1
|
Duan L, Yin H, Liu J, Wang W, Huang P, Liu L, Shen J, Wang Z. Maternal COVID-19 infection associated with offspring neurodevelopmental disorders. Mol Psychiatry 2025; 30:2108-2118. [PMID: 39521839 DOI: 10.1038/s41380-024-02822-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/20/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Maternal COVID-19 infection increases the incidence of neurodevelopmental disorders (NDDs) in offspring, although the underlying mechanisms have not been elucidated. This study demonstrated that COVID-19 infection during pregnancy disrupted the balance of maternal and fetal immune environments, driving alterations in astrocytes, endothelial cells, and excitatory neurons. A risk score was established using 47 unique genes in the single-cell transcriptome of gestational mothers. The high risk score in CD4 proliferating T cell level served as an indicator for increased risk of offspring NDDs. Summary-based Mendelian randomization and phenome-wide association study analyses were conducted to identify the causal association of the transcriptional changes with the increased risk of offspring NDDs. Additionally, 10 drugs were identified as potential therapeutic candidates. Our findings support a model where the maternal COVID-19 infection changed the levels of CD4 proliferating T cells, leading to the alterations of astrocytes, endothelial cells, and excitatory neurons in offspring, contributing to the increased risk of NDDs in these individuals.
Collapse
Affiliation(s)
- Lian Duan
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Huamin Yin
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxin Liu
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Wenhang Wang
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Peijun Huang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Li Liu
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Jingling Shen
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China.
| | - Zhendong Wang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
2
|
Keuls RA, Ochsner SA, O'Neill MB, O'Day DR, Miyauchi A, Campbell KM, Lanners N, Goldstein JA, Yee C, McKenna NJ, Parchem RJ, Parchem JG. Single-nucleus transcriptional profiling of the placenta reveals the syncytiotrophoblast stress response to COVID-19. Am J Obstet Gynecol 2025; 232:S160-S175.e7. [PMID: 40253079 DOI: 10.1016/j.ajog.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND COVID-19 in pregnancy is associated with placental immune activation, inflammation, and vascular malperfusion, but its impact on syncytiotrophoblast biology and function is unclear. OBJECTIVE This study aimed to determine the effects of maternal COVID-19 on placental syncytiotrophoblasts using single-nucleus transcriptional profiling and to compare placental stress responses in COVID-19 and preeclampsia. STUDY DESIGN For transcriptional characterization of syncytiotrophoblasts, we used the single-nucleus RNA sequencing platform, single-cell combinatorial indexing RNA sequencing (sci-RNA-seq3), to profile placental villi and fetal membranes from unvaccinated patients with symptomatic COVID-19 at birth (n = 4), gestational age-matched controls (n = 4), and a case of critical COVID-19 in the second trimester with delivery at term (n = 1). Clustering of nuclei and differential gene expression analysis was performed in Seurat. Gene ontology analysis was conducted using Enrichr. High-confidence transcriptional target analysis was used to identify key transcription factor nodes governing the syncytiotrophoblast response to maternal SARS-CoV-2 infection. Bioinformatic approaches were further used to compare the COVID-19 dataset to published preeclampsia gene signatures. Tissue analysis, including immunofluorescence, was conducted to validate the transcriptional data and to compare COVID-19 and preeclampsia placental histology for an expanded cohort of placentas: controls (n = 6), asymptomatic COVID-19 (n = 3), symptomatic COVID-19 (n = 5), and preeclampsia with severe features (n = 7). RESULTS The analyzed dataset comprised 15 cell clusters and 47,889 nuclei. We identified 3 clusters of syncytiotrophoblasts representing fusing and mature nuclei with overlapping but distinct transcriptional responses to COVID-19. Bioinformatic analyses indicated that COVID-19 is associated with the following alterations in syncytiotrophoblasts: (1) endoplasmic reticulum stress and activation of stress signaling pathways, including the unfolded protein response and integrated stress response; (2) regulation of gene expression by CCAAT/enhancer-binding protein beta (CEBPB), a master transcription factor of the syncytiotrophoblast lineage; and (3) upregulation of preeclampsia-associated genes. Using complementary methods, we confirmed increased levels of stress response proteins (eg, BiP, G3BP1) in syncytiotrophoblasts, unfolded protein response signaling (spliced XBP1 mRNA), and CEBPB activation (phosphorylation) in COVID-19. Increased cytotrophoblast proliferation (Ki-67) was also detected in COVID-19, consistent with a trophoblast response to injury. Markers of stress detected in preeclampsia demonstrated similarities in the placental stress phenotype of COVID-19 and preeclampsia. CONCLUSION Maternal COVID-19 is associated with syncytiotrophoblast endoplasmic reticulum stress and activation of the syncytiotrophoblast lineage transcription factor, CEBPB. Similarities between syncytiotrophoblast stress in COVID-19 and preeclampsia provide insights into their clinical association.
Collapse
Affiliation(s)
- Rachel A Keuls
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Mary B O'Neill
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Diana R O'Day
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Akihiko Miyauchi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Kadeshia M Campbell
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Natalie Lanners
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Jeffery A Goldstein
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Connor Yee
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX; Larry C. Gilstrap MD Center for Perinatal and Women's Health Research, The University of Texas Health Science Center at Houston, Houston, TX
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Ronald J Parchem
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX.
| | - Jacqueline G Parchem
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX.
| |
Collapse
|
3
|
Hardie I, Marryat L, Murray A, King J, Okelo K, Boardman JP, Lombardo MV, Stock SJ, Wood R, Auyeung B. Early childhood developmental concerns following SARS-CoV-2 infection and COVID-19 vaccination during pregnancy: a Scottish population-level retrospective cohort study. THE LANCET. CHILD & ADOLESCENT HEALTH 2025; 9:162-171. [PMID: 39978991 DOI: 10.1016/s2352-4642(25)00008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Understanding the effects of SARS-CoV-2 infection and COVID-19 vaccination during pregnancy can help inform clinical guidance and tackle vaccine hesitancy. We examined relationships between SARS-CoV-2 infection during pregnancy, COVID-19 vaccination during pregnancy, and early child developmental concerns in children aged 13-15 months in Scotland. METHODS We created a large, population-level linked administrative health dataset, combining the COVID-19 in Pregnancy in Scotland (COPS) dataset with age 13-15 month child health review data and other datasets. We included children estimated to have been conceived after May 18, 2020, and born before Sept 30, 2021, and their mothers. We used logistic regression modelling to investigate associations between SARS-CoV-2 infection during pregnancy, COVID-19 vaccination during pregnancy, and developmental concerns (ie, parent or caregiver developmental concerns and health visitor-identified concerns regarding speech-language-communication, problem solving, gross motor, personal-social, and emotional-behavioural development) measured during routine child health reviews at age 13-15 months, including adjustment for confounders and covariates. FINDINGS A total of 24 919 child-mother pairs (12 752 [51·2%] male children; 12 167 [48·8%] female children) were included. 1631 (6·5%) children were prenatally exposed to SARS-CoV-2 and 4943 (19·8%) to COVID-19 vaccination. We found no associations between SARS-CoV-2 infection during pregnancy and developmental concerns. After confounder and covariate adjustment, COVID-19 vaccination during pregnancy was associated with reduced odds of developmental concerns regarding problem solving (odds ratio 0·78 [95% CI 0·64-0·95]), personal-social (0·76 [0·61-0·95]), and emotional-behavioural (0·67 [0·48-0·92]) development, but had no associations with other developmental concerns. INTERPRETATION SARS-CoV-2 infections during pregnancy do not appear to be linked to early childhood developmental concerns, and COVID-19 vaccinations during pregnancy appear to be safe from the perspective of early childhood developmental concerns. As some developmental concerns do not become apparent until children are older than 13-15 months, future research should continue to monitor outcomes as children grow and develop. FUNDING Economic and Social Research Council.
Collapse
Affiliation(s)
- Iain Hardie
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK.
| | - Louise Marryat
- School of Health Sciences, University of Dundee, Dundee, UK
| | - Aja Murray
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| | - Josiah King
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| | - Kenneth Okelo
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| | - James P Boardman
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Sarah J Stock
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Rachael Wood
- Usher Institute, University of Edinburgh, Edinburgh, UK; Public Health Scotland, Edinburgh, UK
| | - Bonnie Auyeung
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Xu L, Zhang Y, Chen X, Hong Y, Zhang X, Hu H, Han X, Zou X, Xu M, Zhu W, Liu Y. Human Brain Organoids Model Abnormal Prenatal Neural Development Induced by Thermal Stimulation. Cell Prolif 2025; 58:e13777. [PMID: 39668124 PMCID: PMC11839188 DOI: 10.1111/cpr.13777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 12/14/2024] Open
Abstract
The developing human foetal brain is sensitive to thermal stimulation during pregnancy. However, the mechanisms by which heat exposure affects human foetal brain development remain unclear, largely due to the lack of appropriate research models for studying thermal stimulation. To address this, we have developed a periodic heating model based on brain organoids derived from human pluripotent stem cells. The model recapitulated neurodevelopmental disruptions under prenatal heat exposure at the early stages, providing a paradigm for studying the altered neurodevelopment under environmental stimulation. Our study found that periodic heat exposure led to decreased size and impaired neural tube development in the brain organoids. Bulk RNA-seq analysis revealed that the abnormal WNT signalling pathway and the reduction of G2/M progenitor cells might be involved in heat stimulation. Further investigation revealed increased neural differentiation and decreased proliferation under heat stimulation, indicating that periodic heat exposure might lead to abnormal brain development by altering key developmental processes. Hence, our model of periodically heating brain organoids provides a platform for modelling the effects of maternal fever on foetal brain development and could be extended to applications in neurodevelopmental disorders intervention.
Collapse
Affiliation(s)
- Lei Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Department of Neurology, Affiliated Zhongda HospitalSoutheast UniversityNanjingChina
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Yufan Zhang
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Xingyi Chen
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Yuan Hong
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Xu Zhang
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Hao Hu
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Xiao Han
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Xiao Zou
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
| | - Min Xu
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Wanying Zhu
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| | - Yan Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Department of Neurology, Affiliated Zhongda HospitalSoutheast UniversityNanjingChina
- Institute of Stem Cell and Neural Regeneration, School of PharmacyNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjingChina
| |
Collapse
|
5
|
Packer CH, Jasset O, Hanniford N, Brigida S, Demidkin S, Perlis RH, Edlow AG, Shook LL. Maternal-fetal cytokine profiles in acute SARS-CoV-2 "breakthrough" infection after COVID-19 vaccination. Front Immunol 2025; 15:1506203. [PMID: 39845965 PMCID: PMC11750656 DOI: 10.3389/fimmu.2024.1506203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Objective Vaccination is protective against severe COVID-19 disease, yet whether vaccination reduces COVID-19-associated inflammation in pregnancy has not been established. The objective of this study is to characterize maternal and cord cytokine profiles of acute SARS-CoV-2 "breakthrough" infection (BTI) after vaccination, compared with unvaccinated infection and uninfected controls. Study design 66 pregnant individuals enrolled in the MGH COVID-19 biorepository (March 2020-April 2022) were included. Maternal sera were collected from 26 unvaccinated and 21 vaccinated individuals with acute SARS-CoV-2 infection. Cord sera were collected at delivery. Maternal and cord sera from 19 term dyads without current or prior SARS-CoV-2 infection were analyzed as controls. Cytokines were quantified using the Human Inflammation 20-Plex ProcartaPlex assay. Results There was a significantly higher incidence of severe/critical maternal illness in unvaccinated pregnant individuals with SARS-CoV-2 compared to vaccinated (10/26 (38%) vs. 0/21 (0%), p<0.01). Significantly higher maternal levels of TNFα and CD62P were observed in vaccinated individuals with SARS-CoV-2 BTI compared with unvaccinated individuals with infection (p<0.05). Network correlation analyses revealed a distinct maternal cytokine response to SARS-CoV-2 in vaccinated vs unvaccinated individuals. Neither unvaccinated nor vaccinated SARS-CoV-2 infection resulted in elevated cord cytokines compared to controls. Multivariate analyses demonstrate distinct maternal and cord cytokine profiles in the setting of maternal SARS-CoV-2 at delivery. Conclusion Vaccination was associated with higher maternal cytokine levels during acute SARS-CoV-2 infection compared to unvaccinated infection, which may reflect vaccine-mediated priming of the immune system. A fetal inflammatory response specific to maternal SARS-CoV-2 infection was not observed.
