1
|
Ozarslan N, Mong C, Ategeka J, Li L, Buarpung S, Robinson JF, Kizza J, Kakuru A, Kamya MR, Dorsey G, Rosenthal PJ, Gaw SL. Placental malaria induces a unique methylation profile associated with fetal growth restriction. Epigenetics 2025; 20:2475276. [PMID: 40051167 PMCID: PMC11901535 DOI: 10.1080/15592294.2025.2475276] [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: 10/22/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/12/2025] Open
Abstract
Fetal growth restriction (FGR) is associated with perinatal death and adverse birth outcomes, as well as long-term complications, including increased childhood morbidity, abnormal neurodevelopment, and cardio-metabolic diseases in adulthood. Placental epigenetic reprogramming associated with FGR may mediate these long-term outcomes. Placental malaria (PM), characterized by sequestration of Plasmodium falciparum-infected erythrocytes in placental intervillous space, is the leading global cause of FGR, but its impact on placental epigenetics is unknown. We hypothesized that placental methylomic profiling would reveal common and distinct mechanistic pathways of non-malarial and PM-associated FGR. We analyzed placentas from a US cohort with no malaria exposure (n = 12) and a cohort from eastern Uganda, a region with a high prevalence of malaria (n = 12). From each site, 8 cases of FGR and 4 healthy controls were analyzed. PM was diagnosed by placental histopathology. We compared the methylation levels of over 850K CpGs of the placentas using Infinium MethylationEPIC v1 microarray. Non-malarial FGR was associated with 65 differentially methylated CpGs (DMCs), whereas PM-FGR was associated with 133 DMCs, compared to their corresponding controls without FGR. One DMC (cg16389901, located in the promoter region of BMP4) was commonly hypomethylated in both groups. We identified 522 DMCs between non-malarial FGR vs. PM-FGR placentas, independent of differing geographic location or cellular composition. Placentas with PM-associated FGR have distinct methylation profiles compared to placentas with non-malarial FGR, suggesting novel epigenetic reprogramming in response to malaria. Larger cohort studies are needed to determine the distinct long-term health outcomes in PM-associated FGR pregnancies.
Collapse
Affiliation(s)
- Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Corina Mong
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - John Ategeka
- Infectious Diseases Research Collaboration, Uganda
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Joshua F. Robinson
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Jimmy Kizza
- Infectious Diseases Research Collaboration, Uganda
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Uganda
| | - Moses R. Kamya
- Infectious Diseases Research Collaboration, Uganda
- Department of Medicine, Makerere University, Kampala, Uganda
| | - Grant Dorsey
- Division of HIV, Global Medicine, and Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Philip J. Rosenthal
- Division of HIV, Global Medicine, and Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA, USA
| |
Collapse
|
2
|
Wang M, Zhang X, Zhong L, Zeng L, Li L, Yao P. Understanding autism: Causes, diagnosis, and advancing therapies. Brain Res Bull 2025; 227:111411. [PMID: 40449388 DOI: 10.1016/j.brainresbull.2025.111411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/28/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental condition marked by difficulties in social communication, languages, and repetitive behaviors. Its rising prevalence has made it a critical global public health issue. ASD is believed to arise from a combination of genetic and environmental influences. While some gene mutations associated with ASD have been identified, most cases lack clear genetic explanations. Evidence increasingly points to early-life environmental factors as key contributors to ASD, including advanced parental age, maternal diabetes during pregnancy, infections, hormonal imbalances, certain medications, and exposure to air pollution. Currently, ASD diagnosis relies on behavioral assessments, but the absence of specific molecular biomarkers poses significant obstacles to early detection and targeted therapies. Encouragingly, research has identified potential biomarkers, such as neuroimaging classifiers, electroencephalography patterns, eye-tracking data, digital analytics, gene expression profiles, inflammatory and chemokine markers, proteomic and metabolomic profiles, and gut microbiota characteristics. Potential therapeutical strategies under investigation include digital therapies, non-invasive brain stimulation, antioxidants, oxytocin, AVPR1a antagonists, PPAR agonists, and mTOR inhibitors. This review explores ASD across five areas: epidemiological trends, genetic mechanisms, early-life environmental factors and their potential roles, diagnostic biomarkers, and therapeutic approaches.
Collapse
Affiliation(s)
- Min Wang
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China
| | - Xiaozhuang Zhang
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China
| | - Liyan Zhong
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China
| | - Liqin Zeng
- Department of gynecology, Sun Yat-Sen University Affiliated No.8 Hospital, Shenzhen 518033, PR China
| | - Ling Li
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China.
| | - Paul Yao
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China.
| |
Collapse
|
3
|
Vellucci L, Mazza B, Barone A, Nasti A, De Simone G, Iasevoli F, de Bartolomeis A. The Role of Astrocytes in the Molecular Pathophysiology of Schizophrenia: Between Neurodevelopment and Neurodegeneration. Biomolecules 2025; 15:615. [PMID: 40427508 PMCID: PMC12109222 DOI: 10.3390/biom15050615] [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] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/05/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Schizophrenia is a chronic and severe psychiatric disorder affecting approximately 1% of the global population, characterized by disrupted synaptic plasticity and brain connectivity. While substantial evidence supports its classification as a neurodevelopmental disorder, non-canonical neurodegenerative features have also been reported, with increasing attention given to astrocytic dysfunction. Overall, in this study, we explore the role of astrocytes as a structural and functional link between neurodevelopment and neurodegeneration in schizophrenia. Specifically, we examine how astrocytes contribute to forming an aberrant substrate during early neurodevelopment, potentially predisposing individuals to later neurodegeneration. Astrocytes regulate neurotransmitter homeostasis and synaptic plasticity, influencing early vulnerability and disease progression through their involvement in Ca2⁺ signaling and dopamine-glutamate interaction-key pathways implicated in schizophrenia pathophysiology. Astrocytes differentiate via nuclear factor I-A, Sox9, and Notch pathways, occurring within a neuronal environment that may already be compromised in the early stages due to the genetic factors associated with the 'two-hits' model of schizophrenia. As a result, astrocytes may contribute to the development of an altered neural matrix, disrupting neuronal signaling, exacerbating the dopamine-glutamate imbalance, and causing excessive synaptic pruning and demyelination. These processes may underlie both the core symptoms of schizophrenia and the increased susceptibility to cognitive decline-clinically resembling neurodegeneration but driven by a distinct, poorly understood molecular substrate. Finally, astrocytes are emerging as potential pharmacological targets for antipsychotics such as clozapine, which may modulate their function by regulating glutamate clearance, redox balance, and synaptic remodeling.
Collapse
Affiliation(s)
- Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Anita Nasti
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona (UB), c. Casanova, 143, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c. Villarroel, 170, 08036 Barcelona, Spain
- Bipolar and Depressive Disorders Unit, Hospìtal Clinic de Barcelona. c. Villarroel, 170, 08036 Barcelona, Spain
| | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
4
|
Tobin NH, Li F, Zhu W, Ferbas KG, Sleasman JW, Raftery D, Kuhn L, Aldrovandi GM. Altered milk tryptophan and tryptophan metabolites and health of children born to women with HIV. RESEARCH SQUARE 2025:rs.3.rs-6229815. [PMID: 40166030 PMCID: PMC11957222 DOI: 10.21203/rs.3.rs-6229815/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Children born to women with HIV (WWH) suffer increased morbidity and, in low-income settings, have two to three times the mortality of infants born to women without HIV. The basis for this increase remains elusive. In low-income settings, breastfeeding is recommended because health benefits outweigh the risk of transmission, especially when maternal antiretroviral therapy is provided. We profiled the milk metabolome of 326 women sampled longitudinally for 18 months postpartum using global metabolomics. We identify perturbations in several metabolites, including tryptophan, dimethylarginine, and a recently discovered antiviral ribonucleotide, that are robustly associated with maternal HIV infection. Quantitative tryptophan and kynurenine levels in both milk and plasma reveal that these perturbations reflect systemic depletion of tryptophan and alterations in tryptophan catabolism in WWH. Our findings provide intriguing evidence that decreases in tryptophan availability and perturbations in tryptophan catabolism in children born to WWH may contribute to their increased morbidity and mortality.
