as potential ASD diagnostic markers. Single-cell sequencing pinpointed activated microglia as key immune cells. Behavioral tests showed MIA offspring mice exhibited typical ASD-like behaviors. Immunofluorescence confirmed the activation of microglia and HIF1A pathway in frontal cortex of ASD mice. Additionally, IL-6 contributed to ASD by activating JUN/HIF1A pathway, affecting CDKN1A, LYN, and SLC16A3 expression in microglia. CONCLUSIONS HIF1A-related genes CDKN1A, ETS2, LYN, and SLC16A3 are strong diagnostic markers for ASD and the activation of IL-6/JUN/HIF1A pathway in microglia contributes to the pathogenesis of ASD.
Collapse
Affiliation(s)
- Jinru Cui
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Heli Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cong Hu
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feiyan Zhang
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yunjie Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Weng
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liping Yang
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yingying Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Minglan Yao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Li
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Hao
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
33
|
Otero AM, Connolly MG, Gonzalez-Ricon RJ, Wang SS, Allen JM, Antonson AM. Influenza A virus during pregnancy disrupts maternal intestinal immunity and fetal cortical development in a dose- and time-dependent manner. Mol Psychiatry 2025; 30:13-28. [PMID: 38961232 PMCID: PMC11649561 DOI: 10.1038/s41380-024-02648-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
Epidemiological studies link exposure to viral infection during pregnancy, including influenza A virus (IAV) infection, with increased incidence of neurodevelopmental disorders (NDDs) in offspring. Models of maternal immune activation (MIA) using viral mimetics demonstrate that activation of maternal intestinal T helper 17 (TH17) cells, which produce effector cytokine interleukin (IL)-17, leads to aberrant fetal brain development, such as neocortical malformations. Fetal microglia and border-associated macrophages (BAMs) also serve as potential cellular mediators of MIA-induced cortical abnormalities. However, neither the inflammation-induced TH17 cell pathway nor fetal brain-resident macrophages have been thoroughly examined in models of live viral infection during pregnancy. Here, we inoculated pregnant mice with two infectious doses of IAV and evaluated peak innate and adaptive immune responses in the dam and fetus. While respiratory IAV infection led to dose-dependent maternal colonic shortening and microbial dysregulation, there was no elevation in intestinal TH17 cells nor IL-17. Systemically, IAV resulted in consistent dose- and time-dependent increases in IL-6 and IFN-γ. Fetal cortical abnormalities and global changes in fetal brain transcripts were observable in the high-but not the moderate-dose IAV group. Profiling of fetal microglia and BAMs revealed dose- and time-dependent differences in the numbers of meningeal but not choroid plexus BAMs, while microglial numbers and proliferative capacity of Iba1+ cells remained constant. Fetal brain-resident macrophages increased phagocytic CD68 expression, also in a dose- and time-dependent fashion. Taken together, our findings indicate that certain features of MIA are conserved between mimetic and live virus models, while others are not. Overall, we provide consistent evidence of an infection severity threshold for downstream maternal inflammation and fetal cortical abnormalities, which recapitulates a key feature of the epidemiological data and further underscores the importance of using live pathogens in NDD modeling to better evaluate the complete immune response and to improve translation to the clinic.
Collapse
Affiliation(s)
- Ashley M Otero
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Meghan G Connolly
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | - Selena S Wang
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jacob M Allen
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Adrienne M Antonson
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
34
|
Mattei D, Guneykaya D, Ugursu B, Buonfiglioli A. From womb to world: The interplay between maternal immune activation, neuroglia, and neurodevelopment. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:269-285. [PMID: 40148048 DOI: 10.1016/b978-0-443-19102-2.00028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
This chapter introduces and discusses maternal immune activation (MIA) as a contributing factor in increasing the risk of neurodevelopmental disorders, particularly in relation to its interactions with neuroglia. Here we first provide an overview of the neuroglia-astroglia, oligodendroglia, microglia, and radial glial cells-and their important role during early brain development and in adulthood. We then present and discuss MIA, followed by a critical overview of inflammatory molecules and temporal stages associated to maternal inflammation during pregnancy. We provide an overview of animal and human models used to mimic and study MIA. Furthermore, we review the possible interaction between MIA and neuroglia, focusing on the current advances in both modeling and therapeutics. Additionally, we discuss and provide preliminary and interesting insights into the most recent pandemic, COVID-19, and how the infection may be associated to MIA and increased risk for neurodevelopmental disorders. Finally, we provide a critical overview of challenges and future opportunities to study how MIA may contribute to higher risk of developing neurodevelopmental disorders.