Collapse
Affiliation(s)
- Claire H. Packer
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Olyvia Jasset
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Nikolina Hanniford
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sara Brigida
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Stepan Demidkin
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Roy H. Perlis
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Andrea G. Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| | - Lydia L. Shook
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
6
|
Kim DH, Croen LA, Iosif AM, Ames JL, Alexeeff S, Qian Y, Yolken RH, Ashwood P, Van de Water J. The association of maternal COVID-19-infection during pregnancy on the neonatal immune profile and associations with later diagnosis of neurodevelopmental disorders. Brain Behav Immun 2025; 123:1071-1080. [PMID: 39532198 DOI: 10.1016/j.bbi.2024.11.014] [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: 05/24/2024] [Revised: 10/15/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
Despite the prevalence and significant concern of COVID-19 in maternal and offspring health, little is known about the impact of COVID-19 during pregnancy on newborn immunity and neurodevelopment. This study aimed to examine 1) the relationship between maternal COVID-19 during pregnancy and newborn immune profiles and investigate the 2) associations between specific newborn immune profiles and the risk of subsequent diagnosis of a neurodevelopmental disorder (NDD) among children with prenatal exposure to COVID-19. Newborn dried bloodspots (NBS) from 545 children born at Kaiser Permanente Northern California between January 2020 and September 2021 (460 [223 males, 237 females] to COVID-19-infected [COVID+] mothers; 85 [45 males, 40 females] to COVID-19-uninfected [COVID-] mothers) were used to profile newborn immune molecules via a 42-plex cytokine/chemokine assay. Among the 460 children born to COVID+ mothers, 73 (47 males, 27 females) were later diagnosed with an NDD. In the first set of analyses examining the association between maternal COVID-19 infection during pregnancy and newborn immune profile, the results adjusted for covariates but uncorrected for multiple comparisons showed that newborns of COVID+ mothers had significantly higher levels of IL-22 (estimate [est.] = 0.16, 95 % Cl 0.01, 0.3, p = 0.04) and GM-CSF (est. = 0.27, 95 % Cl 0.09, 0.46, p = 0.004) compared to newborns of COVID- mothers. These differences were no longer statistically significant after multiple comparison adjustments. In the second analysis exploring the association between newborn profile and later diagnosis of NDD among newborns born to COVID+ mothers, the results adjusted for covariates revealed an association between higher neonatal levels of IL-22 (hazard ratio [HR] = 0.49, 95 % Cl 0.33, 0.75, p = 0.001) and lower risk of a later diagnosis of an NDD, which remained significant after multiple comparison adjustments (p = 0.04). Other neonatal cytokines/chemokines/growth factors such as sCD40L (HR = 0.7, 95 % Cl 0.57, 0.9, p = 0.009), IP-10 (HR = 0.46, 95 % Cl 0.25, 0.83, p = 0.009), MIG (HR = 0.52, 95 % Cl 0.3, 0.9, p = 0.02), FLT-3L (HR = 0.45, 95 % Cl 0.24, 0.83, p = 0.01), PDGF AB/BB (HR = 0.56, 95 % Cl 0.36, 0.99, p = 0.046), VEGF (HR = 0.57, 95 % Cl 0.34, 0.98, p = 0.04), and IL-4 (HR = 0.48, 95 % Cl 0.26, 0.93, p = 0.03) were no longer statistically significant after multiple comparison adjustments. Despite the imbalance between the number of COVID-19 exposed and unexposed newborns in this study cohort, our novel findings enhance our understanding of the potential impact of maternal COVID-19 infection during pregnancy on the developing neonatal immune system. Our findings highlight the role of immune molecules, beyond those considered to be pro-inflammatory, that may be crucial in maternal and newborn immunity against COVID-19 infection during pregnancy. Furthermore, our results suggest that reduced levels of neonatal immune molecules in newborns of COVID + mothers may be linked to an increased risk of a subsequent diagnosis of an NDD.
Collapse
Affiliation(s)
- Danielle Hj Kim
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Lisa A Croen
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
| | - Ana-Maria Iosif
- Department of Public Health Sciences, Division of Biostatistics, University of California, Davis, CA, USA
| | - Jennifer L Ames
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
| | - Stacey Alexeeff
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
| | - Yinge Qian
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, CA, USA; MIND Institute, University of California, Davis, CA, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA; Department of Medical Microbiology and Immunology, University of California, Davis, CA, USA; MIND Institute, University of California, Davis, CA, USA.
| |
Collapse
|
7
|
Ayuk HS, Arnold S, Pierzchalski A, Bauer M, Stojanovska V, Zenclussen AC. SARS-CoV-2 Activated Peripheral Blood Mononuclear Cells (PBMCs) Do Not Provoke Adverse Effects in Trophoblast Spheroids. Am J Reprod Immunol 2025; 93:e70039. [PMID: 39776066 PMCID: PMC11706221 DOI: 10.1111/aji.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/29/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
PROBLEM Although it is still uncertain whether Severe Acute Respiratory Coronavirus (SARS-CoV-2) placental infection and vertical transmission occur, inflammation during early pregnancy can have devastating consequences for gestation itself and the growing fetus. If and how SARS-CoV-2-specific immune cells negatively affect placenta functionality is still unknown. METHOD OF STUDY We stimulated peripheral blood mononuclear cells (PBMCs) from women of reproductive age with SARS-CoV-2 peptides and cocultured them with trophoblast spheroids (HTR-8/SVneo and JEG-3) to dissect if SARS-CoV-2-activated immune cells can interfere with trophoblast functionality. The activation and cytokine profile of the PBMCs were determined using multicolor flow cytometry. The functionality of trophoblast spheroids was assessed using microscopy, enzyme-linked immunosorbent assay (ELISA), and RT-qPCR. RESULTS SARS-CoV-2 S and M peptides significantly activated PBMCs (monocytes, NK cells, and T cells with memory subsets) and induced the upregulation of proinflammatory cytokines, such as IFNγ. The activated PBMCs did not impact the viability, growth rate, and invasion capabilities of trophoblast spheroids. Furthermore, the hormonal production of hCG by JEG-3 spheroids was not compromised upon coculture with the activated PBMCs. mRNA transcript levels of genes involved in trophoblast spheroid functional pathways were also not dysregulated after coculture. CONCLUSIONS Together, the findings of our in vitro coculture model, although not fully representative of in vivo conditions, strongly support the claim that the interaction of SARS-CoV-2-activated peripheral blood immune cells with trophoblast cells at the fetal-maternal interface does not negatively affect trophoblast functionality. This goes in hand with the recommendation of vaccinating pregnant women in their first trimester.
Collapse
Affiliation(s)
| | - Susanne Arnold
- Department of Environmental ImmunologyHelmholtz Centre for Environmental ResearchLeipzigSaxonyGermany
| | - Arkadiusz Pierzchalski
- Department of Environmental ImmunologyHelmholtz Centre for Environmental ResearchLeipzigSaxonyGermany
| | - Mario Bauer
- Department of Environmental ImmunologyHelmholtz Centre for Environmental ResearchLeipzigSaxonyGermany
| | - Violeta Stojanovska
- Department of Environmental ImmunologyHelmholtz Centre for Environmental ResearchLeipzigSaxonyGermany
| | - Ana Claudia Zenclussen
- Department of Environmental ImmunologyHelmholtz Centre for Environmental ResearchLeipzigSaxonyGermany
- Saxon Incubator for Translational ResearchUniversity of LeipzigLeipzigSaxonyGermany
- German Center for Child and Adolescent Health (DZKJ)Partner Site Leipzig/DresdenLeipzig/DresdenGermany
| |
Collapse
|
8
|
Pogledic I, Mankad K, Severino M, Lerman-Sagie T, Jakab A, Hadi E, Jansen AC, Bahi-Buisson N, Di Donato N, Oegema R, Mitter C, Capo I, Whitehead MT, Haldipur P, Mancini G, Huisman TAGM, Righini A, Dobyns B, Barkovich JA, Milosevic NJ, Kasprian G, Lequin M. Prenatal assessment of brain malformations on neuroimaging: an expert panel review. Brain 2024; 147:3982-4002. [PMID: 39054600 PMCID: PMC11730443 DOI: 10.1093/brain/awae253] [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: 04/26/2024] [Revised: 06/11/2024] [Accepted: 07/04/2024] [Indexed: 07/27/2024] Open
Abstract
Brain malformations represent a heterogeneous group of abnormalities of neural morphogenesis, often associated with aberrations of neuronal connectivity and brain volume. Prenatal detection of brain malformations requires a clear understanding of embryology and developmental morphology through the various stages of gestation. This expert panel review is written with the central aim of providing an easy-to-understand road map to improve prenatal detection and characterization of structural malformations based on the current understanding of normal and aberrant brain development. For every developmental stage, the utility of each available neuroimaging modality, including prenatal multiplanar neuro sonography, anatomical MRI and advanced MRI techniques, as well as further insights from post-mortem imaging, has been highlighted.
Collapse
Affiliation(s)
- Ivana Pogledic
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N3JH, UK
- UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | | | - Tally Lerman-Sagie
- Multidisciplinary foetal Neurology Center, Obstetrics & Gynecology Ultrasound Unit, Obstetrics and Gynecology Department, Wolfson Medical Center, Holon 5822012, Israel
- Faculty of Medicine, Tel Aviv University, 5822012 Tel Aviv, Israel
| | - Andras Jakab
- Center for MR Research, University Children's Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
| | - Efrat Hadi
- Department of Obstetrics and Gynecology, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, 6436624 Tel Aviv, Israel
| | - Anna C Jansen
- Pediatric Neurology Unit, Universitair Ziekenhuis Antwerpen, 2650 EdegemAntwerp, Belgium
| | - Nadia Bahi-Buisson
- Pediatric Neurology, Necker Enfants Malades, University Hospital Imagine Institute, 75015 Paris, France
| | - Natalya Di Donato
- Institute for Clinical Genetics, University Hospital, TU Dresden, 01307 Dresden, Germany
| | - Renske Oegema
- Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 AB Utrecht, The Netherlands
| | - Christian Mitter
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Ivan Capo
- Department of Histology and Embryology, Faculty of Medicine, University of Novi Sad, Novi Sad 21000, Serbia
| | - Matthew T Whitehead
- Division of Neuroradiology, Department of Radiology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine of Philadelphia, Philadelphia, PA 19105, USA
| | - Parthiv Haldipur
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Grazia Mancini
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam 3015GD, The Netherlands
| | - Thierry A G M Huisman
- Edward B. Singleton Department of Radiology, Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrea Righini
- Pediatric Radiology and Neuroradiology Department, Children’s Hospital V. Buzzi, 20154 Milan, Italy
| | - Bill Dobyns
- Department of Pediatrics, Division of Genetics and Metabolism, University of Minnesota, Minneapolis, MN 55454, USA
| | - James A Barkovich
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA
| | - Natasa Jovanov Milosevic
- Croatian Institute for Brain Research and Department of Biology, University of Zagreb, School of Medicine, 10000 Zagreb, Croatia
| | - Gregor Kasprian
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Maarten Lequin
- Edward B. Singleton Department of Radiology, Texas Children's Hospital and Baylor College of Medicine, Austin, TX 78717USA
| |
Collapse
|
9
|
Indra S, Chalak K, Das P, Mukhopadhyay A. Placenta a potential gateway of prenatal SARS-CoV-2 infection: A review. Eur J Obstet Gynecol Reprod Biol 2024; 303:123-131. [PMID: 39461078 DOI: 10.1016/j.ejogrb.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
SARS-CoV-2, the causative agent of COVID-19, can infect various tissues in the body apart from the lungs. Although placental infection remains controversial, COVID-19-associated placental abnormalities have been reported worldwide. Therefore, COVID-19 poses a significant risk for fetal distress as well. Scientists are currently debating whether such distress results from direct viral induced assault or placental damage caused by the mother's immune response. The placenta develops different histopathological lesions in response to maternal SARS-CoV-2 infection. While some studies support both theories, the transmission rate through the placenta remains low. Therefore, a more in-depth study is necessary to determine the primary cause of maternal SARS-CoV-2-induced fetal distress. This comprehensive review is aimed to shed light on the possible reasons towards fetal distress among mothers with COVID-19. This review describes the various mechanisms of viral entry along with the mechanisms by which the virus could affect the placenta. Reported cases of placental abnormalities and fetal distress symptoms have been collated to provide an overview of the current state of knowledge on vertical transmission of COVID-19.
Collapse
Affiliation(s)
- Subhashis Indra
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Kuheli Chalak
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Purbasha Das
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | | |
Collapse
|
10
|
Veloso AHN, Barbosa ADM, Ribeiro MFM, Gervásio FM. Neurodevelopment in the first year of children exposed to SARS-CoV-2 during intrauterine period: systematic review. Rev Gaucha Enferm 2024; 45:e20240020. [PMID: 39607231 DOI: 10.1590/1983-1447.2024.20240020.en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/07/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE To identify, in the literature, the implications of gestational exposure to SARS-CoV-2 on neurodevelopment in the first postnatal year, focusing on changes in the motor, personal-social, socio-emotional, and communication and language domains. METHOD Systematic review with narrative synthesis, considering neurodevelopmental outcomes, categorized according to gross and fine motor skills, personal-social interaction, socio-emotional aspects, and communication and language. Searches were conducted in PubMed, LILACS/BIREME, and EMBASE databases between January 2020 and June 2023. Two independent researchers performed selection by reading the title and abstract and applying the inclusion and exclusion criteria. Cohort studies that evaluated children up to one year old, exposed to SARS-CoV-2 in utero, were included. The Newcastle-Ottawa scale was used to assess methodological quality. RESULTS Seventeen articles were included, with methodological quality ranging from intermediate to good. The most frequently used instrument to characterize neurodevelopment was the Ages & Stages Questionnaires. Infants aged 0 to 3 months had lower scores for fine and gross motor skills. Infants aged 3 to 12 months had more fine motor, social and communication and language impairments. CONCLUSION Most infants exposed to SARS-CoV-2 showed development as expected, however delays were identified in the motor, personal-social, socio-emotional and communication and language domains according to the age group.
Collapse
|
11
|
Zeng X, Fan L, Qin Q, Zheng D, Wang H, Li M, Jiang Y, Wang H, Liu H, Liang S, Wu L, Liang S. Exogenous PD-L1 binds to PD-1 to alleviate and prevent autism-like behaviors in maternal immune activation-induced male offspring mice. Brain Behav Immun 2024; 122:527-546. [PMID: 39182588 DOI: 10.1016/j.bbi.2024.08.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder caused by the interaction of multiple pathogenic factors. Epidemiological studies and animal experiments indicate that maternal immune activation (MIA) is closely related to the development of ASD in offspring. A large number of pro-inflammatory cytokines are transferred from the placenta to the fetal brain during MIA, which impedes fetal neurodevelopment and is accompanied by activation of immune cells and microglia. Programmed cell death protein 1 (PD-1) can be highly expressed on the surface of various activated immune cells, when combined with programmed cell death-ligand 1 (PD-L1), it can activate the PD-1/PD-L1 pathway and exert powerful immunosuppressive effects, suggesting that this immune checkpoint may have the potential to treat MIA-induced ASD. This study combined bioinformatics analysis and experimental validation to explore the efficacy of Fc-fused PD-L1 (PD-L1-Fc) in treating MIA-induced ASD. Bioinformatics analysis results showed that in human placental inflammation, IL-6 was upregulated, T cells proliferated significantly, and the PD-1/PD-L1 pathway was significantly enriched. The experimental results showed that intraperitoneal injection of poly(I:C) induced MIA in pregnant mice resulted in significant expression of IL-6 in their serum, placenta, and fetal brain. At the same time, the expression of PD-1 and PD-L1 in the placenta and fetal brain increased, CD4+ T cells in the spleen were significantly activated, and PD-1 expression increased. Their offspring mice exhibited typical ASD-like behaviors. In vitro experiments on primary microglia of offspring mice have confirmed that the expression of IL-6, PD-1, and PD-L1 is significantly increased, and PD-L1-Fc effectively reduced their expression levels. In the prefrontal cortex of MIA offspring mice, there was an increase in the expression of IL-6, PD-1, and PD-L1; activation of microglial cells, and colocalization with PD-1. Then we administered brain stereotaxic injections of PD-L1-Fc to MIA offspring mice and intraperitoneal injections to MIA pregnant mice. The results indicated that PD-L1-Fc effectively suppressed neuroinflammation in the frontal cortex of offspring mice and partially ameliorated ASD-like behaviors; MIA in pregnant mice was significantly alleviated, and the offspring mice they produced did not exhibit neuroinflammation or ASD-like behaviors. In summary, we have demonstrated the therapeutic ability of PD-L1-Fc for MIA-induced ASD, aiming to provide new strategies and insights for the treatment of ASD.