Collapse
Affiliation(s)
- Nicole H Tobin
- Division of InfecGous Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles
| | - Fan Li
- Division of InfecGous Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles
| | | | - Kathie G Ferbas
- Division of InfecGous Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles
| | - John W Sleasman
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | | | - Louise Kuhn
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons; and Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
| | - Grace M Aldrovandi
- Division of InfecGous Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles
| |
Collapse
|
5
|
Guo S, Liu J, Wang B, Zhang X, Zhao Y, Xu J, Cao X, Zhao M, Xiao X, Zhao M. A viral infection prediction model for patients with r/r B-cell malignancies after CAR-T therapy: a retrospective analysis. Front Oncol 2025; 15:1549809. [PMID: 40190552 PMCID: PMC11968754 DOI: 10.3389/fonc.2025.1549809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Background Chimeric antigen receptor T cell (CAR-T) therapy for relapsed/refractory (r/r) B cell acute lymphoblastic leukemia (B-ALL) and B cell non-Hodgkin lymphoma (B-NHL) patients has shown promising effects, but side effects such as viral infections have been observed. Methods A total of 45 patients with r/r B-ALL and r/r B-NHL were included in this retrospective study. Patient demographics were recorded, with the primary endpoint being viral infection within 3 months post CAR-T treatment. Univariate and multivariate logistic regression analyses and least absolute shrinkage and selection operator (LASSO) regression analysis were used to analyze independent factors. The patients were divided into a training cohort of 28 and a validation cohort of 17 to construct a prediction model based on determined independent factors. The model's discrimination and calibration were assessed using the receiver operating characteristic curve (ROC), calibration plot, and decision curve analysis (DCA curve). Results The univariate and multivariate logistic regression analyses of the 43 patients showed that low baseline lymphocyte ratio was an independent risk factor and using granulocyte colony-stimulating factor (G-CSF) early was a protective factor for viral infection after CAR-T therapy in patients with B-ALL and B-NHL. Based on that, the area under the ROC curve (AUC) of the training cohort and validation cohort was 0.935 (95% CI 0.837-1.000) and 0.869 (95%CI 0.696-1.000), respectively, showing excellent predictive value. Conclusions We established a nomogram to predict the factors' influence on viral infection after CAR-T therapy and found that the ratio of baseline lymphocytes and using G-CSF early or lately were able to predict viral infection after CAR-T therapy in r/r B-ALL and B-NHL.
Collapse
Affiliation(s)
- Shujing Guo
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Jile Liu
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Bing Wang
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Xiaomei Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Yifan Zhao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Jianmei Xu
- Department of Hematology, Hebei University Affiliated Hospital, Baoding, Hebei, China
| | - Xinping Cao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Mohan Zhao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Xia Xiao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
6
|
Cilleros-Portet A, Lesseur C, Marí S, Cosin-Tomas M, Lozano M, Irizar A, Burt A, García-Santisteban I, Garrido-Martín D, Escaramís G, Hernangomez-Laderas A, Soler-Blasco R, Breeze CE, Gonzalez-Garcia BP, Santa-Marina L, Chen J, Llop S, Fernández MF, Vrijheid M, Ibarluzea J, Guxens M, Marsit C, Bustamante M, Bilbao JR, Fernandez-Jimenez N. Potentially causal associations between placental DNA methylation and schizophrenia and other neuropsychiatric disorders. Nat Commun 2025; 16:2431. [PMID: 40087310 PMCID: PMC11909199 DOI: 10.1038/s41467-025-57760-3] [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: 03/14/2023] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Increasing evidence supports the role of the placenta in neurodevelopment and in the onset of neuropsychiatric disorders. Recently, mQTL and iQTL maps have proven useful in understanding relationships between SNPs and GWAS that are not captured by eQTL. In this context, we propose that part of the genetic predisposition to complex neuropsychiatric disorders acts through placental DNA methylation. We construct a public placental cis-mQTL database including 214,830 CpG sites calculated in 368 fetal placenta DNA samples from the INMA project, and run cell type-, gestational age- and sex-imQTL models. We combine these data with summary statistics of GWAS on ten neuropsychiatric disorders using summary-based Mendelian randomization and colocalization. We also evaluate the influence of identified DNA methylation sites on placental gene expression in the RICHS cohort. We find that placental cis-mQTLs are enriched in placenta-specific active chromatin regions, and establish that part of the genetic burden for schizophrenia, bipolar disorder, and major depressive disorder confers risk through placental DNA methylation. The potential causality of several of the observed associations is reinforced by secondary association signals identified in conditional analyses, the involvement of cell type-imQTLs, and the correlation of identified DNA methylation sites with the expression levels of relevant genes in the placenta.