Collapse
Affiliation(s)
- Daniele Mattei
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, New York, NY, United States
| | - Dilansu Guneykaya
- Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Bilge Ugursu
- Department of Psychoneuroimmunology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Alice Buonfiglioli
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
35
|
Bose R, Posada-Pérez M, Karvela E, Skandik M, Keane L, Falk A, Spulber S, Joseph B, Ceccatelli S. Bi-allelic NRXN1α deletion in microglia derived from iPSC of an autistic patient increases interleukin-6 production and impairs supporting function on neuronal networking. Brain Behav Immun 2025; 123:28-42. [PMID: 39243986 DOI: 10.1016/j.bbi.2024.09.001] [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: 01/19/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Autism spectrum disorder (ASD) is a set of heterogeneous neurodevelopmental conditions, with a highly diverse genetic hereditary component, including altered neuronal circuits, that has an impact on communication skills and behaviours of the affected individuals. Beside the recognised role of neuronal alterations, perturbations of microglia and the associated neuroinflammatory processes have emerged as credible contributors to aetiology and physiopathology of ASD. Mutations in NRXN1, a member of the neurexin family of cell-surface receptors that bind neuroligin, have been associated to ASD. NRXN1 is known to be expressed by neurons where it facilitates synaptic contacts, but it has also been identified in glial cells including microglia. Asserting the impact of ASD-related genes on neuronal versus microglia functions has been challenging. Here, we present an ASD subject-derived induced pluripotent stem cells (iPSC)-based in vitro system to characterise the effects of the ASD-associated NRXN1 gene deletion on neurons and microglia, as well as on the ability of microglia to support neuronal circuit formation and function. Using this approach, we demonstrated that NRXN1 deletion, impacting on the expression of the alpha isoform (NRXN1α), in microglia leads to microglial alterations and release of IL6, a pro-inflammatory interleukin associated with ASD. Moreover, microglia bearing the NRXN1α-deletion, lost the ability to support the formation of functional neuronal networks. The use of recombinant IL6 protein on control microglia-neuron co-cultures or neutralizing antibody to IL6 on their NRXN1α-deficient counterparts, supported a direct contribution of IL6 to the observed neuronal phenotype. Altogether, our data suggest that, in addition to neurons, microglia are also negatively affected by NRXN1α-deletion, and this significantly contributes to the observed neuronal circuit aberrations.
Collapse
Affiliation(s)
- Raj Bose
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong
| | - Mercedes Posada-Pérez
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong
| | - Eleni Karvela
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Martin Skandik
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Lily Keane
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong; Lund Stem Cell Center, Lund University, 22100 Lund, Sweden
| | - Stefan Spulber
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong
| | - Bertrand Joseph
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong.
| |
Collapse
|
36
|
Wang X, Li Q, Lyu Z, Wu Y. Supplementing with Vitamin D during Pregnancy Reduces Inflammation and Prevents Autism-Related Behaviors in Offspring Caused by Maternal Immune Activation. Biol Pharm Bull 2025; 48:632-640. [PMID: 40383635 DOI: 10.1248/bpb.b25-00008] [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] [Indexed: 05/20/2025]
Abstract
Autism spectrum disorder (ASD), a neurodevelopmental disorder of unknown etiology with limited treatment options, has emerged as a significant public health concern. Studies have demonstrated that prenatal vitamin D deficiency is a risk factor for ASD development in offspring; however, the underlying mechanism remains unclear. In this project, vitamin D was administered orally to pregnant mice with/without the subsequent administration of polyriboinosinic polyribocytidylic acid (Poly(I:C)), which induced the maternal immune activation (MIA). Our results showed that vitamin D supplementation during pregnancy alleviated MIA-induced ASD-like behaviors in offspring. Moreover, vitamin D supplementation reduced the MIA-induced elevation of interleukin-6 (IL-6) and IL-17a levels in both the maternal ileum and fetal brains. It also suppressed signal transducer and activator of transcription 3 (Stat3) activation and the elevated expression of serum amyloid A1 and A2 (SAA1/2) in the ileum of MIA-affected pregnant mice. This study revealed that vitamin D may reduce the expression of IL-17a by inhibiting the IL-6/Stat3/SAA signaling pathway, thereby improving ASD-like behavior in offspring mice, and provide a new theoretical support for the prevention and treatment of ASD by scientific dietary interventions and nutritional supplement during pregnancy.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Qingqing Li
- Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Zhihong Lyu
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yingying Wu
- Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| |
Collapse
|
37
|
Salia S, Burke FF, Hinks ME, Randell AM, Matheson MA, Walling SG, Swift-Gallant A. Gut microbiota transfer from the preclinical maternal immune activation model of autism is sufficient to induce sex-specific alterations in immune response and behavioural outcomes. Brain Behav Immun 2025; 123:813-823. [PMID: 39471905 DOI: 10.1016/j.bbi.2024.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/22/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024] Open