Collapse
Affiliation(s)
- Xin Zeng
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Linlin Fan
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Qian Qin
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Danyang Zheng
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Han Wang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Mengyue Li
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Yutong Jiang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Hui Wang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Hao Liu
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Shengjun Liang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Lijie Wu
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China.
| | - Shuang Liang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
12
|
Jaswa EG, Huddleston HG, Lindquist KJ, Wu AHB, Bishop SL, Kim YS, Kaing A, Prahl M, Gaw SL, Corley J, Hoskin E, Cho YJ, Rogers EE, Cedars MI. In Utero Exposure to Maternal COVID-19 and Offspring Neurodevelopment Through Age 24 Months. JAMA Netw Open 2024; 7:e2439792. [PMID: 39412802 PMCID: PMC11581627 DOI: 10.1001/jamanetworkopen.2024.39792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/18/2024] [Indexed: 11/24/2024] Open
Abstract
Importance In utero exposure to maternal infections has been associated with abnormal neurodevelopment among offspring. The emergence of a new, now endemic infection (SARS-CoV-2) warrants investigating developmental implications for exposed offspring. Objective To assess whether in utero exposure to maternal COVID-19 is associated with abnormal neurodevelopmental scores among children ages 12, 18, and 24 months. Design, Setting, and Participants Data were ascertained from the ASPIRE (Assessing the Safety of Pregnancy in the Coronavirus Pandemic) trial, a prospective cohort of pregnant individuals aged 18 years or older who were enrolled before 10 weeks' gestation and their children. Individuals were recruited online from May 14, 2020, to August 23, 2021, using the Society for Assisted Reproductive Technology and BabyCenter, an online media platform. Participants from all 50 states and Puerto Rico completed activities remotely. Exposure In utero exposure to COVID-19. Main Outcomes and Measures Birth mothers completed the Ages & Stages Questionnaires, Third Edition, a validated screening tool for developmental delays, at 12, 18, and 24 months' post partum. A score below the cutoff in any domain (communication, gross motor, fine motor, problem-solving, and social skills) was considered an abnormal developmental screen (scores range from 0 to 60 in each domain, with higher scores indicating less risk for neurodevelopmental delay). Results The cohort included 2003 pregnant individuals (mean [SD] age, 33.3 [4.2] years) enrolled before 10 weeks' gestation and who completed study activities; 1750 (87.4%) had earned a college degree. Neurodevelopmental outcomes were available for 1757 children at age 12 months, 1522 at age 18 months, and 1523 at age 24 months. The prevalence of abnormal screens for exposed vs unexposed offspring at age 12 months was 64 of 198 (32.3%) vs 458 of 1559 (29.4%); at age 18 months, 36 of 161 (22.4%) vs 279 of 1361 (20.5%); and at age 24 months, 29 of 151 (19.2%) vs 230 of 1372 (16.8%). In an adjusted mixed-effects logistics regression model, no difference in risk of abnormal neurodevelopmental screens was observed at age 12 months (adjusted risk ratio [ARR], 1.07 [95% CI, 0.85-1.34]), age 18 months (ARR, 1.15 [95% CI, 0.84-1.57]), or age 24 months (ARR, 1.01 [95% CI, 0.69-1.48]). Supplemental analyses did not identify differential risk based on trimester of infection, presence vs absence of fever, or breakthrough infection following vaccination vs primary infection. Conclusions and Relevance In this cohort study of pregnant individuals and offspring, exposure to maternal COVID-19 was not associated with abnormal neurodevelopmental screening results through 24 months' post partum. Continued study of diverse groups of children is needed because, among other factors, evidence suggests sensitivity of the developing fetal brain to maternal immune activation.
Collapse
Affiliation(s)
- Eleni G. Jaswa
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Heather G. Huddleston
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Karla J. Lindquist
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Alan H. B. Wu
- Division of Clinical Chemistry, Department of Laboratory Medicine, University of California, San Francisco
| | - Somer L. Bishop
- Department of Psychiatry, University of California, San Francisco
| | - Young-Shin Kim
- Department of Psychiatry, University of California, San Francisco
| | - Amy Kaing
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Mary Prahl
- Division of Pediatric Infectious Disease and Global Health, Department of Pediatrics, University of California, San Francisco
| | - Stephanie L. Gaw
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Jamie Corley
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Elena Hoskin
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| | - Yoon Jae Cho
- Department of Psychiatry, University of California, San Francisco
| | - Elizabeth E. Rogers
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco
| | - Marcelle I. Cedars
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
| |
Collapse
|
13
|
Santana-Coelho D. Does the kynurenine pathway play a pathogenic role in autism spectrum disorder? Brain Behav Immun Health 2024; 40:100839. [PMID: 39263315 PMCID: PMC11387593 DOI: 10.1016/j.bbih.2024.100839] [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: 11/29/2023] [Revised: 05/28/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in communication, sociability, and repetitive/stereotyped behavior. The etiology of autism is diverse, with genetic susceptibility playing an important role alongside environmental insults and conditions. Human and preclinical studies have shown that ASD is commonly accompanied by inflammation, and inhibition of the inflammatory response can ameliorate, or prevent the phenotype in preclinical studies. The kynurenine pathway, responsible for tryptophan metabolism, is upregulated by inflammation. Hence, this metabolic route has drawn the attention of investigators across different disciplines such as cancer, immunology, and neuroscience. Over the past decade, studies have identified evidence that the kynurenine pathway is also altered in autism spectrum disorders. In this mini review, we will explore the current status quo of the link between the kynurenine pathway and ASD, shedding light on the compelling but still preliminary evidence of this relationship.
Collapse
|
14
|
Wilson JD, Dworsky-Fried M, Ismail N. Neurodevelopmental implications of COVID-19-induced gut microbiome dysbiosis in pregnant women. J Reprod Immunol 2024; 165:104300. [PMID: 39004033 DOI: 10.1016/j.jri.2024.104300] [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: 04/04/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
The global public health emergency of COVID-19 in January 2020 prompted a surge in research focusing on the pathogenesis and clinical manifestations of the virus. While numerous reports have been published on the acute effects of COVID-19 infection, the review explores the multifaceted long-term implications of COVID-19, with a particular focus on severe maternal COVID-19 infection, gut microbiome dysbiosis, and neurodevelopmental disorders in offspring. Severe COVID-19 infection has been associated with heightened immune system activation and gastrointestinal symptoms. Severe COVID-19 may also result in gut microbiome dysbiosis and a compromised intestinal mucosal barrier, often referred to as 'leaky gut'. Increased gut permeability facilitates the passage of inflammatory cytokines, originating from the inflamed intestinal mucosa and gut, into the bloodstream, thereby influencing fetal development during pregnancy and potentially elevating the risk of neurodevelopmental disorders such as autism and schizophrenia. The current review discusses the role of cytokine signaling molecules, microglia, and synaptic pruning, highlighting their potential involvement in the pathogenesis of neurodevelopmental disorders following maternal COVID-19 infection. Additionally, this review addresses the potential of probiotic interventions to mitigate gut dysbiosis and inflammatory responses associated with COVID-19, offering avenues for future research in optimizing maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Jacob D Wilson
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada
| | - Michaela Dworsky-Fried
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada
| | - Nafissa Ismail
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada; LIFE Research Institute, Ottawa, Ontario K1N 6N5, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|
15
|
Shook LL, Batorsky RE, De Guzman RM, McCrea LT, Brigida SM, Horng JE, Sheridan SD, Kholod O, Cook AM, Li JZ, Slonim DK, Goods BA, Perlis RH, Edlow AG. Maternal SARS-CoV-2 impacts fetal placental macrophage programs and placenta-derived microglial models of neurodevelopment. J Neuroinflammation 2024; 21:163. [PMID: 38918792 PMCID: PMC11197235 DOI: 10.1186/s12974-024-03157-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND The SARS-CoV-2 virus activates maternal and placental immune responses. Such activation in the setting of other infections during pregnancy is known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models. METHODS AND RESULTS We assessed the impact of maternal SARS-CoV-2 on HBCs isolated from 24 term placentas (N = 10 SARS-CoV-2 positive cases, 14 negative controls). Using single-cell RNA-sequencing, we demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells (HBC-iMGs), with impaired synaptic pruning behavior compared to HBC models from negative controls. CONCLUSION These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.
Collapse
Affiliation(s)
- Lydia L Shook
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, 55 Fruit Street, Thier Research Building, 903B, Boston, MA, 02114, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | | | - Rose M De Guzman
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, 55 Fruit Street, Thier Research Building, 903B, Boston, MA, 02114, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Liam T McCrea
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara M Brigida
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, 55 Fruit Street, Thier Research Building, 903B, Boston, MA, 02114, USA
| | - Joy E Horng
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven D Sheridan
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Olha Kholod
- Thayer School of Engineering and Program, Dartmouth College, Hanover, NH, USA
| | - Aidan M Cook
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Jonathan Z Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - Brittany A Goods
- Thayer School of Engineering and Program, Dartmouth College, Hanover, NH, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Roy H Perlis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea G Edlow
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, 55 Fruit Street, Thier Research Building, 903B, Boston, MA, 02114, USA.
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Auger N, Arbour L, Lewin A, Brousseau É, Healy-Profitós J, Luu TM. Congenital anomalies during Covid-19: artifact of surveillance or a real TORCH? Eur J Epidemiol 2024; 39:613-621. [PMID: 38589643 DOI: 10.1007/s10654-024-01122-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024]
Abstract
Infections in the first trimester of pregnancy can be teratogenic, but the possibility that Covid-19 could lead to birth defects is unclear. We examined whether SARS-CoV-2 infection during pregnancy or exposure to pandemic conditions were associated with the risk of congenital anomalies. We carried out a retrospective study of 420,222 neonates born in Quebec, Canada in two time periods: prepandemic (January 1, 2017 to March 12, 2020) vs. pandemic (March 13, 2020 to March 31, 2022). We classified pandemic births as early (first trimester completed before the pandemic) or late (first trimester during the pandemic), and identified patients with SARS-CoV-2 infections during pregnancy. We applied (1) adjusted log-binomial regression models to assess the association between SARS-CoV-2 infection and congenital anomalies, and (2) autoregressive interrupted time series regression to analyze temporal trends in the monthly number of defects in all patients regardless of infection. In total, 29,263 newborns (7.0%) had a congenital anomaly. First trimester SARS-CoV-2 infections were not associated with a greater risk of birth defects compared with no infection (RR 1.07, 95% CI 0.59-1.95). However, births during the late pandemic period were more likely to be diagnosed with congenital microcephaly compared with prepandemic births (RR 1.44, 95% CI 1.21-1.71). Interrupted time series analysis confirmed that the frequency of microcephaly increased during the late pandemic period, whereas other anomalies did not. We conclude that Covid-19 is likely not teratogenic, but enhanced surveillance of anomalies among late pandemic births may have heightened the detection of infants with microcephaly.
Collapse
Affiliation(s)
- Nathalie Auger
- University of Montreal Hospital Research Centre, Montreal, QC, Canada.
- Institut national de santé publique du Québec, Montreal, QC, Canada.
- Department of Social and Preventive Medicine, School of Public Health, University of Montreal, Montreal, QC, Canada.
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada.
| | - Laura Arbour
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Antoine Lewin
- Medical Affairs and Innovation, Hema-Quebec, Montreal, QC, Canada
| | - Émilie Brousseau
- University of Montreal Hospital Research Centre, Montreal, QC, Canada
- Institut national de santé publique du Québec, Montreal, QC, Canada
| | - Jessica Healy-Profitós
- University of Montreal Hospital Research Centre, Montreal, QC, Canada
- Institut national de santé publique du Québec, Montreal, QC, Canada
| | - Thuy Mai Luu
- Department of Pediatrics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
17
|
Ma Y, Duan L, Reisch B, Kimmig R, Iannaccone A, Gellhaus A. CCN1-Mediated Signaling in Placental Villous Tissues after SARS-CoV-2 Infection in Term Pregnant Women: Implications for Dysregulated Angiogenesis. Curr Issues Mol Biol 2024; 46:3533-3550. [PMID: 38666951 PMCID: PMC11049059 DOI: 10.3390/cimb46040221] [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: 03/11/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The global spread of SARS-CoV-2 has increased infections among pregnant women. This study aimed to explore placental pathology alterations and angiogenic factor levels in term pregnant women after SARS-CoV-2 infection in a retrospective single-center study. Additionally, we investigated the role and underlying mechanism of the vascular inflammation-promoting, cysteine-rich protein 61 (CYR61/CCN1) in this context. All analyses were performed in term pregnant women infected with or without SARS-CoV-2. The sFlt-1, PlGF, and sEng serum levels were quantified using ELISA. Placental protein expressions were examined by immunoblot and immunostaining. Additionally, the effect of CCN1 protein on SGHPL-5 trophoblast cells was examined. We found that SARS-CoV-2 activated the inflammatory response in pregnant women, leading to pronounced vascular alterations in placental villous tissues. Elevated serum anti-angiogenic factors (sFlt-1, sEng) upon SARS-CoV-2 infection may directly contribute to these pathological changes. Upregulated CCN1 and pNF-κB in placental villous tissues of infected patients are identified as crucial factors in placental alterations. As a conclusion, CCN1 was significantly elevated in the placentas of term pregnant women infected with SARS-CoV-2. By activating a cascade of inflammatory responses, CCN1 induced the production of the anti-angiogenic factors sFlt-1 and sEng, which may lead to abnormal placental vascular architecture.