Collapse
Affiliation(s)
- Ariadna Cilleros-Portet
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergi Marí
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Marta Cosin-Tomas
- ISGlobal, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Manuel Lozano
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
- Preventive Medicine and Public Health, Food Sciences, Toxicology and Forensic Medicine Department, Universitat de València, Valencia, Spain
| | - Amaia Irizar
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine and Public Health, University of the Basque Country (UPV/EHU), Leioa, Spain
- Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Amber Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Iraia García-Santisteban
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Diego Garrido-Martín
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Spain
| | - Geòrgia Escaramís
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Alba Hernangomez-Laderas
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Raquel Soler-Blasco
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
- Department of Nursing, Universitat de València, Valencia, Spain
| | | | - Bárbara P Gonzalez-Garcia
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Loreto Santa-Marina
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Biogipuzkoa Health Research Institute, San Sebastian, Spain
- Department of Health of the Basque Government, Subdirectorate of Public Health of Gipuzkoa, San Sebastian, Spain
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sabrina Llop
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
| | - Mariana F Fernández
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Department of Radiology and Physical Medicine, Biomedical Research Center (CIBM), School of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Jesús Ibarluzea
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Biogipuzkoa Health Research Institute, San Sebastian, Spain
- Department of Health of the Basque Government, Subdirectorate of Public Health of Gipuzkoa, San Sebastian, Spain
| | - Mònica Guxens
- ISGlobal, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- ICREA, Barcelona, Spain
| | - Carmen Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Mariona Bustamante
- ISGlobal, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Jose Ramon Bilbao
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Nora Fernandez-Jimenez
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
7
|
Kapur RP, Vo AE, Li A, Li M, Munson J, Huang H, Del Rosario B, Cervantes O, Zhao H, Vong A, Manuel G, Li E, Devaraju M, Deng X, Baldessari A, Durning WM, Wangari S, Menz B, Germond A, English C, Coleman M, Orvis A, Sun S, Parker E, Juul S, Fountaine B, Rajagopal L, Adams Waldorf KM. Granular cytoplasmic inclusions in astrocytes and microglial activation in the fetal brain of pigtail macaques in response to maternal viral infection. Acta Neuropathol Commun 2025; 13:55. [PMID: 40069869 PMCID: PMC11895267 DOI: 10.1186/s40478-025-01970-9] [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: 01/16/2025] [Accepted: 02/21/2025] [Indexed: 03/15/2025] Open
Abstract
The fetal origins of neuropsychiatric disorders are poorly understood but have been linked to viral or inflammatory injury of the developing brain. The fetal white matter is particularly susceptible to injury as myelination, axonal growth, and deep white matter tracts become established. We have used the pigtail macaque (Macaca nemestrina) to study the maternal and fetal effects of influenza A virus (FLUAV) and Zika virus (ZIKV) infection during pregnancy, in cohorts with different time intervals between inoculation and delivery. We observed a striking histopathological alteration in a subset of astrocytes which contained granular cytoplasmic inclusions ("inclusion cells", ICs) within a specific region of the deep cerebral white matter in the fetal brains from specific FLUAV and ZIKV cohorts. Immunohistochemical and ultrastructural characteristics of ICs indicated that they are astrocytes (GFAP+) undergoing autophagocytosis (p62+) with activated lysosomes (LAMP1+, LAMP2+) and reactive changes in neighboring microglia. There was also a positive correlation between the number of ICs and LAMP1 or LAMP2 immunoreactivity in the fetal brain (LAMP1: rho 0.66; LAMP2: rho 0.54, p < 0.001 for both). Interestingly, ICs were significantly more prevalent in the 5-day FLUAV cohort and the 21-day intermediate ZIKV cohort than in controls (p < 0.005 and p = 0.04, respectively), but this relationship was not apparent in the ZIKV cohort with a shorter (2-3 days) or longer (months) time course. Virologic and immunologic assays indicated that the appearance of these cells was not linked with fetal brain infection. ICs were not observed in a macaque model of perinatal hypoxic ischemic encephalopathy. These alterations in fetal white matter are pathologically abnormal and may represent a transient neuropathologic finding that signifies a subtle brain injury in the fetus after maternal viral infection.
Collapse
Affiliation(s)
- Raj P Kapur
- Department of Laboratory Medicine and Pathology, Seattle Children's Hospital, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| | - Andrew E Vo
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Miranda Li
- School of Medicine, University of Washington, Seattle, WA, USA
| | - Jeff Munson
- Department of Psychiatry, University of Washington, Seattle, WA, USA
| | - Hazel Huang
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Briana Del Rosario
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Orlando Cervantes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Hong Zhao
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Ashley Vong
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Gygeria Manuel
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Edmunda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Monica Devaraju
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Xuemei Deng
- Department of Laboratory Medicine and Pathology, Seattle Children's Hospital, Seattle, WA, USA
| | | | | | - Solomon Wangari
- Washington National Primate Research Center, Seattle, WA, USA
| | - Brenna Menz
- Washington National Primate Research Center, Seattle, WA, USA
| | - Audrey Germond
- Washington National Primate Research Center, Seattle, WA, USA
| | - Chris English
- Washington National Primate Research Center, Seattle, WA, USA
| | - Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sidney Sun
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Ed Parker
- Department of Ophthalmology, University of Washington, Seattle, WA, USA
| | - Sandra Juul
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Brendy Fountaine
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA, USA
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| |
Collapse
|
8
|
Borrego-Ruiz A, Borrego JJ. Involvement of virus infections and antiviral agents in schizophrenia. Psychol Med 2025; 55:e73. [PMID: 40059820 PMCID: PMC12055031 DOI: 10.1017/s0033291725000467] [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] [Received: 06/17/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Schizophrenia is a chronic and complex mental disorder resulting from interactions between cumulative and synergistic genetic and environmental factors. Viral infection during the prenatal stage constitutes one of the most relevant risk factors for the development of schizophrenia later in adulthood. METHODS A narrative review was conducted to explore the link between viral infections and schizophrenia, as well as the neuropsychiatric effects of antiviral drugs, particularly in the context of this specific mental condition. Literature searches were performed using the PubMed, Scopus, and Web of Science databases. RESULTS Several viral infections, such as herpesviruses, influenza virus, Borna disease virus, and coronaviruses, can directly or indirectly disrupt normal fetal brain development by modifying gene expression in the maternal immune system, thereby contributing to the pathophysiological symptoms of schizophrenia. In addition, neuropsychiatric effects caused by antiviral drugs are frequent and represent significant adverse outcomes for viral treatment. CONCLUSIONS Epidemiological evidence suggests a potential relationship between viruses and schizophrenia. Increases in inflammatory cytokine levels and changes in the expression of key genes observed in several viral infections may constitute potential links between these viral infections and schizophrenia. Furthermore, antivirals may affect the central nervous system, although for most drugs, their mechanisms of action are still unclear, and a strong relationship between antivirals and schizophrenia has not yet been established.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Juan J. Borrego
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
9
|
Duarte RMF, Ribeiro-Barbosa ER, Ferreira FR, Espindola FS, Spini VBMG. Resveratrol prevents offspring's behavioral impairment associated with immunogenic stress during pregnancy. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111188. [PMID: 39522792 DOI: 10.1016/j.pnpbp.2024.111188] [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: 02/02/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Evidence suggests that prenatal maternal immunological stress is associated with an increased risk of neurological and psychiatric disorders in the developing offspring. Protecting the embryo during this critical period of neurodevelopment, when the brain is especially vulnerable, is therefore crucial. Polyphenols, with their antioxidant and anti-inflammatory properties, offer promising therapeutic approaches. This study demonstrated a series of behavioral changes induced by maternal immune activation (MIA) triggered by an antigenic solution derived from the H1N1 virus. These changes include significant differences in anxiety and risk assessment behaviors, increased immobility in the forced swim test, impairments in memory and object recognition, and social deficits resembling autism. The phenolic compound resveratrol (RSV) was evaluated for its in vitro antioxidant capacity and characterized using infrared spectroscopy. Administering RSV from embryonic day 14 (E14) to embrionyc day 19 (E19) during MIA effectively reduced its harmful effects on the offspring. This was evidenced by a significant restoration of social behaviors, memory, and recognition, as well as anxiolytic and antidepressant effects in the adult offspring. These findings contribute to new therapeutic strategies for preventing psychiatric disorders associated with neurodevelopmental stressors.
Collapse
Affiliation(s)
- Rener Mateus Francisco Duarte
- Department of Biochemistry and Molecular Biology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil.
| | - Erika Renata Ribeiro-Barbosa
- Department of Physiological Sciences, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | | | - Foued Salmen Espindola
- Department of Biochemistry and Molecular Biology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
| | | |
Collapse
|
10
|
Abdelbasset M, Saron WAA, Ma D, Rathore APS, Kozaki T, Zhong C, Mantri CK, Tan Y, Tung CC, Tey HL, Chu JJH, Chen J, Ng LG, Wang H, Ginhoux F, St John AL. Differential contributions of fetal mononuclear phagocytes to Zika virus neuroinvasion versus neuroprotection during congenital infection. Cell 2024; 187:7511-7532.e20. [PMID: 39532096 DOI: 10.1016/j.cell.2024.10.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/08/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Fetal immune cell functions during congenital infections are poorly understood. Zika virus (ZIKV) can vertically transmit from mother to fetus, causing nervous system infection and congenital ZIKV syndrome (CZS). We identified differential functional roles for fetal monocyte/macrophage cell types and microglia in ZIKV dissemination versus clearance using mouse models. Trafficking of ZIKV-infected primitive macrophages from the yolk sac allowed initial fetal virus inoculation, while recruited monocytes promoted non-productive neuroinflammation. Conversely, brain-resident differentiated microglia were protective, limiting infection and neuronal death. Single-cell RNA sequencing identified transcriptional profiles linked to the protective versus detrimental contributions of mononuclear phagocyte subsets. In human brain organoids, microglia also promoted neuroprotective transcriptional changes and infection clearance. Thus, microglia are protective before birth, contrasting with the disease-enhancing roles of primitive macrophages and monocytes. Differential modulation of myeloid cell phenotypes by genetically divergent ZIKVs underscores the potential of immune cells to regulate diverse outcomes during fetal infections.