Abstract
The gut microbiome plays a vital role in health and disease, including neurodevelopmental disorders like autism spectrum disorder (ASD). ASD affects 4:1 males-to-females, and sex differences are apparent in gut microbiota composition among ASD individuals and in animal models of this condition, such as the maternal immune activation (MIA) mouse model. However, few studies have included sex as a biological variable when assessing the role of gut microbiota in mediating ASD symptoms. Using the MIA model of ASD, we assessed whether gut microbiota contributes to the sex differences in the presentation of ASD-like behaviors. Gut microbiota transplantation from MIA or vehicle/control male and female mice into healthy, otherwise unmanipulated, 4-week-old C57Bl/6 mice was performed for 6 treatments over 12 days. Colonization with male, but not female, MIA microbiota was sufficient to reduce sociability, decrease microbiota diversity and increase neuroinflammation with more pronounced deficits in male recipients. Colonization with both male and female donor microbiota altered juvenile ultrasonic vocalizations and anxiety-like behavior in recipients of both sexes, and there was an accompanied change in the gut microbiota and serum cytokine IL-4 and IL-7 levels of all recipients of MIA gut microbiota. In addition to the increases in gut microbes associated with pathological states, the female donor microbiota profile also had increases in gut microbes with known neural protective effects (e.g., Lactobacillus and Rikenella). These results suggest that gut reactivity to environmental insults, such as in the MIA model, may play a role in shaping the sex disparity in ASD development.
Collapse
Affiliation(s)
- Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada.
| | - Francine F Burke
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Meagan E Hinks
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Alison M Randell
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Mairead Anna Matheson
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Susan G Walling
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
38
|
Munarriz‐Cuezva E, Meana JJ. Poly (I:C)-induced maternal immune activation generates impairment of reversal learning performance in offspring. J Neurochem 2025; 169:e16212. [PMID: 39183542 PMCID: PMC11657921 DOI: 10.1111/jnc.16212] [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: 01/29/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024]
Abstract
Maternal immune activation (MIA) induces a variety of behavioral and brain abnormalities in offspring of rodent models, compatible with neurodevelopmental disorders, such as schizophrenia or autism. However, it remains controversial whether MIA impairs reversal learning, a basic expression of cognitive flexibility that seems to be altered in schizophrenia. In the present study, MIA was induced by administration of a single dose of polyriboinosinic-polyribocytidylic acid (Poly (I:C) (5 mg/kg i.p.)) or saline to mouse pregnant dams in gestational day (GD) 9.5. Immune activation was monitored through changes in weight and temperature. The offspring were evaluated when they reached adulthood (8 weeks) using a touchscreen-based system to investigate the effects of Poly (I:C) on discrimination and reversal learning performance. After an initial pre-training, mice were trained to discriminate between two different stimuli, of which only one was rewarded (acquisition phase). When the correct response reached above 80% values for two consecutive days, the images were reversed (reversal phase) to assess the adaptation capacity to a changing environment. Maternal Poly (I:C) treatment did not interfere with the learning process but induced deficits in reversal learning compared to control saline animals. Thus, the accuracy in the reversal phase was lower, and Poly (I:C) animals required more sessions to complete it, suggesting impairments in cognitive flexibility. This study advances the knowledge of how MIA affects behavior, especially cognitive domains that are impaired in schizophrenia. The findings support the validity of the Poly (I:C)-based MIA model as a tool to develop pharmacological treatments targeting cognitive deficits associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Eva Munarriz‐Cuezva
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque CountryLeioaBizkaiaSpain
- Centro de Investigación Biomédica en Red de Salud MentalLeioaBizkaiaSpain
| | - Jose Javier Meana
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque CountryLeioaBizkaiaSpain
- Centro de Investigación Biomédica en Red de Salud MentalLeioaBizkaiaSpain
- Biobizkaia Health Research InstituteBarakaldoBizkaiaSpain
| |
Collapse
|
39
|