Collapse
|
18
|
Campisciano G, Sorz A, Cason C, Zanotta N, Gionechetti F, Piazza M, Carli P, Uliana FM, Ballaminut L, Ricci G, De Seta F, Maso G, Comar M. Genital Dysbiosis and Different Systemic Immune Responses Based on the Trimester of Pregnancy in SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:4298. [PMID: 38673883 PMCID: PMC11050260 DOI: 10.3390/ijms25084298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Respiratory infections are common in pregnancy with conflicting evidence supporting their association with neonatal congenital anomalies, especially during the first trimester. We profiled cytokine and chemokine systemic responses in 242 pregnant women and their newborns after SARS-CoV-2 infection, acquired in different trimesters. Also, we tested transplacental IgG passage and maternal vaginal-rectal microbiomes. IgG transplacental passage was evident, especially with infection acquired in the first trimester. G-CSF concentration-involved in immune cell recruitment-decreased in infected women compared to uninfected ones: a beneficial event for the reduction of inflammation but detrimental to ability to fight infections at birth. The later the infection was acquired, the higher the systemic concentration of IL-8, IP-10, and MCP-1, associated with COVID-19 disease severity. All infected women showed dysbiosis of vaginal and rectal microbiomes, compared to uninfected ones. Two newborns tested positive for SARS-CoV-2 within the first 48 h of life. Notably, their mothers had acute infection at delivery. Although respiratory infections in pregnancy are reported to affect babies' health, with SARS-CoV-2 acquired early during gestation this risk seems low because of the maternal immune response. The observed vaginal and rectal dysbiosis could be relevant for neonatal microbiome establishment, although in our series immediate neonatal outcomes were reassuring.
Collapse
Affiliation(s)
- Giuseppina Campisciano
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (C.C.); (N.Z.); (P.C.); (F.M.U.); (L.B.); (M.C.)
| | - Alice Sorz
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health–IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (A.S.); (M.P.); (G.R.); (F.D.S.); (G.M.)
| | - Carolina Cason
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (C.C.); (N.Z.); (P.C.); (F.M.U.); (L.B.); (M.C.)
| | - Nunzia Zanotta
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (C.C.); (N.Z.); (P.C.); (F.M.U.); (L.B.); (M.C.)
| | - Fabrizia Gionechetti
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy;
| | - Maria Piazza
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health–IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (A.S.); (M.P.); (G.R.); (F.D.S.); (G.M.)
| | - Petra Carli
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (C.C.); (N.Z.); (P.C.); (F.M.U.); (L.B.); (M.C.)
| | - Francesca Maria Uliana
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (C.C.); (N.Z.); (P.C.); (F.M.U.); (L.B.); (M.C.)
| | - Lisa Ballaminut
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (C.C.); (N.Z.); (P.C.); (F.M.U.); (L.B.); (M.C.)
| | - Giuseppe Ricci
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health–IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (A.S.); (M.P.); (G.R.); (F.D.S.); (G.M.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Francesco De Seta
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health–IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (A.S.); (M.P.); (G.R.); (F.D.S.); (G.M.)
- Department of Obstetrics and Gynecology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| | - Gianpaolo Maso
- Department of Obstetrics and Gynecology, Institute for Maternal and Child Health–IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (A.S.); (M.P.); (G.R.); (F.D.S.); (G.M.)
| | - Manola Comar
- Department of Advanced Translational Microbiology, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65, 34137 Trieste, Italy; (C.C.); (N.Z.); (P.C.); (F.M.U.); (L.B.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| |
Collapse
|
19
|
Fortin O, DeBiasi RL, Mulkey SB. Congenital infectious encephalopathies from the intrapartum period to postnatal life. Semin Fetal Neonatal Med 2024:101526. [PMID: 38677956 DOI: 10.1016/j.siny.2024.101526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Congenital infections are a common but often underrecognized cause of fetal brain abnormalities, as well as fetal-neonatal morbidity and mortality, that should be considered by all healthcare professionals providing neurological care to fetuses and newborns. Maternal infection with various pathogens (cytomegalovirus, Toxoplasmosis, Rubella virus, Parvovirus B19, lymphocytic choriomeningitis virus, syphilis, Zika virus, varicella zoster virus) during pregnancy can be transmitted to the developing fetus, which can cause multisystem dysfunction and destructive or malformative central nervous system lesions. These can be recognized on fetal and neonatal imaging, including ultrasound and MRI. Imaging and clinical features often overlap, but some distinguishing features can help identify specific pathogens and guide subsequent testing strategies. Some pathogens can be specifically treated, and others can be managed with targeted interventions or symptomatic therapy based on expected complications. Neurological and neurodevelopmental complications related to congenital infections vary widely and are likely driven by a combination of pathophysiologic factors, alone or in combination. These include direct invasion of the fetal central nervous system by pathogens, inflammation of the maternal-placental-fetal triad in response to infection, and long-term effects of immunogenic and epigenetic changes in the fetus in response to maternal-fetal infection. Congenital infections and their neurodevelopmental impacts should be seen as an issue of public health policy, given that infection and the associated complications disproportionately affect woman and children from low- and middle-income countries and those with lower socio-economic status in high-income countries. Congenital infections may be preventable and treatable, which can improve long-term neurodevelopmental outcomes in children.
Collapse
Affiliation(s)
- Olivier Fortin
- Zickler Family Prenatal Pediatrics Institute, Children's National Hospital, Washington DC, USA
| | - Roberta L DeBiasi
- Division of Pediatric Infectious Disease, Children's National Hospital, Washington DC, USA; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington DC, USA; Department of Tropical Medicine, Microbiology and Infectious Diseases, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Sarah B Mulkey
- Zickler Family Prenatal Pediatrics Institute, Children's National Hospital, Washington DC, USA; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington DC, USA; Department of Neurology and Rehabilitation Medicine, The George Washington University School of Medicine and Health Sciences, Washington DC, USA.
| |
Collapse
|
20
|
Ho SJ, Chaput D, Sinkey RG, Garces AH, New EP, Okuka M, Sang P, Arlier S, Semerci N, Steffensen TS, Rutherford TJ, Alsina AE, Cai J, Anderson ML, Magness RR, Uversky VN, Cummings DAT, Tsibris JCM. Proteomic studies of VEGFR2 in human placentas reveal protein associations with preeclampsia, diabetes, gravidity, and labor. Cell Commun Signal 2024; 22:221. [PMID: 38594674 PMCID: PMC11003095 DOI: 10.1186/s12964-024-01567-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024] Open
Abstract
VEGFR2 (Vascular endothelial growth factor receptor 2) is a central regulator of placental angiogenesis. The study of the VEGFR2 proteome of chorionic villi at term revealed its partners MDMX (Double minute 4 protein) and PICALM (Phosphatidylinositol-binding clathrin assembly protein). Subsequently, the oxytocin receptor (OT-R) and vasopressin V1aR receptor were detected in MDMX and PICALM immunoprecipitations. Immunogold electron microscopy showed VEGFR2 on endothelial cell (EC) nuclei, mitochondria, and Hofbauer cells (HC), tissue-resident macrophages of the placenta. MDMX, PICALM, and V1aR were located on EC plasma membranes, nuclei, and HC nuclei. Unexpectedly, PICALM and OT-R were detected on EC projections into the fetal lumen and OT-R on 20-150 nm clusters therein, prompting the hypothesis that placental exosomes transport OT-R to the fetus and across the blood-brain barrier. Insights on gestational complications were gained by univariable and multivariable regression analyses associating preeclampsia with lower MDMX protein levels in membrane extracts of chorionic villi, and lower MDMX, PICALM, OT-R, and V1aR with spontaneous vaginal deliveries compared to cesarean deliveries before the onset of labor. We found select associations between higher MDMX, PICALM, OT-R protein levels and either gravidity, diabetes, BMI, maternal age, or neonatal weight, and correlations only between PICALM-OT-R (p < 2.7 × 10-8), PICALM-V1aR (p < 0.006), and OT-R-V1aR (p < 0.001). These results offer for exploration new partnerships in metabolic networks, tissue-resident immunity, and labor, notably for HC that predominantly express MDMX.
Collapse
Grants
- Department of Obstetrics and Gynecology, University of South Florida
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida
- Lisa Muma Weitz Microscopy Laboratory, University of South Florida
- Department of Chemistry, University of South Florida
- Tampa General Hospital, Tampa, Florida
- Teasley Foundation
- Department of Molecular Medicine, University of South Florida
- Department of Biology, University of Florida
- Emerging Pathogens Institute, University of Florida
Collapse
Affiliation(s)
- Shannon J Ho
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Rachel G Sinkey
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Amanda H Garces
- Lisa Muma Weitz Microscopy Laboratory, University of South Florida, Tampa, FL, USA
| | - Erika P New
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Maja Okuka
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Peng Sang
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Sefa Arlier
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Nihan Semerci
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | | | - Thomas J Rutherford
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
- Cancer Center, Tampa General Hospital, Tampa, FL, USA
| | - Angel E Alsina
- Transplant Surgery Center, Tampa General Hospital, Tampa, FL, USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Matthew L Anderson
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
- Cancer Center, Tampa General Hospital, Tampa, FL, USA
| | - Ronald R Magness
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Derek A T Cummings
- Department of Biology and Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - John C M Tsibris
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA.
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
21
|
Goyal M, Mascarenhas D, RR P, Nanavati R. Long-Term Growth and Neurodevelopmental Outcomes of Neonates Infected with SARS-CoV-2 during the COVID-19 Pandemic at 18-24 Months Corrected Age: A Prospective Observational Study. Neonatology 2024; 121:450-459. [PMID: 38583433 PMCID: PMC11318580 DOI: 10.1159/000537803] [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: 08/30/2023] [Accepted: 02/07/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION During the early coronavirus disease (COVID-19) pandemic in 2020, researchers cautioned about the potential neuroinvasive capability of the virus and long-term neurological consequences. Although a few preliminary studies have found delayed communication, fine motor, and problem-solving skills in infants after COVID-19 infection, there continues to be a paucity of data on long-term development of neonates diagnosed with COVID-19. METHODS We conducted a prospective study of 20 neonates who acquired severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection during the first wave of the pandemic (April-July 2020). At 18-24 months corrected age, we assessed neurodevelopment by Bayley Scales of Infant and Toddler Development, the third edition (BSID-III), along with growth, hearing, and vision evaluation. RESULTS The mean corrected age at assessment was 21 months 11 days ± 1 month 28 days. We found developmental delay in nearly half of the children with scores below one standard deviation in either of the BSID-III domains. Mild delay in either motor, cognitive, or language domains was found in 9 (45%) children and moderate delay in 2 (10%). Expressive language, fine motor, and receptive language were predominantly affected. None of the children had hearing impairment, blindness, or significant growth faltering including clinically severe microcephaly. The mean composite cognitive, language, and motor scores were significantly lower in those with neurodevelopmental delay (p value - 0.02, 0.000, and 0.03, respectively) without any differences in their disease characteristics. CONCLUSION Neonates infected with SARS-CoV-2 have an increased risk of developmental delays in expressive language, fine motor, and receptive language skills at 18-24 months of age. The severity of delays is predominantly mild.
Collapse
Affiliation(s)
- Medha Goyal
- Division of Neonatology, McMaster Children’s Hospital, Hamilton, ON, Canada
| | - Dwayne Mascarenhas
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Prashanth RR
- Department of Neonatology, Seth GS Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Ruchi Nanavati
- Department of Neonatology, Seth GS Medical College and King Edward Memorial Hospital, Mumbai, India
| |
Collapse
|
22
|
Wardhana MP, Kuntaman K, Utomo B, Aryananda RA, Rifdah SN, Wafa IA, Shahnaz AA, Ningrum D, Cininta NI, Ariani G, Van Lith JM, Dachlan EG. Evidence of Placental Villous Inflammation and Apoptosis in Third-Trimester Symptomatic SARS-CoV-2 Maternal Infection. Yonsei Med J 2024; 65:202-209. [PMID: 38515357 PMCID: PMC10973560 DOI: 10.3349/ymj.2023.0309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/03/2023] [Accepted: 10/30/2023] [Indexed: 03/23/2024] Open
Abstract
PURPOSE In view of conflicting reports on the ability of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) to infect placental tissue, this study aimed to further evaluate the impact of inflammation and placental damage from symptomatic third-trimester maternal COVID-19 infection. MATERIALS AND METHODS This case-control study included 32 placenta samples each from symptomatic COVID-19 pregnancy and normal non-COVID-19 pregnancy. The villous placental area's inflammatory expression [angiotensin converting enzyme-2 (ACE-2), transmembrane protease serine-2 (TMPRSS2), interferon-γ (IFN-γ), interleukin-6 (IL-6), and SARS-CoV-2 spike protein] and apoptotic rate were examined using immunohistochemistry and Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling (TUNEL) assay. Comparison and correlation analysis were used based on COVID-19 infection, placental SARS-CoV-2 spike protein evidence, and maternal severity status. RESULTS Higher expressions of TMPRSS2, IFN-γ, and trophoblast apoptotic rate were observed in the COVID-19 group (p<0.001), whereas ACE-2 and IL-6 expressions were not significantly different from the control group (p>0.05). Additionally, SARS-CoV-2 spike protein was detected in 8 (25%) placental samples of COVID-19 pregnancy. COVID-19 subgroup analysis revealed increased IFN-γ, trophoblast, and stromal apoptosis (p<0.01). Moreover, the results of the current study revealed no correlation between maternal COVID-19 severity and placental inflammation as well as the apoptotic process. CONCLUSION The presence of SARS-CoV-2 spike protein as well as altered inflammatory and apoptotic processes may indicate the presence of placental disturbance in third-trimester maternal COVID-19 infection. The lack of correlation between placental disruption and maternal severity status suggests the need for more research to understand the infection process and any potential long-term impacts on all offsprings born to COVID-19-infected pregnant women.