Collapse
Affiliation(s)
- Muhammad Abdelbasset
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wilfried A A Saron
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Dongliang Ma
- Neuroscience & Behavioral Disorders Programme, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Abhay P S Rathore
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA
| | - Tatsuya Kozaki
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chengwei Zhong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chinmay Kumar Mantri
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Yingrou Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Chi-Ching Tung
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Hong Liang Tey
- National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine affiliated Renji Hospital, Shanghai, China
| | - Hongyan Wang
- Neuroscience & Behavioral Disorders Programme, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Ashley L St John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA; SingHealth Duke-NUS Global Health Institute, Singapore, Singapore.
| |
Collapse
|
11
|
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
|
12
|
Ozarslan N, Mong C, Ategeka J, Li L, Buarpung S, Robinson JF, Kizza J, Kakuru A, Kamya MR, Dorsey G, Rosenthal PJ, Gaw SL. Placental Malaria Induces a Unique Methylation Profile Associated with Fetal Growth Restriction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593431. [PMID: 38798500 PMCID: PMC11118523 DOI: 10.1101/2024.05.09.593431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND Fetal growth restriction (FGR) is associated with perinatal death and adverse birth outcomes, as well as long-term complications, including increased childhood morbidity, abnormal neurodevelopment, and cardio-metabolic diseases in adulthood. Placental epigenetic reprogramming associated with FGR may mediate these long-term outcomes. Placental malaria (PM), characterized by sequestration of Plasmodium falciparum-infected erythrocytes in placental intervillous space, is the leading global cause of FGR, but its impact on placental epigenetics is unknown. We hypothesized that placental methylomic profiling would reveal common and distinct mechanistic pathways of non-malarial and PM-associated FGR. RESULTS We analyzed placentas from a US cohort with no malaria exposure (n = 12) and a cohort from eastern Uganda, a region with a high prevalence of malaria (n = 12). From each site, 8 cases of FGR (defined as birth weight <10%ile for gestational age by Intergrowth-21 standard curves) and 4 healthy controls with normal weight were analyzed. PM was diagnosed by placental histopathology. We compared the methylation levels of over 850K CpGs of the placentas using Infinium MethylationEPIC v1 microarray. Non-malarial FGR was associated with 65 differentially methylated CpGs (DMCs), whereas PM-FGR was associated with 133 DMCs, compared to their corresponding controls without FGR. One DMC (cg16389901, located in the promoter region of BMP4) was commonly hypomethylated in both groups. We identified 522 DMCs between non-malarial FGR vs. PM-FGR placentas, which was independent of differing geographic location or cellular composition. CONCLUSION Placentas with PM-associated FGR have distinct methylation profiles as compared to placentas with non-malarial FGR, suggesting novel epigenetic reprogramming in response to malaria. Larger cohort studies are needed to determine the distinct long-term health outcomes in PM-associated FGR pregnancies.
Collapse
|
13
|
Prentice RE, Hunt RW, Spittle AJ, Ditchfield M, Chen J, Burns M, Flanagan EK, Wright E, Ross AL, Goldberg R, Bell SJ. Well controlled maternal inflammatory bowel disease does not increase the risk of abnormal neurocognitive outcome screening in offspring. Brain Behav Immun Health 2024; 40:100827. [PMID: 39149622 PMCID: PMC11326492 DOI: 10.1016/j.bbih.2024.100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/26/2024] [Accepted: 07/20/2024] [Indexed: 08/17/2024] Open
Abstract
Background Exposure to maternal inflammation is associated with an increased risk of neurocognitive and developmental disorders in offspring. Early diagnosis and intervention improves childhood motor and cognitive functioning. Neonatal cerebral MRI and remote app-based generalised movement assessments (GMAs) are both predictive of adverse neurocognitive outcomes but have only been used in infants at significantly increased risk for these outcomes, rather than following in utero exposure to maternal inflammatory disorders. Methods Pregnant women with inflammatory bowel disease were assessed clinically and biochemically in each trimester of pregnancy in this single centre prospective study. Neonatal cerebral MRIs were performed at 6-12 weeks post-corrected term. Two GMA videos were filmed using the 'BabyMoves' app from 12 to 16 weeks of age. MRIs and GMAs were assessed by a blinded highly qualified practitioner using validated scoring systems. Results 40/53 of invited maternal-infant dyads were recruited. C-reactive protein was elevated antenatally in less than 13%. 5/37 neonatal MRIs had incidental or obstetric trauma related gross anatomical abnormalities, with none abnormal on validated gross abnormality scoring. 3/35 GMAs were abnormal, with one GMA abnormality being clinically significant. Of those with abnormal GMAs, 2/3 were in exposed to severely active IBD in-utero. Conclusion Neonatal cerebral MRI and GMA for neurocognitive screening is feasible in the setting of maternal inflammatory bowel disease, where the risk of cerebral palsy is poorly defined and thus burdensome screening interventions are less appealing to parents. Larger studies are required to stratify adverse neurocognitive outcome risk in infants born to women with maternal inflammatory disorders, but these data are reassuring for women with IBD in remission antenatally.
Collapse
Affiliation(s)
- Ralley E Prentice
- Department of Gastroenterology, Monash Health, Melbourne, VIC, Australia
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Rod W Hunt
- Department of Neonatal Medicine, Monash Health, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Cerebral Palsy Alliance, Australia
| | - Alicia J Spittle
- Department of Physiotherapy, University of Melbourne, Melbourne, VIC, Australia
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Michael Ditchfield
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Department of Medical Imaging, Monash Children's Hospital, Melbourne, VIC, Australia
| | - Jeff Chen
- Department of Medical Imaging, Monash Children's Hospital, Melbourne, VIC, Australia
| | - Megan Burns
- Department of Gastroenterology, Monash Health, Melbourne, VIC, Australia
| | - Emma K Flanagan
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Emily Wright
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Alyson L Ross
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - Rimma Goldberg
- Department of Gastroenterology, Monash Health, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Sally J Bell
- Department of Gastroenterology, Monash Health, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Benazzato C, Lojudice F, Pöehlchen F, Leite PEC, Manucci AC, Van der Linden V, Jungmann P, Sogayar MC, Bruni-Cardoso A, Russo FB, Beltrão-Braga P. Zika virus vertical transmission induces neuroinflammation and synapse impairment in brain cells derived from children born with Congenital Zika Syndrome. Sci Rep 2024; 14:18002. [PMID: 39097642 PMCID: PMC11297915 DOI: 10.1038/s41598-024-65392-8] [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: 12/01/2023] [Accepted: 06/18/2024] [Indexed: 08/05/2024] Open
Abstract
Zika virus (ZIKV) infection was first reported in 2015 in Brazil as causing microcephaly and other developmental abnormalities in newborns, leading to the identification of Congenital Zika Syndrome (CZS). Viral infections have been considered an environmental risk factor for neurodevelopmental disorders outcome, such as Autism Spectrum Disorder (ASD). Moreover, not only the infection per se, but maternal immune system activation during pregnancy, has been linked to fetal neurodevelopmental disorders. To understand the impact of ZIKV vertical infection on brain development, we derived induced pluripotent stem cells (iPSC) from Brazilian children born with CZS, some of the patients also being diagnosed with ASD. Comparing iPSC-derived neurons from CZS with a control group, we found lower levels of pre- and postsynaptic proteins and reduced functional synapses by puncta co-localization. Furthermore, neurons and astrocytes derived from the CZS group showed decreased glutamate levels. Additionally, the CZS group exhibited elevated levels of cytokine production, one of which being IL-6, already associated with the ASD phenotype. These preliminary findings suggest that ZIKV vertical infection may cause long-lasting disruptions in brain development during fetal stages, even in the absence of the virus after birth. These disruptions could contribute to neurodevelopmental disorders manifestations such as ASD. Our study contributes with novel knowledge of the CZS outcomes and paves the way for clinical validation and the development of potential interventions to mitigate the impact of ZIKV vertical infection on neurodevelopment.