Mamun AA, Geng P, Wang S, Shao C, Xiao J. IUPHAR review: Targeted therapies of signaling pathways based on the gut microbiome in autism spectrum disorders: Mechanistic and therapeutic applications. Pharmacol Res 2025; 211:107559. [PMID: 39733842 DOI: 10.1016/j.phrs.2024.107559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders characterized by impairments in social interaction, communication and repetitive activities. Gut microbiota significantly influences behavior and neurodevelopment by regulating the gut-brain axis. This review explores gut microbiota-influenced treatments for ASD, focusing on their therapeutic applications and mechanistic insights. In addition, this review discusses the interactions between gut microbiota and the immune, metabolic and neuroendocrine systems, focusing on crucial microbial metabolites including short-chain fatty acids (SCFAs) and several neurotransmitters. Furthermore, the review explores various therapy methods including fecal microbiota transplantation, dietary modifications, probiotics and prebiotics and evaluates their safety and efficacy in reducing ASD symptoms. The discussion shows the potential of customized microbiome-based therapeutics and the integration of multi-omics methods to understand the underlying mechanisms. Moreover, the review explores the intricate relationship between gut microbiota and ASD, aiming to develop innovative therapies that utilize the gut microbiome to improve the clinical outcomes of ASD patients. Microbial metabolites such as neurotransmitter precursors, tryptophan metabolites and SCFAs affect brain development and behavior. Symptoms of ASD are linked to changes in these metabolites. Dysbiosis in the gut microbiome may impact neuroinflammatory processes linked to autism, negatively affecting immune signaling pathways. Research indicates that probiotics and prebiotics can improve gut microbiota and alleviate symptoms in ASD patients. Fecal microbiota transplantation may also improve behavioral symptoms and restore gut microbiota balance. The review emphasizes the need for further research on gut microbiota modification as a potential therapeutic approach for ASD, highlighting its potential in clinical settings.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China.
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
40
|
Niskanen A, Barron A, Azaryah H, Kerkelä M, Pulli E, Tuulari JJ, Lukkarinen M, Karlsson L, Muetzel RL, Campoy C, Catena A, Tiemeier H, Khandaker GM, Karlsson H, Veijola J, Björnholm L. Sex-specific associations between maternal prenatal inflammation and offspring cortical morphology in youth: A harmonised study across four birth cohorts. Brain Behav Immun 2025; 123:1081-1090. [PMID: 39505051 DOI: 10.1016/j.bbi.2024.11.010] [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: 04/23/2024] [Revised: 09/29/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024] Open
Abstract
Maternal immune activation (MIA) during pregnancy is implicated in offspring psychiatric disorders. However, it is unknown to what extent MIA affects neurodevelopment, particularly cerebrocortical anatomy, in the general population, and whether effects differ by sex. The current study used vertex-wise statistics to examine the association between maternal prenatal CRP, an archetypal systemic inflammatory marker, and offspring cortical thickness, surface area, and volume, in 2635 mother-child dyads (5.4-26.5 years) from three population-based cohorts, and one clinical cohort enriched for presence of inflammation markers. Maternal CRP within a normal physiological range (<10 mg/L) exhibited sex-specific quadratic associations with cortical morphological measures in 2 regions in males and 1 region in females at childhood. Elevated (>10 mg/L) CRP was associated with regional cortical morphology in females and in a pooled sample of sexes. Overall, MIA is associated with cortical development in a regional and sex-specific manner in studies spanning childhood to adulthood.
Collapse
Affiliation(s)
- Anni Niskanen
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Aaron Barron
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Clinical Medicine, Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Hatim Azaryah
- Department of Pediatrics, University of Granada, Granada, Spain; EURISTIKOS Excellence Centre for Pediatric Research, School of Medicine, University of Granada, Granada, Spain; Instituto Biosanitario de Granada (Ibs-Granada), Granada, Spain
| | - Martta Kerkelä
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland
| | - Elmo Pulli
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland
| | - Jetro J Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Clinical Medicine, Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland; Turku Collegium for Science, Medicine and Technology (TCSMT), University of Turku, Turku, Finland
| | - Minna Lukkarinen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland; The Department of Pediatrics and Adolescent Medicine, University of Turku, and Turku University Hospital, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland; Department of Clinical Medicine, Unit of Public Health, University of Turku, Finland; Department of Child Psychiatry, Turku University Hospital, Finland
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, Netherlands (the); Department of Radiology and Nuclear Medicine, Erasmus University Medical Center,Netherlands (the)
| | - Cristina Campoy
- Department of Pediatrics, University of Granada, Granada, Spain; EURISTIKOS Excellence Centre for Pediatric Research, School of Medicine, University of Granada, Granada, Spain; Instituto Biosanitario de Granada (Ibs-Granada), Granada, Spain; CIBERESP, Spanish Research Network on Epidemiology and Public Health, ISCIII, Madrid, Spain
| | - Andrés Catena
- Department of Experimental Psychology, School of Psychology, University of Granada, Granada, Spain; Mind, Brain & Behaviour Centre (CIMCYC), University of Granada, Spain
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, Netherlands (the); Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Golam M Khandaker
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Centre for Academic Mental Health, Bristol Medical School, University of Bristol, Bristol, UK; National Institute of Health and Care Research Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK; Avon and Wiltshire Mental Health Partnership NHS Trust, Bristol, UK