Collapse
Affiliation(s)
- Manggala Pasca Wardhana
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Kuntaman Kuntaman
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Clinical Microbiology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Budi Utomo
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Public Health, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Rozi Aditya Aryananda
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
| | | | - Ifan Ali Wafa
- Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Almira Aulia Shahnaz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Dahlia Ningrum
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Nareswari Imanadha Cininta
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Grace Ariani
- Anatomical Pathology Department, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Jan Mm Van Lith
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
- Department of Gynaecology and Obstetrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Erry Gumilar Dachlan
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia.
| |
Collapse
|
23
|
Kim E, Huh JR, Choi GB. Prenatal and postnatal neuroimmune interactions in neurodevelopmental disorders. Nat Immunol 2024; 25:598-606. [PMID: 38565970 DOI: 10.1038/s41590-024-01797-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024]
Abstract
The intricate relationship between immune dysregulation and neurodevelopmental disorders (NDDs) has been observed across the stages of both prenatal and postnatal development. In this Review, we provide a comprehensive overview of various maternal immune conditions, ranging from infections to chronic inflammatory conditions, that impact the neurodevelopment of the fetus during pregnancy. Furthermore, we examine the presence of immunological phenotypes, such as immune-related markers and coexisting immunological disorders, in individuals with NDDs. By delving into these findings, we shed light on the potential underlying mechanisms responsible for the high occurrence of immune dysregulation alongside NDDs. We also discuss current mouse models of NDDs and their contributions to our understanding of the immune mechanisms underlying these diseases. Additionally, we discuss how neuroimmune interactions contribute to shaping the manifestation of neurological phenotypes in individuals with NDDs while also exploring potential avenues for mitigating these effects.
Collapse
Affiliation(s)
- Eunha Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
24
|
Kim M, Choi Y, Lee M, Kang J, Kang SM, Lee DG, Yon DK. Maternal SARS-CoV-2 infection during pregnancy and subsequent risk of atopic dermatitis in offspring: a nationwide birth cohort study in South Korea. Br J Dermatol 2024; 190:576-577. [PMID: 38035775 DOI: 10.1093/bjd/ljad478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/23/2023] [Accepted: 01/13/2024] [Indexed: 12/02/2023]
Abstract
Although previous studies suggest an association between SARS-CoV-2 infection and atopic dermatitis (AD) in adults, insufficient attention has been given to the impact of SARS-CoV-2 infection on pregnant woman and their offspring. Evidence on prenatal SARS-CoV-2 infection and subsequent risk of AD is lacking; therefore, we aimed to investigate the risk of AD in infants born to women who were infected with SARS-CoV-2 during pregnancy.
Collapse
Affiliation(s)
- Minji Kim
- Center for Digital Health, Medical Science Research Institute
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
| | - Yujin Choi
- Center for Digital Health, Medical Science Research Institute
- Department of Korean Medicine, Kyung Hee University College of Korean Medicine, Seoul, South Korea
| | - Myeongcheol Lee
- Center for Digital Health, Medical Science Research Institute
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
| | - Jiseung Kang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - So Min Kang
- Center for Digital Health, Medical Science Research Institute
| | - Dong-Geol Lee
- Department of Microbiology, Dankook University, Cheonan, South Korea
| | - Dong Keon Yon
- Center for Digital Health, Medical Science Research Institute
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
- Department of Pediatrics, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, South Korea
| |
Collapse
|
25
|
Gumusoglu SB. The role of the placenta-brain axis in psychoneuroimmune programming. Brain Behav Immun Health 2024; 36:100735. [PMID: 38420039 PMCID: PMC10900837 DOI: 10.1016/j.bbih.2024.100735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/06/2024] [Accepted: 02/04/2024] [Indexed: 03/02/2024] Open
Abstract
Gestational exposures have enduring impacts on brain and neuroimmune development and function. Perturbations of pregnancy leading to placental structure/function deficits, cell stress, immune activation, and endocrine changes (metabolic, growth factors, etc.) all increase neuropsychiatric risk in offspring. The existing literature links obstetric diseases with placental involvement to offspring neuroimmune outcomes and neurodevelopmental risk. Psychoneuroimmune outcomes in offspring brain include changes to microglia, cytokine/chemokine production, cell stress, and long-term immunoreactivity. These outcomes are altered by structural, anti-angiogenic/hypoxic, inflammatory, and metabolic diseases of the placenta. This fetal programming occurs via direct placental passage or production of factors which can act directly on fetal brain substrates, or indirectly via action of circulating factors on intermediates in the placenta. Placental neuroendocrine, vascular/angiogenic, immune, and extracellular vesicular mechanisms are detailed. These mechanisms interact within various placental and pregnancy conditions. An increased understanding of the placental origins of psychoneuroimmunology will yield dividends for human health. Identifying maternal and placental biomarkers for fetal neuroimmune health may also revolutionize early diagnosis and precision psychiatry, empowering patients to make the best healthcare decisions for their families. Targeting placental mechanisms may be a valuable approach for the prevention and mitigation of intergenerational, lifelong neuropathology.
Collapse
Affiliation(s)
- Serena B. Gumusoglu
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, 200 Hawkins Dr. Iowa City, IA, 52327, USA
- Department of Psychiatry, University of Iowa Carver College of Medicine, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
26
|
Jaswa EG, Cedars MI, Lindquist KJ, Bishop SL, Kim YS, Kaing A, Prahl M, Gaw SL, Corley J, Hoskin E, Cho YJ, Rogers E, Huddleston HG. In Utero Exposure to Maternal COVID-19 Vaccination and Offspring Neurodevelopment at 12 and 18 Months. JAMA Pediatr 2024; 178:258-265. [PMID: 38252445 PMCID: PMC10804280 DOI: 10.1001/jamapediatrics.2023.5743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 01/23/2024]
Abstract
Importance Uptake of COVID-19 vaccines among pregnant individuals was hampered by safety concerns around potential risks to unborn children. Data clarifying early neurodevelopmental outcomes of offspring exposed to COVID-19 vaccination in utero are lacking. Objective To determine whether in utero exposure to maternal COVID-19 vaccination was associated with differences in scores on the Ages and Stages Questionnaire, third edition (ASQ-3), at 12 and 18 months of age. Design, Setting, and Participants This prospective cohort study, Assessing the Safety of Pregnancy During the Coronavirus Pandemic (ASPIRE), enrolled pregnant participants from May 2020 to August 2021; follow-up of children from these pregnancies is ongoing. Participants, which included pregnant individuals and their offspring from all 50 states, self-enrolled online. Study activities were performed remotely. Exposure In utero exposure of the fetus to maternal COVID-19 vaccination during pregnancy was compared with those unexposed. Main Outcomes and Measures Neurodevelopmental scores on validated ASQ-3, completed by birth mothers at 12 and 18 months. A score below the established cutoff in any of 5 subdomains (communication, gross motor, fine motor, problem solving, social skills) constituted an abnormal screen for developmental delay. Results A total of 2487 pregnant individuals (mean [SD] age, 33.3 [4.2] years) enrolled at less than 10 weeks' gestation and completed research activities, yielding a total of 2261 and 1940 infants aged 12 and 18 months, respectively, with neurodevelopmental assessments. In crude analyses, 471 of 1541 exposed infants (30.6%) screened abnormally for developmental delay at 12 months vs 203 of 720 unexposed infants (28.2%; χ2 = 1.32; P = .25); the corresponding prevalences at 18 months were 262 of 1301 (20.1%) vs 148 of 639 (23.2%), respectively (χ2 = 2.35; P = .13). In multivariable mixed-effects logistic regression models adjusting for maternal age, race, ethnicity, education, income, maternal depression, and anxiety, no difference in risk for abnormal ASQ-3 screens was observed at either time point (12 months: adjusted risk ratio [aRR], 1.14; 95% CI, 0.97-1.33; 18 months: aRR, 0.88; 95% CI, 0.72-1.07). Further adjustment for preterm birth and infant sex did not affect results (12 months: aRR, 1.16; 95% CI, 0.98-1.36; 18 months: aRR, 0.87; 95% CI, 0.71-1.07). Conclusions and Relevance Results of this cohort study suggest that COVID-19 vaccination was safe during pregnancy from the perspective of infant neurodevelopment to 18 months of age. Additional longer-term research should be conducted to corroborate these findings and buttress clinical guidance with a strong evidence base.
Collapse
Affiliation(s)
- Eleni G. Jaswa
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Marcelle I. Cedars
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Karla J. Lindquist
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco
| | - Somer L. Bishop
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Young-Shin Kim
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Amy Kaing
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Mary Prahl
- Department of Pediatrics, Division of Pediatric Infectious Disease and Global Health, University of California, San Francisco, San Francisco
| | - Stephanie L. Gaw
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal Fetal Medicine, University of California, San Francisco, San Francisco
| | - Jamie Corley
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Elena Hoskin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| | - Yoon Jae Cho
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Elizabeth Rogers
- Department of Pediatrics, Division of Neonatology, University of California, San Francisco, San Francisco
| | - Heather G. Huddleston
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, University of California, San Francisco, San Francisco
| |
Collapse
|
27
|
Pinheiro GSMA, Lemos SMA, Martins IDA, Januário GC, Cintra ML, Farias AVSR, Oliveira RMDS, Januário JN, Azevedo VMGDO, Bentes AA, Alves CRL. Effects of SARS-CoV-2 gestational exposure and risk factors on neurodevelopment until 12 months: A prospective cohort study in Brazil. Early Hum Dev 2024; 188:105918. [PMID: 38104363 DOI: 10.1016/j.earlhumdev.2023.105918] [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: 08/16/2023] [Revised: 11/26/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND The effects of SARS-CoV-2 gestational exposure on child development remain inconclusive. AIMS To analyze the effects of SARS-CoV-2 gestational exposure on neurodevelopment until 12 months. STUDY DESIGN Prospective cohort study conducted in five municipalities in Southeast Brazil from August 2021 to September 2022. SUBJECTS Infants were recruited from a serological survey performed during neonatal screening and followed up to 12 months old. We included 224 infants exposed to SARS-CoV-2 during pregnancy and 225 non-exposed, according to the serology results of the newborn as well as their mothers and the maternal antenatal RT-PCR results. OUTCOME MEASURES Developmental assessments were performed at 6 and 12 months using the Survey of Wellbeing of Young Children-Brazilian Version (SWYC-BR). Children with suspected developmental delay (SDD) at 6 and 12 months were considered at high risk for developmental delay (HRDD). Additionally, risk factors associated with SDD were examined. RESULTS There were 111 children identified with SDD and 52 with HRDD. SARS-CoV-2 gestational exposure was not associated with SDD. Exposure in the first gestational trimester increased SDD risk by 2.15 times compared to the third. Cesarean delivery predicted SDD (OR 1.56; 95%CI 1.01-2.42) and HRDD (OR 1.91; 95%CI 1.04-3.48). Additionally, suspected maternal depression predicted SDD (OR 1.76; 95%CI 1.01-3.10). CONCLUSION SARS-CoV-2 gestational exposure did not increase the developmental delay risk. However, our findings suggest that the earlier the gestational exposure, the greater the developmental delay risk at 12 months. Cesarean delivery and suspected maternal depression increased the developmental delay risk, independent of virus exposure.
Collapse
Affiliation(s)
| | - Stela Maris Aguiar Lemos
- Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190 - Santa Efigênia, Belo Horizonte, MG 30130-100, Brazil
| | - Isadora de Araújo Martins
- Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190 - Santa Efigênia, Belo Horizonte, MG 30130-100, Brazil
| | - Gabriela Cintra Januário
- Secretaria de Estado de Saúde de Minas Gerais, Rodovia Papa João Paulo II, 4143. Cidade Administrativa - Serra Verde, Belo Horizonte, MG 31630-900, Brazil
| | - Mila Lemos Cintra
- Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190 - Santa Efigênia, Belo Horizonte, MG 30130-100, Brazil
| | | | | | - José Nélio Januário
- Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190 - Santa Efigênia, Belo Horizonte, MG 30130-100, Brazil.
| | | | - Aline Almeida Bentes
- Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190 - Santa Efigênia, Belo Horizonte, MG 30130-100, Brazil.
| | - Claudia Regina Lindgren Alves
- Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190 - Santa Efigênia, Belo Horizonte, MG 30130-100, Brazil.
| |
Collapse
|
28
|
Celik IH, Tanacan A, Canpolat FE. Neonatal outcomes of maternal prenatal coronavirus infection. Pediatr Res 2024; 95:445-455. [PMID: 38057579 DOI: 10.1038/s41390-023-02950-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has led to significant changes in life and healthcare all over the world. Pregnant women and their newborns require extra attention due to the increased risk of adverse outcomes. Adverse pregnancy outcomes include intensive care unit (ICU) admission, pulmonary, cardiac, and renal impairment leading to mortality. Immaturity and variations of the neonatal immune system may be advantageous in responding to the virus. Neonates are at risk of vertical transmission and in-utero infection. Impaired intrauterine growth, prematurity, vertical transmission, and neonatal ICU admission are the most concerning issues. Data on maternal and neonatal outcomes should be interpreted cautiously due to study designs, patient characteristics, clinical variables, the effects of variants, and vaccination beyond the pandemic. Cesarean section, immediate separation of mother-infant dyads, isolation of neonates, and avoidance of breast milk were performed to reduce transmission risk at the beginning of the pandemic in the era of insufficient knowledge. Vertical transmission was found to be low with favorable short-term outcomes. Serious fetal and neonatal outcomes are not expected, according to growing evidence. Long-term effects may be associated with fetal programming. Knowledge and lessons from COVID-19 will be helpful for the next pandemic if it occurs. IMPACT: Prenatal infection with SARS-CoV-2 is associated with adverse maternal and neonatal outcomes. Our review includes the effects of COVID-19 on the fetus and neonates, transmission routes, placental effects, fetal and neonatal outcomes, and long-term effects on neonates. There is a growing body of data and evidence about the COVID-19 pandemic. Knowledge and lessons from the pandemic will be helpful for the next pandemic if it happens.