Collapse
Affiliation(s)
- Cecilia Benazzato
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, Av. Prof Lineu Prestes, 1374, 2Nd Floor, Room 235, São Paulo, SP, 05508-000, Brazil
| | - Fernando Lojudice
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo-SP, 01246-903, Brazil
| | - Felizia Pöehlchen
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, Av. Prof Lineu Prestes, 1374, 2Nd Floor, Room 235, São Paulo, SP, 05508-000, Brazil
- Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Paulo Emílio Corrêa Leite
- Clinical Research Unit of the Antonio Pedro Hospital, Federal Fluminense University, Rio de Janeiro, 24220-900, Brazil
| | - Antonio Carlos Manucci
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | | | - Patricia Jungmann
- Pathology Department, University of Pernambuco, Recife, 50670-901, Brazil
| | - Mari C Sogayar
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo-SP, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Alexandre Bruni-Cardoso
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Fabiele B Russo
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, Av. Prof Lineu Prestes, 1374, 2Nd Floor, Room 235, São Paulo, SP, 05508-000, Brazil.
| | - Patricia Beltrão-Braga
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, Av. Prof Lineu Prestes, 1374, 2Nd Floor, Room 235, São Paulo, SP, 05508-000, Brazil.
- Institute Pasteur of São Paulo, Av. Prof. Lucio Martins Rodrigues 370, A-Building, 4Th Floor, São Paulo-SP, 05508-020, Brazil.
| |
Collapse
|
15
|
Yu Y, Pan J, Zhao Y, Guo X, Yu W, Zhou F, Shu J, Huang Q. SARS-CoV-2 infection in pregnant patients on TNFα inhibitor: Real-life data with a review of literature. J Reprod Immunol 2024; 163:104220. [PMID: 38447289 DOI: 10.1016/j.jri.2024.104220] [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: 06/14/2023] [Revised: 02/04/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
Tumor necrosis factor alpha (TNFα) is involved in the occurrence of negative pregnancy outcomes. The study aimed to evaluate the safety and efficacy of the immunosuppressive TNFα inhibitors (TNFαi) in the treatment of patients with a history of recurrent reproductive failure in the context of COVID-19 pandemics. We reviewed 85 patients who received TNFαi (certolizumab pegol) during Mainland China's first wave of COVID-19 pandemic, from 21st Nov 2022-11 th Jan 2023. We also collected corresponding data from 130 pregnant patients who never used TNFαi for comparison. There were no significant differences in the history of previous pregnancy loss, miscarriage, embryo implantation failure, comorbidities and doses of COVID-19 vaccination. 82.2% and 87.7% pregnant patients contracted primary COVID-19 with symptoms in TNFαi group and no-TNFαi group. Duration of symptoms was significantly longer in TNFαi group and the incidences of cough and lethargy was significantly higher in TNFαi group. Both groups reported similar severity to same-aged close contacts, similar rates of other symptoms and hospitalization. No deaths were reported. In the in vitro fertilization (IVF) subgroup, we achieved a biochemical pregnancy loss rate of 17.4%, miscarriage rate of 21.7%, ongoing pregnancy rate and live birth rate of 34.2%. COVID-19 did not influence the live birth rate. We concluded that TNFαi administration in pregnancy was not associated with increased susceptivity to and severity of COVID-19. However, TNFαi users showed more prominent symptoms and longer recovery time. The pregnancy outcomes with TNFαi in such high-risk group for pregnancy loss was satisfactory.
Collapse
Affiliation(s)
- Yiqi Yu
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China; Reproductive Medicine Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China.
| | - Jiaying Pan
- Department of Obstetrics and Gynecology, Xianju County People's Hospital, Taizhou, Zhejiang 317399, China
| | - Yiqi Zhao
- Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
| | - Xiaoyan Guo
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China; Reproductive Medicine Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| | - Wenting Yu
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
| | - Feifei Zhou
- Center for Reproductive Medicine, Department of Traditional Chinese Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
| | - Jing Shu
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China; Reproductive Medicine Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| | - Qiongxiao Huang
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
16
|
Leung PBM, Liu Z, Zhong Y, Tubbs JD, Di Forti M, Murray RM, So HC, Sham PC, Lui SSY. Bidirectional two-sample Mendelian randomization study of differential white blood cell counts and schizophrenia. Brain Behav Immun 2024; 118:22-30. [PMID: 38355025 DOI: 10.1016/j.bbi.2024.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/15/2024] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Schizophrenia and white blood cell counts (WBC) are both complex and polygenic traits. Previous evidence suggests that increased WBC are associated with higher all-cause mortality, and other studies have found elevated WBC in first-episode psychosis and chronic schizophrenia. However, these observational findings may be confounded by antipsychotic exposures and their effects on WBC. Mendelian randomization (MR) is a useful method for examining the directions of genetically-predicted relationships between schizophrenia and WBC. METHODS We performed a two-sample MR using summary statistics from genome-wide association studies (GWAS) conducted by the Psychiatric Genomics Consortium Schizophrenia Workgroup (N = 130,644) and the Blood Cell Consortium (N = 563,946). The MR methods included inverse variance weighted (IVW), MR Egger, weighted median, MR-PRESSO, contamination mixture, and a novel approach called mixture model reciprocal causal inference (MRCI). False discovery rate was employed to correct for multiple testing. RESULTS Multiple MR methods supported bidirectional genetically-predicted relationships between lymphocyte count and schizophrenia: IVW (b = 0.026; FDR p-value = 0.008), MR Egger (b = 0.026; FDR p-value = 0.008), weighted median (b = 0.013; FDR p-value = 0.049), and MR-PRESSO (b = 0.014; FDR p-value = 0.010) in the forward direction, and IVW (OR = 1.100; FDR p-value = 0.021), MR Egger (OR = 1.231; FDR p-value < 0.001), weighted median (OR = 1.136; FDR p-value = 0.006) and MRCI (OR = 1.260; FDR p-value = 0.026) in the reverse direction. MR Egger (OR = 1.171; FDR p-value < 0.001) and MRCI (OR = 1.154; FDR p-value = 0.026) both suggested genetically-predicted eosinophil count is associated with schizophrenia, but MR Egger (b = 0.060; FDR p-value = 0.010) and contamination mixture (b = -0.013; FDR p-value = 0.045) gave ambiguous results on whether genetically predicted liability to schizophrenia would be associated with eosinophil count. MR Egger (b = 0.044; FDR p-value = 0.010) and MR-PRESSO (b = 0.009; FDR p-value = 0.045) supported genetically predicted liability to schizophrenia is associated with elevated monocyte count, and the opposite direction was also indicated by MR Egger (OR = 1.231; FDR p-value = 0.045). Lastly, unidirectional genetic liability from schizophrenia to neutrophil count were proposed by MR-PRESSO (b = 0.011; FDR p-value = 0.028) and contamination mixture (b = 0.011; FDR p-value = 0.045) method. CONCLUSION This MR study utilised multiple MR methods to obtain results suggesting bidirectional genetic genetically-predicted relationships for elevated lymphocyte counts and schizophrenia risk. In addition, moderate evidence also showed bidirectional genetically-predicted relationships between schizophrenia and monocyte counts, and unidirectional effect from genetic liability for eosinophil count to schizophrenia and from genetic liability for schizophrenia to neutrophil count. The influence of schizophrenia to eosinophil count is less certain. Our findings support the role of WBC in schizophrenia and concur with the hypothesis of neuroinflammation in schizophrenia.