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Clinical Medicine, Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland
| | - Juha Veijola
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Lassi Björnholm
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| |
Collapse
|
41
|
Guma E, Chakravarty MM. Immune Alterations in the Intrauterine Environment Shape Offspring Brain Development in a Sex-Specific Manner. Biol Psychiatry 2025; 97:12-27. [PMID: 38679357 PMCID: PMC11511788 DOI: 10.1016/j.biopsych.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Exposure to immune dysregulation in utero or in early life has been shown to increase risk for neuropsychiatric illness. The sources of inflammation can be varied, including acute exposures due to maternal infection or acute stress, or persistent exposures due to chronic stress, obesity, malnutrition, or autoimmune diseases. These exposures may cause subtle alteration in brain development, structure, and function that can become progressively magnified across the lifespan, potentially increasing the likelihood of developing a neuropsychiatric conditions. There is some evidence that males are more susceptible to early-life inflammatory challenges than females. In this review, we discuss the various sources of in utero or early-life immune alteration and the known effects on fetal development with a sex-specific lens. To do so, we leveraged neuroimaging, behavioral, cellular, and neurochemical findings. Gaining clarity about how the intrauterine environment affects offspring development is critically important for informing preventive and early intervention measures that may buffer against the effects of these early-life risk factors.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, Maryland; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Heine VM, Dooves S. Neuroglia in autism spectrum disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:303-311. [PMID: 40148051 DOI: 10.1016/b978-0-443-19102-2.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Autism spectrum disorder (ASD) is characterized by difficulties in social interaction, communication, and repetitive behavior, typically diagnosed during early childhood and attributed to altered neuronal network connectivity. Several genetic and environmental risk factors contribute to ASD, including pre- or early life immune activation, which can trigger microglial and astroglial reactivity, impacting early neurodevelopment. In ASD, astrocytes show altered glutamate metabolism, directly influencing neuronal network activity, while microglia display impaired synaptic pruning, an essential developmental process for the refinement of neuronal connections. Additionally, reduced myelination in specific cortical and subcortical regions may affect brain connectivity in ASD, with white matter integrity correlating with the severity of the disorder, suggesting an important role for oligodendrocytes and myelin in ASD. This chapter provides an overview of current literature on the role of neuroglia cells in ASD, with a focus on immune activation, glutamate signaling, synaptic pruning, and myelination.
Collapse
Affiliation(s)
- Vivi M Heine
- Department of Child and Adolescence Psychiatry, Emma Center for Personalized Medicine, Amsterdam Neuroscience, Emma Children's Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Stephanie Dooves
- Department of Child and Adolescence Psychiatry, Emma Center for Personalized Medicine, Amsterdam Neuroscience, Emma Children's Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Ziętek MM, Jaszczyk A, Stankiewicz AM, Sampino S. Prenatal gene-environment interactions mediate the impact of advanced maternal age on mouse offspring behavior. Sci Rep 2024; 14:31733. [PMID: 39738558 PMCID: PMC11685589 DOI: 10.1038/s41598-024-82070-x] [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: 05/24/2024] [Accepted: 12/02/2024] [Indexed: 01/02/2025] Open
Abstract
Autism spectrum disorders encompass diverse neurodevelopmental conditions marked by alterations in social communication and repetitive behaviors. Advanced maternal age is associated with an increased risk of bearing children affected by autism but the etiological factors underlying this association are not well known. Here, we investigated the effects of advanced maternal age on offspring health and behavior in two genetically divergent mouse strains: the BTBR T+ Itpr3tf/J (BTBR) mouse model of idiopathic autism, and the C57BL/6 J (B6) control strain, as a model of genetic variability. In both strains, advanced maternal age negatively affected female reproductive and pregnancy outcomes, and perturbed placental and fetal growth, and the expression of genes in the fetal brain tissues. Postnatally, advanced maternal age had strain-dependent effects on offspring sociability, learning skills, and the occurrence of perseverative behaviors, varying between male and female offspring. These findings disentangle the relationship between genetic determinants and maternal age-related factors in shaping the emergence of autism-like behaviors in mice, highlighting the interplay between maternal age, genetic variability, and prenatal programming, in the occurrence of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Marta Marlena Ziętek
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland
| | - Aneta Jaszczyk
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland
| | - Adrian Mateusz Stankiewicz
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland
| | - Silvestre Sampino
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland.