Collapse
Affiliation(s)
- Istemi Han Celik
- Department of Pediatrics, Division of Neonatology, University of Health Sciences Türkiye; Etlik Zubeyde Hanim Women's Health Teaching and Research Hospital, 06010, Ankara, Türkiye.
| | - Atakan Tanacan
- Department of Obstetrics and Gynecology, Perinatology Clinic, University of Health Sciences Turkiye, Ankara Bilkent City Hospital, 06800, Ankara, Türkiye
| | - Fuat Emre Canpolat
- Department of Pediatrics, Division of Neonatology, University of Health Sciences Türkiye, Ankara Bilkent City Hospital, 06800, Ankara, Türkiye
| |
Collapse
|
29
|
Shook LL, Batorsky RA, De Guzman RM, McCrea LT, Brigida SM, Horng JE, Sheridan SD, Kholod O, Cook AM, Li JZ, Goods BA, Perlis RH, Edlow AG. Maternal SARS-CoV-2 impacts fetal placental macrophage programs and placenta-derived microglial models of neurodevelopment. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.29.23300544. [PMID: 38234776 PMCID: PMC10793528 DOI: 10.1101/2023.12.29.23300544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The SARS-CoV-2 virus activates maternal and placental immune responses, which in the setting of other infections occurring during pregnancy are known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models. Here, we assessed the impact of maternal SARS-CoV-2 on HBCs isolated from term placentas using single-cell RNA-sequencing. We demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells, with altered morphology and impaired synaptic pruning behavior compared to HBC models from negative controls. These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.
Collapse
|
30
|
Mostallino R, Santoni M, Sagheddu C, Serra V, Orrù V, Pistis M, Castelli MP. The PPARα agonist fenofibrate reduces the cytokine imbalance in a maternal immune activation model of schizophrenia. Eur J Pharmacol 2023; 961:176172. [PMID: 37939988 DOI: 10.1016/j.ejphar.2023.176172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Maternal infections during pregnancy may increase the risk of psychiatric disorders in offspring. We recently demonstrated that activation of peroxisome proliferator-activate receptor-α (PPARα), with the clinically available agonist fenofibrate (FEN), attenuates the neurodevelopmental disturbances induced by maternal immune activation (MIA) in rat offspring. We hypothesized that fenofibrate might reduce MIA-induced cytokine imbalance using a MIA model based on the viral mimetic polyriboinosinic-polyribocytidilic acid [poly (I:C)]. By using the Bio-Plex Multiplex-Immunoassay-System, we measured cytokine/chemokine/growth factor levels in maternal serum and in the fetal brain of rats treated with fenofibrate, at 6 and 24 h after poly (I:C). We found that MIA induced time-dependent changes in the levels of several cytokines/chemokines/colony-stimulating factors (CSFs). Specifically, the maternal serum of the poly (I:C)/control (CTRL) group showed increased levels of (i) proinflammatory chemokine macrophage inflammatory protein 1-alpha (MIP-1α), (ii) tumor necrosis factor-alpha (TNF-α), the monocyte chemoattractant protein-1 (MCP-1), the macrophage (M-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Conversely, in the fetal brain of the poly (I:C)/CTRL group, interleukin 12p70 and MIP-1α levels were lower than in vehicle (veh)/CTRL group. Notably, MIP-1α, TNF-α, keratinocyte derived chemokine (GRO/KC), GM-CSF, and M-CSF levels were lower in the poly (I:C)/FEN than in poly (I:C)/CTRL rats, suggesting the protective role of the PPARα agonist. PPARα might represent a therapeutic target to attenuate MIA-induced inflammation.
Collapse
Affiliation(s)
- Rafaela Mostallino
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Michele Santoni
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Valentina Serra
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; Neuroscience Institute, National Research Council of Italy, Section of Cagliari, Italy; Unit of Clinical Pharmacology, University Hospital, Cagliari, Italy.
| | - M Paola Castelli
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy.
| |
Collapse
|
31
|
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
|
32
|
Zhang L, Lin C, Zhu J, He Y, Zhan M, Xia X, Yang N, Yang K, Wang B, Zhong Z, Wang Y, Ding W, Yang Y. Restoring prefrontal cortical excitation-inhibition balance with cannabidiol ameliorates neurobehavioral abnormalities in a mouse model of neurodevelopmental disorders. Neuropharmacology 2023; 240:109715. [PMID: 37716533 DOI: 10.1016/j.neuropharm.2023.109715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/18/2023]
Abstract
Maternal immune activation (MIA) resulting from viral infections during pregnancy is linked to increased rates of neurodevelopmental disorders in offspring. However, the mechanisms underlying MIA-induced neurobehavioral abnormalities remain unclear. Here, we used a poly (I:C)-induced MIA mouse model to demonstrate the presence of multiple behavioral deficits in male offspring. Through RNA sequencing (RNA-seq), we identified significant upregulation of genes involved in axonogenesis, synaptogenesis, and glutamatergic synaptic neurotransmission in the mPFC of MIA mice. Electrophysiological analyses further revealed an excitatory-inhibitory (E/I) synaptic imbalance in mPFC pyramidal neurons, leading to hyperactivity in this brain region. Cannabidiol (CBD) effectively alleviated the behavioral abnormalities observed in MIA offspring by reducing glutamatergic transmission and enhancing GABAergic neurotransmission of mPFC pyramidal neurons. Activation of GPR55 by lipid lysophosphatidylinositol (LPI), an endogenous GPR55 agonist, specifically in the mPFC of healthy animals led to MIA-associated behavioral phenotypes, which CBD could effectively reverse. Moreover, we found that a GPR55 antagonist can mimic CBD's beneficial effects, indicating that CBD's therapeutic effects are mediated via the LPI-GPR55 signaling pathway. Therefore, we identified mPFC as a primary node of a neural network that mediates MIA-induced behavioral abnormalities in offspring. Our work provides insights into the mechanisms underlying the developmental consequences of MIA and identifies CBD as a promising therapeutic approach to alleviate these effects.
Collapse
Affiliation(s)
- Lu Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chunqiao Lin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jiushuang Zhu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yan He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Meng Zhan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiuwen Xia
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ni Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Kun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Baojia Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhanqion Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yili Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Youjun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
33
|
Erdogan MA, Turk M, Doganay GD, Sever IH, Ozkul B, Sogut I, Eroglu E, Uyanikgil Y, Erbas O. Prenatal SARS-CoV-2 Spike Protein Exposure Induces Autism-Like Neurobehavioral Changes in Male Neonatal Rats. J Neuroimmune Pharmacol 2023; 18:573-591. [PMID: 37889404 DOI: 10.1007/s11481-023-10089-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023]
Abstract
Recent research on placental, embryo, and brain organoids suggests that the COVID-19 virus may potentially affect embryonic organs, including the brain. Given the established link between SARS-CoV-2 spike protein and neuroinflammation, we sought to investigate the effects of exposure to this protein during pregnancy. We divided pregnant rats into three groups: Group 1 received a 1 ml/kg saline solution, Group 2 received 150 μg/kg adjuvant aluminum hydroxide (AAH), and Group 3 received 40 μg/kg spike protein + 150 μg/kg AAH at 10 and 14 days of gestation. On postnatal day 21 (P21), we randomly separated 60 littermates (10 male-female) into control, AAH-exposed, and spike protein-exposed groups. At P50, we conducted behavioral analyses on these mature animals and performed MR spectroscopy. Subsequently, all animals were sacrificed, and their brains were subject to biochemical and histological analysis. Our findings indicate that male rats exposed to the spike protein displayed a higher rate of impaired performance on behavioral studies, including the three-chamber social test, passive avoidance learning analysis, open field test, rotarod test, and novelty-induced cultivation behavior, indicative of autistic symptoms. Exposure to the spike protein (male) induced gliosis and neuronal cell death in the CA1-CA3 regions of the hippocampus and cerebellum. The spike protein-exposed male rats exhibited significantly greater levels of malondialdehyde (MDA), tumor necrosis factor alpha (TNF-α), interleukin-17 (IL-17), nuclear factor kappa B (NF-κB), and lactate and lower levels of brain-derived neurotrophic factor (BDNF) than the control group. Our study suggests a potential association between prenatal exposure to COVID-19 spike protein and neurodevelopmental problems, such as ASD. These findings highlight the importance of further research into the potential effects of the COVID-19 virus on embryonic and fetal development and the potential long-term consequences for neurodevelopment.
Collapse
Affiliation(s)
- Mumin Alper Erdogan
- Faculty of Medicine, Department of Physiology, Izmir Katip Celebi University, Izmir, Turkey.
| | - Miray Turk
- Graduate School, Department of Molecular Biology-Genetics and Biotechnology, Istanbul Technical University, 34469, Istanbul, Turkey
| | - Gizem Dinler Doganay
- Graduate School, Department of Molecular Biology-Genetics and Biotechnology, Istanbul Technical University, 34469, Istanbul, Turkey
- Faculty of Science and Letters, Department of Molecular Biology and Genetics, Istanbul Technical University, 34469, Istanbul, Turkey
| | - Ibrahim Halil Sever
- Faculty of Medicine, Department of Radiology, Demiroğlu Bilim University, Istanbul, Turkey
| | - Bahattin Ozkul
- School of Medicine, Department of Radiology, Istanbul Atlas University, Istanbul, Turkey
| | - Ibrahim Sogut
- Faculty of Medicine, Department of Biochemistry, Demiroğlu Bilim University, Istanbul, Turkey
| | - Ebru Eroglu
- Faculty of Medicine, Department of Histology and Embryology, Ege University, Izmir, Turkey
| | - Yigit Uyanikgil
- Faculty of Medicine, Department of Histology and Embryology, Ege University, Izmir, Turkey
| | - Oytun Erbas
- Faculty of Medicine, Department of Physiology, Demiroğlu Bilim University, Istanbul, Turkey
| |
Collapse
|
34
|
Power SD, Stewart E, Zielke LG, Byrne EP, Douglas A, Ortega-de San Luis C, Lynch L, Ryan TJ. Immune activation state modulates infant engram expression across development. SCIENCE ADVANCES 2023; 9:eadg9921. [PMID: 37939176 PMCID: PMC10631722 DOI: 10.1126/sciadv.adg9921] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Infantile amnesia is possibly the most ubiquitous form of memory loss in mammals. We investigated how memories are stored in the brain throughout development by integrating engram labeling technology with mouse models of infantile amnesia. Here, we found a phenomenon in which male offspring in maternal immune activation models of autism spectrum disorder do not experience infantile amnesia. Maternal immune activation altered engram ensemble size and dendritic spine plasticity. We rescued the same apparently forgotten infantile memories in neurotypical mice by optogenetically reactivating dentate gyrus engram cells labeled during complex experiences in infancy. Furthermore, we permanently reinstated lost infantile memories by artificially updating the memory engram, demonstrating that infantile amnesia is a reversible process. Our findings suggest not only that infantile amnesia is due to a reversible retrieval deficit in engram expression but also that immune activation during development modulates innate, and reversible, forgetting switches that determine whether infantile amnesia will occur.
Collapse
Affiliation(s)
- Sarah D. Power
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Erika Stewart
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Louisa G. Zielke
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Eric P. Byrne
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Aaron Douglas
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomás J. Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, Canada
| |
Collapse
|
35
|
Ramasauskaite D, Grinciute D. Review of short-term and long-term adverse effects of covid-19 vaccination during pregnancy. Travel Med Infect Dis 2023; 56:102667. [PMID: 37951411 DOI: 10.1016/j.tmaid.2023.102667] [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: 05/31/2023] [Revised: 09/22/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND The covid-19 pandemic sparked a debate about the safety of vaccines during pregnancy. However, pregnant women were excluded from the Pfizer-BioNTech vaccine phase 3 trials. As two years have passed since the first Covid-19 vaccine and more studies have been conducted, we want to evaluate the scientific literature to determine any actual risks in taking the vaccine during pregnancy. METHODS We conducted literature research using PubMed and Google Scholar databases from January to April 2023. As the review considers short- and long-term adverse effects it was divided into two parts. The first part was conducted as a systematic review. The second concerning long-term negative effects due to lack of research is a literature review. The inclusion criteria for the systematic review part were singleton pregnancies, women vaccinated during pregnancy, and studies from 2020 and later. The most common short-term pregnancy adverse effects were included in the search: preterm delivery, small gestation age, intrauterine death, congenital defects, stillborn, fetal growth retardation, spontaneous abortion. Maternal immune activation was the primary concern for the long-term adverse effects and whether vaccination could cause it. The search terms included maternal immune activation, fetal neurodevelopment, neuropsychiatric disorders and the studies used were from 2019. RESULTS Most studies showed no significant difference in short-term adverse effects between vaccinated and non-vaccinated women and their fetuses. However, the literature is insufficient to evaluate possible long-term adverse effects. CONCLUSION Available evidence supports the safety of administering SARS-CoV-2 vaccines to pregnant women, but further systematic reviews and meta-analyses are essential. Maternal immune activation caused by vaccination may impact a child's neurodevelopment and should be a concern for future studies.
Collapse
Affiliation(s)
- Diana Ramasauskaite
- Center of Obstetrics and Gynaecology, Vilnius University Faculty of Medicine, PO: Santariškių 2, Vilnius, LT08661, Lithuania.
| | | |
Collapse
|
36
|
Bacchini PL, Sammartano A, Manfredi P, Bidetti ML, Malpeli M, Magliani M, Maradini F, Ippolito L. Is SARS-CoV-2 vertical transmission still a current problem? A case report on a diagnosed SARS-CoV-2 infection with a positive sample of urines. ACTA BIO-MEDICA : ATENEI PARMENSIS 2023; 94:e2023234. [PMID: 37850765 PMCID: PMC10644935 DOI: 10.23750/abm.v94i5.14864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/02/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND AND AIM Current data suggest little to no possibility of original COVID-19 transmission in pregnant women to the fetus during pregnancy or childbirth. Warning with Omicron new variants has decreased. CASE REPORT A clinical case of a SARS-CoV-2 virus transplacental infection of a newborn, born at the end of 2022, from a mother who tested positive for Sars-covid-2 and positive IgM SARS-CoV-2 anti-virus. The newborn tested positive for SARS-CoV-2 12 hours after birth, and was clinically symptomatic after three days, an increase in IgM antibodies was not found, although the virus was identified in the urine samples through molecular tests. The insufficient time to determine the presence of antibodies and the immune system's state of immaturity can explain the lack of IgM in the newborn's blood at 14 days after birth. CONCLUSIONS The Omicron SARS-CoV-2 keeps provoking infections among newborns, especially if the mother contracts it during the third trimester. The host response is most likely influenced by the newborn's peculiar state of immune immaturity. Just before birth, a positive nasal swab and the presence of a positive urine examination confirmed the diagnosis of intraplacental exposure. Research on the virus through molecular tests of urines can represent an additional technique when an aetiological framework of the infection is necessary and a distinction between congenital and post-natal forms.