Collapse
Affiliation(s)
- Perry B M Leung
- Department of Psychiatry, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Zipeng Liu
- Department of Psychiatry, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Centre for Child Health, Guangzhou, China
| | - Yuanxin Zhong
- Department of Psychiatry, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Justin D Tubbs
- Department of Psychiatry, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marta Di Forti
- Social, Genetics and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region; Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region.
| | - Pak C Sham
- Department of Psychiatry, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; Centre for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region.
| | - Simon S Y Lui
- Department of Psychiatry, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
17
|
Lim JW. Impacts of maternal COVID-19 during pregnancy on neonatal health and epidemiology. Clin Exp Pediatr 2024; 67:149-151. [PMID: 38186261 PMCID: PMC10915457 DOI: 10.3345/cep.2023.01452] [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: 10/20/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 01/09/2024] Open
Affiliation(s)
- Jae Woo Lim
- Department of Pediatrics, Konyang University College of Medicine, Daejeon, Korea
| |
Collapse
|
18
|
Xu P, Yu Y, Wu P. Role of microglia in brain development after viral infection. Front Cell Dev Biol 2024; 12:1340308. [PMID: 38298216 PMCID: PMC10825034 DOI: 10.3389/fcell.2024.1340308] [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: 11/17/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
Microglia are immune cells in the brain that originate from the yolk sac and enter the developing brain before birth. They play critical roles in brain development by supporting neural precursor proliferation, synaptic pruning, and circuit formation. However, microglia are also vulnerable to environmental factors, such as infection and stress that may alter their phenotype and function. Viral infection activates microglia to produce inflammatory cytokines and anti-viral responses that protect the brain from damage. However, excessive or prolonged microglial activation impairs brain development and leads to long-term consequences such as autism spectrum disorder and schizophrenia spectrum disorder. Moreover, certain viruses may attack microglia and deploy them as "Trojan horses" to infiltrate the brain. In this brief review, we describe the function of microglia during brain development and examine their roles after infection through microglia-neural crosstalk. We also identify limitations for current studies and highlight future investigated questions.
Collapse
Affiliation(s)
- Pei Xu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Yongjia Yu
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ping Wu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
19
|
Manti S, Spoto G, Nicotera AG, Di Rosa G, Piedimonte G. Impact of respiratory viral infections during pregnancy on the neurological outcomes of the newborn: current knowledge. Front Neurosci 2024; 17:1320319. [PMID: 38260010 PMCID: PMC10800711 DOI: 10.3389/fnins.2023.1320319] [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: 10/13/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Brain development is a complex process that begins during pregnancy, and the events occurring during this sensitive period can affect the offspring's neurodevelopmental outcomes. Respiratory viral infections are frequently reported in pregnant women, and, in the last few decades, they have been related to numerous neuropsychiatric sequelae. Respiratory viruses can disrupt brain development by directly invading the fetal circulation through vertical transmission or inducing neuroinflammation through the maternal immune activation and production of inflammatory cytokines. Influenza virus gestational infection has been consistently associated with psychotic disorders, such as schizophrenia and autism spectrum disorder, while the recent pandemic raised some concerns regarding the effects of severe acute respiratory syndrome coronavirus 2 on neurodevelopmental outcomes of children born to affected mothers. In addition, emerging evidence supports the possible role of respiratory syncytial virus infection as a risk factor for adverse neuropsychiatric consequences. Understanding the mechanisms underlying developmental dysfunction allows for improving preventive strategies, early diagnosis, and prompt interventions.
Collapse
Affiliation(s)
- Sara Manti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Giulia Spoto
- Unit of Child Neurology and Psychiatry, Department of Biomedical and Dental Sciences and of Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Antonio Gennaro Nicotera
- Unit of Child Neurology and Psychiatry, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Biomedical and Dental Sciences and of Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Giovanni Piedimonte
- Department of Pediatrics, Biochemistry and Molecular Biology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
20
|
Egorova M, Egorov V, Zabrodskaya Y. Maternal Influenza and Offspring Neurodevelopment. Curr Issues Mol Biol 2024; 46:355-366. [PMID: 38248325 PMCID: PMC10814929 DOI: 10.3390/cimb46010023] [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: 11/22/2023] [Revised: 12/23/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
This review examines the complex interactions between maternal influenza infection, the immune system, and the neurodevelopment of the offspring. It highlights the importance of high-quality studies to clarify the association between maternal exposure to the virus and neuropsychiatric disorders in the offspring. Additionally, it emphasizes that the development of accurate animal models is vital for studying the impact of infectious diseases during pregnancy and identifying potential therapeutic targets. By drawing attention to the complex nature of these interactions, this review underscores the need for ongoing research to improve the understanding and outcomes for pregnant women and their offspring.
Collapse
Affiliation(s)
- Marya Egorova
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia; (M.E.); (V.E.)
| | - Vladimir Egorov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia; (M.E.); (V.E.)
- Institute of Experimental Medicine, 12 Ulitsa Akademika Pavlova, St. Petersburg 197376, Russia
| | - Yana Zabrodskaya
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia; (M.E.); (V.E.)
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Ulitsa Polytechnicheskaya, St. Petersburg 194064, Russia
| |
Collapse
|
21
|
Jo S, Alejandro EU. RISING STARS: Mechanistic insights into maternal-fetal cross talk and islet beta-cell development. J Endocrinol 2023; 259:e230069. [PMID: 37855321 PMCID: PMC10692651 DOI: 10.1530/joe-23-0069] [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: 05/22/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023]
Abstract
The metabolic health trajectory of an individual is shaped as early as prepregnancy, during pregnancy, and lactation period. Both maternal nutrition and metabolic health status are critical factors in the programming of offspring toward an increased propensity to developing type 2 diabetes in adulthood. Pancreatic beta-cells, part of the endocrine islets, which are nutrient-sensitive tissues important for glucose metabolism, are primed early in life (the first 1000 days in humans) with limited plasticity later in life. This suggests the high importance of the developmental window of programming in utero and early in life. This review will focus on how changes to the maternal milieu increase offspring's susceptibility to diabetes through changes in pancreatic beta-cell mass and function and discuss potential mechanisms by which placental-driven nutrient availability, hormones, exosomes, and immune alterations that may impact beta-cell development in utero, thereby affecting susceptibility to type 2 diabetes in adulthood.