| |
Collapse
|
44
|
Jhanji M, Krall CL, Guevara A, Yoon B, Sajish M, Boccuto L, Lizarraga SB. The intersection of inflammation and DNA damage as a novel axis underlying the pathogenesis of autism spectrum disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627854. [PMID: 39713319 PMCID: PMC11661205 DOI: 10.1101/2024.12.11.627854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Autism spectrum disorders (ASD) affects 1 in 36 children and is characterized by repetitive behaviors and difficulties in social interactions and social communication. The etiology of ASD is extremely heterogeneous, with a large number of ASD cases that are of unknown or complex etiology, which suggests the potential contribution of epigenetic risk factors. In particular, epidemiological and animal model studies suggest that inflammation during pregnancy could lead to an increased risk of ASD in the offspring. However, the molecular mechanisms that contribute to ASD pathogenesis in relation to maternal inflammation during pregnancy in humans are underexplored. Several pro-inflammatory cytokines have been associated with increased autistic-like behaviors in animal models of maternal immune activation, including IL-17A. Using a combination of ASD patient lymphocytes and stem cell-derived human neurons exposed to IL-17A we discovered a shared molecular signature that highlights a metabolic and translational node that could lead to altered neuronal excitability. Further, our work on human neurons brings forward the possibility that defects in the DNA damage response could be underlying the effect of IL-17A on human excitatory neurons, linking exacerbated unrepaired DNA damage to the pathogenicity of maternal inflammation in connection to ASD.
Collapse
|
45
|
Hasegawa M, Niijima M, Kunisawa K, Teshigawara T, Kubota H, Fujigaki S, Fujigaki H, Yamamoto Y, Kim HC, Saito K, Nabeshima T, Mouri A. Maternal immune activation induces neurodevelopmental impairments of adult offspring through alterations in tryptophane-kynurenine pathway in the placenta. Biochem Biophys Res Commun 2024; 737:150922. [PMID: 39486137 DOI: 10.1016/j.bbrc.2024.150922] [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: 10/19/2024] [Revised: 10/22/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Maternal immune activation (MIA) is recognized as one of the significant environmental risk factors for neuropsychiatric disorders such as schizophrenia in adult offspring. However, the pathophysiological mechanisms remain unknown. The tryptophan (TRP)-kynurenine (KYN) pathway, influenced by inflammation, may be implicated in the pathophysiology of neuropsychiatric disorders. We investigated whether abnormal behaviors in adult offspring could be induced by MIA through alterations in the TRP-KYN pathway. MIA increased not only IL-6 expression in the placenta but also reactive oxygen species (ROS) levels in both the placenta and fetal brain and disrupted cortical layering in the fetal brain. We observed increased levels of 3-hydroxykynurenine (3-HK), a metabolite with oxidative stress properties, in both the placenta and fetal brain. In the knockout mice of kynurenine 3-monooxygenase (KMO), the enzyme responsible for 3-HK production, MIA failed to induce the abnormal behaviors in adult offspring. Notably, RO-618048, a KMO inhibitor that does not cross the blood-brain barrier (BBB), also blocked MIA-induced abnormal behaviors in adult offspring, reduced not only increased IL-6 expression in the placenta but also ROS levels in both the placenta and fetal brain, and prevented abnormal cortical development in the fetal brain. These findings suggest that MIA-induced abnormal behaviors in adult offspring may result from the increase in 3-HK levels through activation of KMO. Therefore, KMO is an attractive target for the prevention of neuropsychiatric disorders associated with MIA.
Collapse
Affiliation(s)
- Masaya Hasegawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Science, Aichi, Japan
| | - Moe Niijima
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Science, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Science, Aichi, Japan; International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Tomoaki Teshigawara
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Science, Aichi, Japan
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Science, Aichi, Japan
| | - Suwako Fujigaki
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Hidetsugu Fujigaki
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Yasuko Yamamoto
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, South Korea
| | - Kuniaki Saito
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan; Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Medical Science, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Medical Science, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan; International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Science, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan.