Collapse
Affiliation(s)
| | | | - Piera Manfredi
- Pediatrics, Fidenza Hospital, 43036, Fidenza (PR), Italy.
| | | | - Monica Malpeli
- Clinical Pathology Unit, Medical and Diagnostic Department, Fidenza Hospital, Fidenza (PR), Italy.
| | - Magda Magliani
- Clinical Pathology Unit, Medical and Diagnostic Department, Fidenza Hospital, Fidenza (PR), Italy.
| | - Fabio Maradini
- Clinical Pathology Unit, Medical and Diagnostic Department, Fidenza Hospital, Fidenza (PR), Italy.
| | - Luigi Ippolito
- Clinical Pathology Unit, Medical and Diagnostic Department, Fidenza Hospital, Fidenza (PR), Italy.
| |
Collapse
|
37
|
Jain S, Allen IE, Song D, Piao X. Cytokine responses to SARS-COV2 infection in mother-infant dyads: a systematic review and meta-analysis. Front Pediatr 2023; 11:1277697. [PMID: 37915987 PMCID: PMC10616592 DOI: 10.3389/fped.2023.1277697] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Background The COVID-19 pandemic has affected a significant number of pregnant women worldwide, but studies on immune responses have presented conflicting results. This study aims to systematically review cytokine profiles in pregnant women with SARS-CoV-2 infection and their infants to evaluate immune responses and potential transplacental transfer of cytokines. Materials and methods A comprehensive search of 4 databases was conducted to identify relevant studies. Inclusion criteria included studies measuring individual cytokines in pregnant women and/or their neonates. Studies were evaluated for quality, and data were extracted for analysis. Meta-analyses were performed using the random-effects model. Results Seventeen studies met the inclusion criteria, including data from 748 pregnant women and 287 infants. More than three of these studies evaluated data of 20 cytokines in maternal serum, and data of 10 cytokines was available from cord blood samples. Only the serum level of CXCL10 was significantly up-regulated in SARS-CoV-2 positive pregnant women (n = 339) compared to SARS-CoV-2 negative pregnant women (n = 409). Subset analysis of maternal samples (n = 183) collected during the acute phase of COVID-19 infection showed elevated CXCL10 and IFN-γ. No significant differences in cytokine levels were found between cord blood samples collected from infants born to mothers with (n = 97) and without (n = 190) COVID-19 during gestation. Subset analysis of cord blood samples collected during the acute phase of maternal infection was limited by insufficient data. The heterogeneity among the studies was substantial. Conclusion The findings suggest that maternal cytokines responses to SARS-CoV-2 infection during pregnancy are not significantly dysregulated, except for CXCL10 and IFN-γ during the acute phase of illness. No evidence of increased cytokine levels in cord blood samples was observed, although this could be impacted by the time period between initial maternal infection and cord blood collection. These results provide some reassurance to parents and healthcare providers but should be interpreted cautiously due to study variations and limitations.
Collapse
Affiliation(s)
- Samhita Jain
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Isabel Elaine Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Dongli Song
- Department of Pediatrics, Division of Neonatology, Santa Clara Valley Medical Center, San Jose, CA, United States
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Xianhua Piao
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Newborn Brain Research Institute, University of California, San Francisco, San Francisco, CA, United States
- Weill Institute for Neuroscience, University of California, San Francisco, San Francisco CA, United States
| |
Collapse
|
38
|
Sawada K, Kamiya S, Kobayashi T. Neonatal Exposure to Lipopolysaccharide Promotes Neurogenesis of Subventricular Zone Progenitors in the Developing Neocortex of Ferrets. Int J Mol Sci 2023; 24:14962. [PMID: 37834410 PMCID: PMC10573966 DOI: 10.3390/ijms241914962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Lipopolysaccharide (LPS) is a natural agonist of toll-like receptor 4 that serves a role in innate immunity. The current study evaluated the LPS-mediated regulation of neurogenesis in the subventricular zone (SVZ) progenitors, that is, the basal radial glia and intermediate progenitors (IPs), in ferrets. Ferret pups were subcutaneously injected with LPS (500 μg/g of body weight) on postnatal days (PDs) 6 and 7. Furthermore, 5-ethynyl-2'-deoxyuridine (EdU) and 5-bromo-2'-deoxyuridine (BrdU) were administered on PDs 5 and 7, respectively, to label the post-proliferative and proliferating cells in the inner SVZ (iSVZ) and outer SVZ (oSVZ). A significantly higher density of BrdU single-labeled proliferating cells was observed in the iSVZ of LPS-exposed ferrets than in controls but not in post-proliferative EdU single-labeled and EdU/BrdU double-labeled self-renewing cells. BrdU single-labeled cells exhibited a lower proportion of Tbr2 immunostaining in LPS-exposed ferrets (22.2%) than in controls (42.6%) and a higher proportion of Ctip2 immunostaining in LPS-exposed ferrets (22.2%) than in controls (8.6%). The present findings revealed that LPS modified the neurogenesis of SVZ progenitors. Neonatal LPS exposure facilitates the proliferation of SVZ progenitors, followed by the differentiation of Tbr2-expressing IPs into Ctip2-expressing immature neurons.
Collapse
Affiliation(s)
- Kazuhiko Sawada
- Department of Nutrition, Faculty of Medical and Health Sciences, Tsukuba International University, Tsuchiura 300-0051, Japan
| | - Shiori Kamiya
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan; (S.K.); (T.K.)
| | - Tetsuya Kobayashi
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan; (S.K.); (T.K.)
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Mughis H, Lye P, Matthews SG, Bloise E. Hypoxia modifies levels of the SARS-CoV-2 cell entry proteins, angiotensin-converting enzyme 2, and furin in fetal human brain endothelial cells. Am J Obstet Gynecol MFM 2023; 5:101126. [PMID: 37562534 DOI: 10.1016/j.ajogmf.2023.101126] [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: 05/31/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND It is not known whether human fetal brain endothelial cells that form the blood-brain barrier express angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin, which are SARS-CoV-2 cell entry proteins. Moreover, it is unclear whether hypoxia, commonly observed during severe maternal COVID-19, can modify their level of expression. We hypothesized that human fetal brain endothelial cells isolated from early- and midpregnancy brain microvessels express angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin. Furthermore, we hypothesized that hypoxia modifies their expression levels in a gestational age- and time-of-exposure-dependent manner. OBJECTIVE This study aimed to investigate whether early- and midpregnancy human fetal brain endothelial cells express angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin SARS-CoV-2-associated cell entry proteins and to determine the effects of hypoxia on angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin expression levels in human fetal brain endothelial cells. STUDY DESIGN This was a prospective study where human fetal brain endothelial cells isolated from early-pregnancy (12.4±0.7 weeks of gestation) and midpregnancy (17.9±0.5 weeks of gestation) fetal brain microvessels (6 per group) were exposed to different oxygen tensions (20%, 5%, and 1% oxygen) for 6, 24, and 48 hours. Angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin messenger RNA and protein levels and localization were assessed using quantitative polymerase chain reaction, Western blot testing, and immunofluorescence. RESULTS Angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin co-localize with the endothelial cell marker von Willebrand factor in human fetal brain endothelial cells isolated from early pregnancy and midpregnancy. In early pregnancy, TMPRSS2 messenger RNA expression was decreased by 5% oxygen compared with 20% oxygen after 6 hours of exposure (P<.05). In midpregnancy, 5% oxygen down-regulated ACE2 messenger RNA compared with 20% oxygen after 24 hours (P<.05). Furin messenger RNA expression was decreased under 5% and 1% oxygen compared with 20% oxygen (P<.05) after 24 hours. In midpregnancy, angiotensin-converting enzyme 2 protein levels were decreased under 5% and 1% oxygen (P<.001) after 24 hours. In contrast, furin protein levels were increased under 1% oxygen compared with 20% oxygen after 24 hours (P<.05). At 48 hours, 1% oxygen increased angiotensin-converting enzyme 2 protein levels compared with 20% oxygen (P<.01). CONCLUSION Hypoxia modifies the expression of selected SARS-CoV-2 cell entry proteins in human fetal brain endothelial cells in a gestational age- and time-of-exposure-dependent manner. As severe COVID-19 may lead to maternal hypoxia, an altered expression of these proteins in the developing human blood-brain barrier could potentially lead to altered SARS-CoV-2 brain invasion and neurologic sequelae in neonates born to pregnancies complicated by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hafsah Mughis
- Department of Physiology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada (Mses Mughis and Lye and Dr Matthews)
| | - Phetcharawan Lye
- Department of Physiology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada (Mses Mughis and Lye and Dr Matthews)
| | - Stephen G Matthews
- Department of Physiology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada (Mses Mughis and Lye and Dr Matthews); Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (Dr Matthews)
| | - Enrrico Bloise
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil (Dr Bloise).
| |
Collapse
|
42
|
Fields NJ, Palmer KR, Nisi A, Marshall SA. Preeclampsia to COVID-19: A journey towards improved placental and vascular function using sulforaphane. Placenta 2023; 141:84-93. [PMID: 37591715 DOI: 10.1016/j.placenta.2023.08.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023]
Abstract
Excess inflammation and oxidative stress are common themes in many pathologies of pregnancy including preeclampsia and more recently severe COVID-19. The risk of preeclampsia increases following maternal infection with COVID-19, potentially relating to significant overlap in pathophysiology with endothelial, vascular and immunological dysfunction common to both. Identifying a therapy which addresses these injurious processes and stabilises the endothelial and vascular maternal system would help address the significant global burden of maternal and neonatal morbidity and mortality they cause. Sulforaphane is a naturally occurring phytonutrient found most densely within cruciferous vegetables. It has anti-inflammatory, antioxidant and immune modulating properties via upregulation of phase-II detoxification enzymes. This review will cover the common pathways shared by COVID-19 and preeclampsia and offer a potential therapeutic target via nuclear factor erythroid 2-related factor upregulation in the form of sulforaphane.
Collapse
Affiliation(s)
- Neville J Fields
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, 27-31 Wright Street, Clayton, Victoria, Australia; Monash Health, Monash Medical Centre, 246 Clayton Road, Clayton, Victoria, Australia.
| | - Kirsten R Palmer
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, 27-31 Wright Street, Clayton, Victoria, Australia; Monash Health, Monash Medical Centre, 246 Clayton Road, Clayton, Victoria, Australia
| | - Anthony Nisi
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, 27-31 Wright Street, Clayton, Victoria, Australia
| | - Sarah A Marshall
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, 27-31 Wright Street, Clayton, Victoria, Australia
| |
Collapse
|
43
|
Fields NJ, Palmer KR, Rolnik DL, Yo J, Nold MF, Giles ML, Krishnaswamy S, Serpa Neto A, Hodges RJ, Marshall SA. CO-Sprout-A Pilot Double-Blinded Placebo-Controlled Randomised Trial of Broccoli Sprout Powder Supplementation for Pregnant Women with COVID-19 on the Duration of COVID-19-Associated Symptoms: Study Protocol. Nutrients 2023; 15:3980. [PMID: 37764764 PMCID: PMC10537772 DOI: 10.3390/nu15183980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Since its discovery in late 2019, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been estimated to be responsible for at least 769.3 million infections and over 6.95 million deaths. Despite significant global vaccination efforts, there are limited therapies that are considered safe and effective for use in the management of COVID-19 during pregnancy despite the common knowledge that pregnant patients have a much higher risk of adverse outcomes. A bioactive compound found in broccoli sprout-sulforaphane-is a potent inducer of phase-II detoxification enzymes promoting a series of potentially beneficial effects notably as an antioxidant, anti-inflammatory, and anti-viral. A pilot, double-blinded, placebo-controlled randomised trial is to be conducted in Melbourne, Australia, across both public and private hospital sectors. We will assess a commercially available broccoli sprout extract in pregnant women between 20+0 and 36+0 weeks gestation with SARS-CoV-2 infection to investigate (i) the duration of COVID-19 associated symptoms, (ii) maternal and neonatal outcomes, and (iii) biomarkers of infection and inflammation. We plan to enrol 60 outpatient women with COVID-19 irrespective of vaccination status diagnosed by PCR swab or RAT (rapid antigen test) within five days and randomised to 14 days of oral broccoli sprout extract (42 mg of sulforaphane daily) or identical microcrystalline cellulose placebo. The primary outcome of this pilot trial will be to assess the feasibility of conducting a larger trial investigating the duration (days) of COVID-19-associated symptoms using a broccoli sprout supplement for COVID-19-affected pregnancies. Pregnant patients remain an at-risk group for severe disease following infection with SARS-CoV-2 and currently unclear consequences for the offspring. Therefore, this study will assess feasibility of using a broccoli sprout supplement, whilst providing important safety data for the use of sulforaphane in pregnancy.
Collapse
Affiliation(s)
- Neville J. Fields
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne 3168, Australia (D.L.R.); (M.F.N.); (M.L.G.); (R.J.H.); (S.A.M.)
- Monash Health, Monash Medical Centre, Melbourne 3168, Australia
| | - Kirsten R. Palmer
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne 3168, Australia (D.L.R.); (M.F.N.); (M.L.G.); (R.J.H.); (S.A.M.)
- Monash Health, Monash Medical Centre, Melbourne 3168, Australia
| | - Daniel L. Rolnik
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne 3168, Australia (D.L.R.); (M.F.N.); (M.L.G.); (R.J.H.); (S.A.M.)
- Monash Health, Monash Medical Centre, Melbourne 3168, Australia
| | - Jennifer Yo
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne 3168, Australia (D.L.R.); (M.F.N.); (M.L.G.); (R.J.H.); (S.A.M.)