Collapse
Affiliation(s)
- Seokwon Jo
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Emilyn U Alejandro
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
22
|
Suleri A, Cecil C, Rommel AS, Hillegers M, White T, de Witte LD, Muetzel RL, Bergink V. Long-term effects of prenatal infection on the human brain: a prospective multimodal neuroimaging study. Transl Psychiatry 2023; 13:306. [PMID: 37789021 PMCID: PMC10547711 DOI: 10.1038/s41398-023-02597-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/05/2023] Open
Abstract
There is convincing evidence from rodent studies suggesting that prenatal infections affect the offspring's brain, but evidence in humans is limited. Here, we assessed the occurrence of common infections during each trimester of pregnancy and examined associations with brain outcomes in adolescent offspring. Our study was embedded in the Generation R Study, a large-scale sociodemographically diverse prospective birth cohort. We included 1094 mother-child dyads and investigated brain morphology (structural MRI), white matter microstructure (DTI), and functional connectivity (functional MRI), as outcomes at the age of 14. We focused on both global and focal regions. To define prenatal infections, we composed a score based on the number and type of infections during each trimester of pregnancy. Models were adjusted for several confounders. We found that prenatal infection was negatively associated with cerebral white matter volume (B = -0.069, 95% CI -0.123 to -0.015, p = 0.011), and we found an association between higher prenatal infection scores and smaller volumes of several frontotemporal regions of the brain. After multiple testing correction, we only observed an association between prenatal infections and the caudal anterior cingulate volume (B = -0.104, 95% CI -0.164 to -0.045, p < 0.001). We did not observe effects of prenatal infection on other measures of adolescent brain morphology, white matter microstructure, or functional connectivity, which is reassuring. Our results show potential regions of interest in the brain for future studies; data on the effect of severe prenatal infections on the offspring's brain in humans are needed.
Collapse
Affiliation(s)
- Anna Suleri
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Charlotte Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anna-Sophie Rommel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Manon Hillegers
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Tonya White
- Section on Social and Cognitive Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Congenital infections are a major cause of childhood multidomain neurodevelopmental disabilities. They contribute to a range of structural brain abnormalities that can cause severe neurodevelopmental impairment, cerebral palsy, epilepsy, and neurosensory impairments. New congenital infections and global viral pandemics have emerged, with some affecting the developing brain and causing neurodevelopmental concerns. This review aims to provide current understanding of fetal infections and their impact on neurodevelopment. RECENT FINDINGS There are a growing list of congenital infections causing neurodevelopmental issues, including cytomegalovirus, Zika virus, syphilis, rubella, lymphocytic choriomeningitis virus, and toxoplasmosis. Fetal exposure to maternal SARS-CoV-2 may also pose risk to the developing brain and impact neurodevelopmental outcomes, although studies have conflicting results. As Zika virus was a recently identified congenital infection, there are several new reports on child neurodevelopment in the Caribbean and Central and South America. For many congenital infections, children with in-utero exposure, even if asymptomatic at birth, may have neurodevelopmental concerns manifest over time. SUMMARY Congenital infections should be considered in the differential diagnosis of a child with neurodevelopmental impairments. Detailed pregnancy history, exposure risk, and testing should guide diagnosis and multidisciplinary evaluation. Children with congenital infections should have long-term follow-up to assess for neurodevelopmental delays and other neurosensory impairments. Children with confirmed delays or high-risk should be referred for rehabilitation therapies.
Collapse
Affiliation(s)
- Olivier Fortin
- Prenatal Pediatrics Institute, Children’s National Hospital, Washington, DC
| | - Sarah B. Mulkey
- Prenatal Pediatrics Institute, Children’s National Hospital, Washington, DC
- Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
24
|
Perez-Palomar B, Erdozain AM, Erkizia-Santamaría I, Ortega JE, Meana JJ. Maternal Immune Activation Induces Cortical Catecholaminergic Hypofunction and Cognitive Impairments in Offspring. J Neuroimmune Pharmacol 2023; 18:348-365. [PMID: 37208550 PMCID: PMC10577104 DOI: 10.1007/s11481-023-10070-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Impairment of specific cognitive domains in schizophrenia has been associated with prefrontal cortex (PFC) catecholaminergic deficits. Among other factors, prenatal exposure to infections represents an environmental risk factor for schizophrenia development in adulthood. However, it remains largely unknown whether the prenatal infection-induced changes in the brain may be associated with concrete switches in a particular neurochemical circuit, and therefore, if they could alter behavioral functions. METHODS In vitro and in vivo neurochemical evaluation of the PFC catecholaminergic systems was performed in offspring from mice undergoing maternal immune activation (MIA). The cognitive status was also evaluated. Prenatal viral infection was mimicked by polyriboinosinic-polyribocytidylic acid (poly(I:C)) administration to pregnant dams (7.5 mg/kg i.p., gestational day 9.5) and consequences were evaluated in adult offspring. RESULTS MIA-treated offspring showed disrupted recognition memory in the novel object recognition task (t = 2.30, p = 0.031). This poly(I:C)-based group displayed decreased extracellular dopamine (DA) concentrations compared to controls (t = 3.17, p = 0.0068). Potassium-evoked release of DA and noradrenaline (NA) were impaired in the poly(I:C) group (DA: Ft[10,90] = 43.33, p < 0.0001; Ftr[1,90] = 1.224, p = 0.2972; Fi[10,90] = 5.916, p < 0.0001; n = 11); (NA: Ft[10,90] = 36.27, p < 0.0001; Ftr[1,90] = 1.841, p = 0.208; Fi[10,90] = 8.686, p < 0.0001; n = 11). In the same way, amphetamine-evoked release of DA and NA were also impaired in the poly(I:C) group (DA: Ft[8,328] = 22.01, p < 0.0001; Ftr[1,328] = 4.507, p = 0.040; Fi[8,328] = 2.319, p = 0.020; n = 43); (NA: Ft[8,328] = 52.07; p < 0.0001; Ftr[1,328] = 4.322; p = 0.044; Fi[8,398] = 5.727; p < 0.0001; n = 43). This catecholamine imbalance was accompanied by increased dopamine D1 and D2 receptor expression (t = 2.64, p = 0.011 and t = 3.55, p = 0.0009; respectively), whereas tyrosine hydroxylase, DA and NA tissue content, DA and NA transporter (DAT/NET) expression and function were unaltered. CONCLUSIONS MIA induces in offspring a presynaptic catecholaminergic hypofunction in PFC with cognitive impairment. This poly(I:C)-based model reproduces catecholamine phenotypes reported in schizophrenia and represents an opportunity for the study of cognitive impairment associated to this disorder.
Collapse
Affiliation(s)
- Blanca Perez-Palomar
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, ISCIII, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, 63110, USA
| | - Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, ISCIII, Leioa, Spain
| | - Ines Erkizia-Santamaría
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain
| | - Jorge E Ortega
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, ISCIII, Leioa, Spain.
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain.