| |
Collapse
|
46
|
Chen Y, Zhang J, Zhang T, Wu Y, Xi Y, Wu T, Li M, Li Y, Zhou S, Wu M, Wang S. Circulating Interleukin-6 Mediates PM 2.5-Induced Ovarian Injury by Suppressing the PPARγ Pathway. RESEARCH (WASHINGTON, D.C.) 2024; 7:0538. [PMID: 39639885 PMCID: PMC11617621 DOI: 10.34133/research.0538] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Exposure to airborne fine particulate matter (PM2.5) is strongly associated with poor fertility and ovarian damage. However, the mechanism underlying this remains largely unclear. Here, we found that PM2.5 markedly impaired murine ovarian reserve, decreased hormone levels, and aggravated ovarian inflammation. Circulating interleukin-6 (IL-6) was elevated in PM2.5-exposed mice and was further confirmed to mediate this damage by IL-6 recombinant protein intervention. PM2.5 exposure led to increased alveolar macrophage infiltration in the lungs. However, alveolar macrophage clearance with clodronate liposomes could not fully reverse the elevated IL-6 levels and ovarian injury, suggesting that alveolar macrophages were probably not the only source of circulating IL-6. Further experiments indicated that IL-6 mainly targeted ovarian theca-interstitial cells and impaired testosterone synthesis via suppressing the peroxisome proliferator-activated receptor γ (PPARγ) pathway. In addition, apoptosis of granulosa cells and restriction of follicular growth were observed in co-cultures with IL-6-treated theca-interstitial cells, which could be further reversed by the PPARγ agonist. Moreover, IL-6-neutralizing antibodies ameliorated PM2.5-induced ovarian damage. Notably, increased levels of circulating IL-6 were observed in premature ovarian aging patients and were inversely associated with their ovarian function. In summary, our findings offer a mechanistic explanation for PM2.5-induced ovarian dysfunction and verify IL-6 as a biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University,
Zhengzhou University, Zhengzhou, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Zhang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Yaling Wu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Yueyue Xi
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Mo Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Yan Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Su Zhou
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Mingfu Wu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
47
|
Mohebalizadeh M, Babapour G, Maleki Aghdam M, Mohammadi T, Jafari R, Shafiei-Irannejad V. Role of Maternal Immune Factors in Neuroimmunology of Brain Development. Mol Neurobiol 2024; 61:9993-10005. [PMID: 38057641 DOI: 10.1007/s12035-023-03749-2] [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: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 12/08/2023]
Abstract
Inflammation during pregnancy may occur due to various factors. This condition, in which maternal immune system activation occurs, can affect fetal brain development and be related to neurodevelopmental diseases. MIA interacts with the fetus's brain development through maternal antibodies, cytokines, chemokines, and microglial cells. Antibodies are associated with the development of the nervous system by two mechanisms: direct binding to brain inflammatory factors and binding to brain antigens. Cytokines and chemokines have an active presence in inflammatory processes. Additionally, glial cells, defenders of the nervous system, play an essential role in synaptic modulation and neurogenesis. Maternal infections during pregnancy are the most critical factors related to MIA; however, several studies show the relation between these infections and neurodevelopmental diseases. Infection with specific viruses, such as Zika, cytomegalovirus, influenza A, and SARS-CoV-2, has revealed effects on neurodevelopment and the onset of diseases such as schizophrenia and autism. We review the relationship between maternal infections during pregnancy and their impact on neurodevelopmental processes.
Collapse
Affiliation(s)
- Mehdi Mohebalizadeh
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Urmia, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Urmia, Iran
| | - Golsa Babapour
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Mahdi Maleki Aghdam
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Urmia, Iran
| | - Tooba Mohammadi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Urmia, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Vahid Shafiei-Irannejad
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
48
|
Kawikova I, Hakenova K, Lebedeva M, Kleteckova L, Jakob L, Spicka V, Wen L, Spaniel F, Vales K. Perinatal Hypoxia and Immune System Activation in Schizophrenia Pathogenesis: Critical Considerations During COVID-19 Pandemic. Physiol Res 2024; 73:S615-S639. [PMID: 39589306 PMCID: PMC11627263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/01/2024] [Indexed: 11/27/2024] Open
Abstract
Schizophrenia, a severe psychiatric, neurodevelopmental disorder affecting about 0.29-1 % of the global population, is characterized by hallucinations, delusions, cognitive impairments, disorganized thoughts and speech, leading to significant social withdrawal and emotional blunting. During the 1980s, considerations about diseases that result from complex interactions of genetic background and environmental factors started to appear. One of the critical times of vulnerability is the perinatal period. Concerning schizophrenia, obstetric complications that are associated with hypoxia of the fetus or neonate were identified as a risk. Also, maternal infections during pregnancy were linked to schizophrenia by epidemiological, serologic and genetic studies. Research efforts then led to the development of experimental models testing the impact of perinatal hypoxia or maternal immune activation on neurodevelopmental disorders. These perinatal factors are usually studied separately, but given that the models are now validated, it is feasible to investigate both factors together. Inclusion of additional factors, such as metabolic disturbances or chronic stress, may need to be considered also. Understanding the interplay of perinatal factors in schizophrenia's etiology is crucial for developing targeted prevention and therapeutic strategies.