- Monash Health, Monash Medical Centre, Melbourne 3168, Australia
| | - Marcel F. Nold
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne 3168, Australia (D.L.R.); (M.F.N.); (M.L.G.); (R.J.H.); (S.A.M.)
- Department of Paediatrics, Monash University, Melbourne 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne 3168, Australia
| | - Michelle L. Giles
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne 3168, Australia (D.L.R.); (M.F.N.); (M.L.G.); (R.J.H.); (S.A.M.)
- Monash Health, Monash Medical Centre, Melbourne 3168, Australia
| | | | - Ary Serpa Neto
- Australian and New Zealand Intensive Care Research Centre (ANZIC-RC), School of Public Health and Preventive Medicine, Melbourne 3004, Australia;
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Austin Hospital, Melbourne 3084, Australia
- Department of Intensive Care, Austin Hospital, Melbourne 3084, Australia
- Department of Critical Care Medicine, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
| | - Ryan J. Hodges
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne 3168, Australia (D.L.R.); (M.F.N.); (M.L.G.); (R.J.H.); (S.A.M.)
- Monash Health, Monash Medical Centre, Melbourne 3168, Australia
| | - Sarah A. Marshall
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne 3168, Australia (D.L.R.); (M.F.N.); (M.L.G.); (R.J.H.); (S.A.M.)
| |
Collapse
|
44
|
Murlanova K, Pletnikov MV. Modeling psychotic disorders: Environment x environment interaction. Neurosci Biobehav Rev 2023; 152:105310. [PMID: 37437753 DOI: 10.1016/j.neubiorev.2023.105310] [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: 12/14/2022] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Schizophrenia is a major psychotic disorder with multifactorial etiology that includes interactions between genetic vulnerability and environmental risk factors. In addition, interplay of multiple environmental adversities affects neurodevelopment and may increase the individual risk of developing schizophrenia. Consistent with the two-hit hypothesis of schizophrenia, we review rodent models that combine maternal immune activation as the first hit with other adverse environmental exposures as the second hit. We discuss the strengths and pitfalls of the current animal models of environment x environment interplay and propose some future directions to advance the field.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
45
|
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
|
46
|
Kowalski K, Misiak B. Schizophrenia and the COVID-19 pandemic: A narrative review from the biomedical perspective. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2023:S1888-9891(23)00015-0. [PMID: 37544807 DOI: 10.1016/j.rpsm.2023.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/29/2023] [Accepted: 04/12/2023] [Indexed: 08/08/2023]
Abstract
The outbreak of the Coronavirus Disease 2019 (COVID-19) pandemic in 2020 caused a rapid worsening of global mental health. Patients with severe mental disorders, including schizophrenia, are at higher risk of being infected. The neuroinvasive potential of the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has been confirmed. The aim of this article was to present a narrative and comprehensive review of multidimensional associations between schizophrenia and COVID-19 with special emphasis on common biological pathways. Online searches were performed in the PubMed database and covered the publication period until September 17, 2022. Search terms included "psychosis", "schizophrenia", "inflammation" and "COVID-19". Viewed as a neuroinflammatory state, schizophrenia shares several neurobiological mechanisms with the COVID-19. Environmental stress, common comorbidities of schizophrenia and adverse effects of antipsychotic treatment are associated with the higher severity and mortality of the COVID-19. Additionally, more frequent relapses of psychosis have been observed, and might be related to lower treatment adherence. In the context of clinical manifestation, higher level of negative symptoms has been identified among patients with schizophrenia during the pandemic. Improvements in mental health care policy and treatment adjustment are necessary to protect people with schizophrenia who are the population that is particularly vulnerable to the consequences of the COVID-19 pandemic. Future research will show if prenatal infection with the SARS-CoV-2 increases a risk of psychosis.
Collapse
Affiliation(s)
- Krzysztof Kowalski
- Department of Psychiatry, Wroclaw Medical University, Pasteura 10 Street, 50-367 Wroclaw, Poland.
| | - Błażej Misiak
- Department of Psychiatry, Wroclaw Medical University, Pasteura 10 Street, 50-367 Wroclaw, Poland
| |
Collapse
|
47
|
Yotova AY, Li LL, O’Leary A, Tegeder I, Reif A, Courtney MJ, Slattery DA, Freudenberg F. Embryonic and adult synaptic proteome perturbations after maternal immune activation: Identification of persistent changes relevant for early intervention. RESEARCH SQUARE 2023:rs.3.rs-3100753. [PMID: 37461513 PMCID: PMC10350178 DOI: 10.21203/rs.3.rs-3100753/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Maternal infections during pregnancy pose an increased risk for neurodevelopmental psychiatric disorders (NPDs) in the offspring. Here, we examined age- and sex-dependent dynamic changes of the hippocampal synaptic proteome after maternal immune activation (MIA) in embryonic and adult mice. Adult male and female MIA offspring exhibited social deficits and sex-specific depression-like behaviours, among others, validating the model. Furthermore, we observed dose-, age-, and sex-dependent synaptic proteome differences. Analysis of the embryonic synaptic proteome implicates sphingolipid and ketoacid metabolism pathway disruptions during neurodevelopment for NPD-pertinent sequelae. In the embryonic hippocampus, prenatal immune activation also led to changes in neuronal guidance, glycosphingolipid metabolism important for signalling and myelination, and post-translational modification of proteins that regulate intercellular interaction and developmental timing. In adulthood, the observed changes in synaptoneurosomes revealed a dynamic shift toward transmembrane trafficking, intracellular signalling cascades, and hormone-mediated metabolism. Importantly, 68 of the proteins with differential abundance in the embryonic brains of MIA offspring were also altered in adulthood, 75% of which retained their directionality. These proteins are involved in synaptic organisation, neurotransmitter receptor regulation, and the vesicle cycle. A cluster of persistently upregulated proteins, including AKT3, PAK1/3, PPP3CA, formed a functional network enriched in the embryonic brain that is involved in cellular responses to environmental stimuli. To infer a link between the overlapping protein alterations and cognitive and psychiatric traits, we probed human phenome-wise association study data for cognitive and psychiatric phenotypes and all, but PORCN were significantly associated with the investigated domains. Our data provide insights into the dynamic effects of an early prenatal immune activation on developing and mature hippocampi and highlights targets for early intervention in individuals exposed to such immune challenges.
Collapse
Affiliation(s)
- Anna Y. Yotova
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
- Goethe University Frankfurt, Faculty of Biological Sciences, Institute of Cell Biology and Neuroscience, Frankfurt, Germany
| | - Li-Li Li
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Turku Brain and Mind Center, University of Turku and Åbo Akademi University, 20014, Turku, Finland
| | - Aet O’Leary
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
- Department of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Irmgard Tegeder
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt, Germany
| | - Andreas Reif
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Michael J Courtney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Turku Brain and Mind Center, University of Turku and Åbo Akademi University, 20014, Turku, Finland
| | - David A. Slattery
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Florian Freudenberg
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
- Goethe University Frankfurt, Faculty of Biological Sciences, Institute of Cell Biology and Neuroscience, Frankfurt, Germany
| |
Collapse
|
48
|
Woods R, Lorusso J, Fletcher J, ElTaher H, McEwan F, Harris I, Kowash H, D'Souza SW, Harte M, Hager R, Glazier JD. Maternal immune activation and role of placenta in the prenatal programming of neurodevelopmental disorders. Neuronal Signal 2023; 7:NS20220064. [PMID: 37332846 PMCID: PMC10273029 DOI: 10.1042/ns20220064] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Maternal infection during pregnancy, leading to maternal immune activation (mIA) and cytokine release, increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. Animal models have provided evidence to support these mechanistic links, with placental inflammatory responses and dysregulation of placental function implicated. This leads to changes in fetal brain cytokine balance and altered epigenetic regulation of key neurodevelopmental pathways. The prenatal timing of such mIA-evoked changes, and the accompanying fetal developmental responses to an altered in utero environment, will determine the scope of the impacts on neurodevelopmental processes. Such dysregulation can impart enduring neuropathological changes, which manifest subsequently in the postnatal period as altered neurodevelopmental behaviours in the offspring. Hence, elucidation of the functional changes that occur at the molecular level in the placenta is vital in improving our understanding of the mechanisms that underlie the pathogenesis of NDDs. This has notable relevance to the recent COVID-19 pandemic, where inflammatory responses in the placenta to SARS-CoV-2 infection during pregnancy and NDDs in early childhood have been reported. This review presents an integrated overview of these collective topics and describes the possible contribution of prenatal programming through placental effects as an underlying mechanism that links to NDD risk, underpinned by altered epigenetic regulation of neurodevelopmental pathways.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jennifer Fletcher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Heidi ElTaher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Francesca McEwan
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Hager M. Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Stephen W. D'Souza
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Michael Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
49
|
Falahi S, Abdoli A, Kenarkoohi A. Maternal COVID-19 infection and the fetus: Immunological and neurological perspectives. New Microbes New Infect 2023; 53:101135. [PMID: 37143853 PMCID: PMC10133021 DOI: 10.1016/j.nmni.2023.101135] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/02/2023] [Accepted: 04/18/2023] [Indexed: 05/06/2023] Open
Abstract
Immunoneuropsychiatry is an emerging field about the interaction between the immune and nervous systems. Infection and infection-related inflammation (in addition to genetics and environmental factors) can act as the etiopathogenesis of neuropsychiatric disorders (NPDs). Exposure to COVID-19 in utero may be a risk factor for developing NPDs in offspring in the future. Maternal immune activation (MIA) and subsequent inflammation can affect fetal brain development. Inflammatory mediators, cytokines, and autoantibodies can pass through the placenta and the compromised blood-brain barrier after MIA, leading to neuroinflammation. Neuroinflammation also affects multiple neurobiological pathways; for example, it decreases the production of the neurotransmitter serotonin. Fetal sex may affect the mother's immune response. Pregnant women with male fetuses have been reported to have decreased maternal and placental humoral responses. This suggests that in pregnancies with a male fetus, fewer antibodies may be transferred to the fetus and contribute to males' increased susceptibility/vulnerability to infectious diseases compared to female infants. Here, we want to discuss maternal COVID-19 infection and its consequences for the fetus, particularly the neurological outcomes and the interaction between fetal sex and possible changes in maternal immune responses.
Collapse
Affiliation(s)
- Shahab Falahi
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Amir Abdoli
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Azra Kenarkoohi
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
- Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
50
|
Alves de Araujo Junior D, Motta F, Fernandes GM, Castro MECD, Sasaki LMP, Luna LP, Rodrigues TS, Kurizky PS, Soares AADSM, Nobrega ODT, Espindola LS, Zaconeta AM, Gomes CM, Martins-Filho OA, de Albuquerque CP, da Mota LMH. Neuroimaging assessment of pediatric cerebral changes associated with SARS-CoV-2 infection during pregnancy. Front Pediatr 2023; 11:1194114. [PMID: 37292371 PMCID: PMC10244818 DOI: 10.3389/fped.2023.1194114] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 04/26/2023] [Indexed: 06/10/2023] Open
Abstract
Background SARS-CoV-2 infection and perinatal neurologic outcomes are still not fully understood. However, there is recent evidence of white matter disease and impaired neurodevelopment in newborns following maternal SARS-CoV-2 infection. These appear to occur as a consequence of both direct viral effects and a systemic inflammatory response, with glial cell/myelin involvement and regional hypoxia/microvascular dysfunction. We sought to characterize the consequences of maternal and fetal inflammatory states in the central nervous system of newborns following maternal SARS-CoV-2 infection. Methods We conducted a longitudinal prospective cohort study from June 2020 to December 2021, with follow-up of newborns born to mothers exposed or not exposed to SARS-CoV-2 infection during pregnancy. Brain analysis included data from cranial ultrasound scans (CUS) with grayscale, Doppler studies (color and spectral), and ultrasound-based brain elastography (shear-wave mode) in specific regions of interest (ROIs): deep white matter, superficial white matter, corpus callosum, basal ganglia, and cortical gray matter. Brain elastography was used to estimate brain parenchymal stiffness, which is an indirect quantifier of cerebral myelin tissue content. Results A total of 219 single-pregnancy children were enrolled, including 201 born to mothers exposed to SARS-CoV-2 infection and 18 from unexposed controls. A neuroimaging evaluation was performed at 6 months of adjusted chronological age and revealed 18 grayscale and 21 Doppler abnormalities. Predominant findings were hyperechogenicity of deep brain white matter and basal ganglia (caudate nuclei/thalamus) and a reduction in the resistance and pulsatility indices of intracranial arterial flow. The anterior brain circulation (middle cerebral and pericallosal arteries) displayed a wider range of flow variation than the posterior circulation (basilar artery). Shear-wave US elastography analysis showed a reduction in stiffness values in the SARS-CoV-2 exposed group in all analyzed regions of interest, especially in the deep white matter elasticity coefficients (3.98 ± 0.62) compared to the control group (7.76 ± 0.77); p-value < 0.001. Conclusion This study further characterizes pediatric structural encephalic changes associated with SARS-CoV-2 infection during pregnancy. The maternal infection has been shown to be related to cerebral deep white matter predominant involvement, with regional hyperechogenicity and reduction of elasticity coefficients, suggesting zonal impairment of myelin content. Morphologic findings may be subtle, and functional studies such as Doppler and elastography may be valuable tools to more accurately identify infants at risk of neurologic damage.
Collapse
Affiliation(s)
- David Alves de Araujo Junior
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
- Department of Medicine, Austin Health, University of Melbourne, Melbourne, VIC, Australia
| | - Felipe Motta
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
| | - Geraldo Magela Fernandes
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
| | - Maria Eduarda Canellas De Castro
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
| | - Lizandra Moura Paravidine Sasaki
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
| | - Licia Pacheco Luna
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | - Patricia Shu Kurizky
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
| | | | | | | | | | - Ciro Martins Gomes
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
| | | | - Cleandro Pires de Albuquerque
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
| | - Licia Maria Henrique da Mota
- Department of Medicine, University of Brasilia (UnB), Brasilia, Brazil
- Hospital Universitario de Brasília (HUB), Brasilia, Brazil
- Medical Sciences, University of Brasilia, Brasilia, Brazil
| |
Collapse
|