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, ISCIII, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
| |
Collapse
|
25
|
Recaioglu H, Kolk SM. Developing brain under renewed attack: viral infection during pregnancy. Front Neurosci 2023; 17:1119943. [PMID: 37700750 PMCID: PMC10493316 DOI: 10.3389/fnins.2023.1119943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/26/2023] [Indexed: 09/14/2023] Open
Abstract
Living in a globalized world, viral infections such as CHIKV, SARS-COV-2, and ZIKV have become inevitable to also infect the most vulnerable groups in our society. That poses a danger to these populations including pregnant women since the developing brain is sensitive to maternal stressors including viral infections. Upon maternal infection, the viruses can gain access to the fetus via the maternofetal barrier and even to the fetal brain during which factors such as viral receptor expression, time of infection, and the balance between antiviral immune responses and pro-viral mechanisms contribute to mother-to-fetus transmission and fetal infection. Both the direct pro-viral mechanisms and the resulting dysregulated immune response can cause multi-level impairment in the maternofetal and brain barriers and the developing brain itself leading to dysfunction or even loss of several cell populations. Thus, maternal viral infections can disturb brain development and even predispose to neurodevelopmental disorders. In this review, we discuss the potential contribution of maternal viral infections of three relevant relative recent players in the field: Zika, Chikungunya, and Severe Acute Respiratory Syndrome Coronavirus-2, to the impairment of brain development throughout the entire route.
Collapse
Affiliation(s)
| | - Sharon M. Kolk
- Faculty of Science, Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| |
Collapse
|
26
|
Kotsiri I, Resta P, Spyrantis A, Panotopoulos C, Chaniotis D, Beloukas A, Magiorkinis E. Viral Infections and Schizophrenia: A Comprehensive Review. Viruses 2023; 15:1345. [PMID: 37376644 DOI: 10.3390/v15061345] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Schizophrenia is a complex mental disorder with multiple genetic and environmental factors contributing to its pathogenesis. Viral infections have been suggested to be one of the environmental factors associated with the development of this disorder. We comprehensively review all relevant published literature focusing on the relationship between schizophrenia and various viral infections, such as influenza virus, herpes virus 1 and 2 (HSV-1 and HSV-2), cytomegalovirus (CMV), Epstein-Barr virus (EBV), retrovirus, coronavirus, and Borna virus. These viruses may interfere with the normal maturation of the brain directly or through immune-induced mediators, such as cytokines, leading to the onset of schizophrenia. Changes in the expression of critical genes and elevated levels of inflammatory cytokines have been linked to virally-induced infections and relevant immune activities in schizophrenia. Future research is necessary to understand this relationship better and provide insight into the molecular mechanisms underlying the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Ioanna Kotsiri
- Department of Internal Medicine, Asklipeion General Hospital, Voulas, 16673 Athens, Greece
| | - Panagiota Resta
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
- National AIDS Reference Centre of Southern Greece, Department of Public Health Policy, University of West Attica, 11521 Athens, Greece
| | - Alexandros Spyrantis
- Department of Internal Medicine, Asklipeion General Hospital, Voulas, 16673 Athens, Greece
| | | | - Dimitrios Chaniotis
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
| | - Apostolos Beloukas
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
- National AIDS Reference Centre of Southern Greece, Department of Public Health Policy, University of West Attica, 11521 Athens, Greece
| | - Emmanouil Magiorkinis
- Department of Laboratory Medicine, Sotiria General Hospital for Chest Diseases, 11527 Athens, Greece
| |
Collapse
|
27
|
Schmitt A, Falkai P, Papiol S. Neurodevelopmental disturbances in schizophrenia: evidence from genetic and environmental factors. J Neural Transm (Vienna) 2023; 130:195-205. [PMID: 36370183 PMCID: PMC9660136 DOI: 10.1007/s00702-022-02567-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/03/2022] [Indexed: 11/13/2022]
Abstract
Since more than 3 decades, schizophrenia (SZ) has been regarded as a neurodevelopmental disorder. The neurodevelopmental hypothesis proposes that SZ is associated with genetic and environmental risk factors, which influence connectivity in neuronal circuits during vulnerable developmental periods. We carried out a non-systematic review of genetic/environmental factors that increase SZ risk in light of its neurodevelopmental hypothesis. We also reviewed the potential impact of SZ-related environmental and genetic risk factors on grey and white matter pathology and brain function based on magnetic resonance imaging and post-mortem studies. Finally, we reviewed studies that have used patient-derived neuronal models to gain knowledge of the role of genetic and environmental factors in early developmental stages. Taken together, these studies indicate that a variety of environmental factors may interact with genetic risk factors during the pre- or postnatal period and/or during adolescence to induce symptoms of SZ in early adulthood. These risk factors induce disturbances of macro- and microconnectivity in brain regions involving the prefrontal, temporal and parietal cortices and the hippocampus. On the molecular and cellular level, a disturbed synaptic plasticity, loss of oligodendrocytes and impaired myelination have been shown in brain regions of SZ patients. These cellular/histological phenotypes are related to environmental risk factors such as obstetric complications, maternal infections and childhood trauma and genetic risk factors identified in recent genome-wide association studies. SZ-related genetic risk may contribute to active processes interfering with synaptic plasticity in the adult brain. Advances in stem cell technologies are providing promising mechanistic insights into how SZ risk factors impact the developing brain. Further research is needed to understand the timing of the different complex biological processes taking place as a result of the interplay between genetic and environmental factors.
Collapse
Affiliation(s)
- Andrea Schmitt
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany.
- Laboratory of Neuroscience (LIM27), Institute of Psychiatry, University of São Paulo, São Paulo, Brazil.
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, Munich, Germany
| | - Sergi Papiol
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstr. 7, 80336, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
28
|
Benoit I, Di Curzio D, Civetta A, Douville RN. Drosophila as a Model for Human Viral Neuroinfections. Cells 2022; 11:cells11172685. [PMID: 36078091 PMCID: PMC9454636 DOI: 10.3390/cells11172685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
The study of human neurological infection faces many technical and ethical challenges. While not as common as mammalian models, the use of Drosophila (fruit fly) in the investigation of virus–host dynamics is a powerful research tool. In this review, we focus on the benefits and caveats of using Drosophila as a model for neurological infections and neuroimmunity. Through the examination of in vitro, in vivo and transgenic systems, we highlight select examples to illustrate the use of flies for the study of exogenous and endogenous viruses associated with neurological disease. In each case, phenotypes in Drosophila are compared to those in human conditions. In addition, we discuss antiviral drug screening in flies and how investigating virus–host interactions may lead to novel antiviral drug targets. Together, we highlight standardized and reproducible readouts of fly behaviour, motor function and neurodegeneration that permit an accurate assessment of neurological outcomes for the study of viral infection in fly models. Adoption of Drosophila as a valuable model system for neurological infections has and will continue to guide the discovery of many novel virus–host interactions.
Collapse
Affiliation(s)
- Ilena Benoit
- Department of Biology, University of Winnipeg, 599 Portage Avenue, Winnipeg, MB R3B 2G3, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Ave, Winnipeg, MB R2H 2A6, Canada
| | - Domenico Di Curzio
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Ave, Winnipeg, MB R2H 2A6, Canada
| | - Alberto Civetta
- Department of Biology, University of Winnipeg, 599 Portage Avenue, Winnipeg, MB R3B 2G3, Canada
| | - Renée N. Douville
- Department of Biology, University of Winnipeg, 599 Portage Avenue, Winnipeg, MB R3B 2G3, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Ave, Winnipeg, MB R2H 2A6, Canada
- Correspondence:
| |
Collapse
|