Collapse
Affiliation(s)
- I Kawikova
- Department of Medicine, Yale University, New Haven, CT, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Collins B, Lemanski EA, Wright-Jin E. The Importance of Including Maternal Immune Activation in Animal Models of Hypoxic-Ischemic Encephalopathy. Biomedicines 2024; 12:2559. [PMID: 39595123 PMCID: PMC11591850 DOI: 10.3390/biomedicines12112559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a perinatal brain injury that is the leading cause of cerebral palsy, developmental delay, and poor cognitive outcomes in children born at term, occurring in about 1.5 out of 1000 births. The only proven therapy for HIE is therapeutic hypothermia. However, despite this treatment, many children ultimately suffer disability, brain injury, and even death. Barriers to implementation including late diagnosis and lack of resources also lead to poorer outcomes. This demonstrates a critical need for additional treatments for HIE, and to facilitate this, we need translational models that accurately reflect risk factors and interactions present in HIE. Maternal or amniotic infection is a significant risk factor and possible cause of HIE in humans. Maternal immune activation (MIA) is a well-established model of maternal infection and inflammation that has significant developmental consequences largely characterized within the context of neurodevelopmental disorders such as autism spectrum disorder and schizophrenia. MIA can also lead to long-lasting changes within the neuroimmune system, which lead to compounding negative outcomes following a second insult. This supports the importance of understanding the interaction of maternal inflammation and hypoxic-ischemic outcomes. Animal models have been invaluable to understanding the pathophysiology of this injury and to the development of therapeutic hypothermia. However, each model system has its own limitations. Large animal models such as pigs may more accurately represent the brain and organ development and complexity in humans, while rodent models are more cost-effective and offer more possible molecular techniques. Recent studies have utilized MIA or direct inflammation prior to HIE insult. Investigators should thoughtfully consider the risk factors they wish to include in their HIE animal models. In the incorporation of MIA, investigators should consider the type, timing, and dose of the inflammatory stimulus, as well as the timing, severity, and type of hypoxic insult. Using a variety of animal models that incorporate the maternal-placental-fetal system of inflammation will most likely lead to a more robust understanding of the mechanisms of this injury that can guide future clinical decisions and therapies.
Collapse
Affiliation(s)
- Bailey Collins
- Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA; (B.C.); (E.A.L.)
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elise A. Lemanski
- Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA; (B.C.); (E.A.L.)
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elizabeth Wright-Jin
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
- Division of Neurology, Nemours Children’s Health, Wilmington, DE 19803, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
50
|
Viola MF, Franco Taveras E, Mass E. Developmental programming of tissue-resident macrophages. Front Immunol 2024; 15:1475369. [PMID: 39575254 PMCID: PMC11578957 DOI: 10.3389/fimmu.2024.1475369] [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: 08/03/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Macrophages are integral components of the innate immune system that colonize organs early in development and persist into adulthood through self-renewal. Their fate, whether they are replaced by monocytes or retain their embryonic origin, depends on tissue type and integrity. Macrophages are influenced by their environment, a phenomenon referred to as developmental programming. This influence extends beyond the local tissue microenvironment and includes soluble factors that can reach the macrophage niche. These factors include metabolites, antibodies, growth factors, and cytokines, which may originate from maternal diet, lifestyle, infections, or other developmental triggers and perturbations. These influences can alter macrophage transcriptional, epigenetic, and metabolic profiles, affecting cell-cell communication and tissue integrity. In addition to their crucial role in tissue immunity, macrophages play vital roles in tissue development and homeostasis. Consequently, developmental programming of these long-lived cells can modulate tissue physiology and pathology throughout life. In this review, we discuss the ontogeny of macrophages, the necessity of developmental programming by the niche for macrophage identity and function, and how developmental perturbations can affect the programming of macrophages and their subtissular niches, thereby influencing disease onset and progression in adulthood. Understanding these effects can inform targeted interventions or preventive strategies against diseases. Finally, understanding the consequences of developmental programming will shed light on how maternal health and disease may impact the well-being of future generations.
Collapse
Affiliation(s)
| | | | - Elvira Mass
- Developmental Biology of the Immune System, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|