1
|
Eleftheriades A, Koulouraki S, Belegrinos A, Eleftheriades M, Pervanidou P. Maternal Obesity and Neurodevelopment of the Offspring. Nutrients 2025; 17:891. [PMID: 40077761 PMCID: PMC11901708 DOI: 10.3390/nu17050891] [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: 02/08/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND An increasing amount of evidence, derived from both human epidemiological studies and animal research, suggests that exposure to maternal obesity in utero is linked to adverse neurodevelopmental outcomes in the offspring. These can include attention deficit hyperactivity disorder, autism spectrum disorders, intellectual disability, and cerebral palsy. METHODS A thorough search in Medline/PubMed and Google Scholar databases was performed by two independent reviewers in order to investigate the link between the exposure to maternal obesity and neurodevelopmental outcomes in the offspring. A list of keywords, including maternal obesity, maternal overweight, maternal diet, neurodevelopment, and neuropsychiatric disorders, was used in the search algorithm. RESULTS The existing evidence regarding the potential mechanisms through which maternal obesity may impact offspring neurodevelopment and programming, such as inflammation, hormone dysregulation, alterations to the microbiome, and epigenetics, as well as evidence from animal studies, was summarized in this narrative review. CONCLUSIONS Maternal obesity seems to be overall associated with various neuropsychiatric and neurodevelopmental disorders. However, more robust data from future studies are needed to establish this association, which will take into account the role of potential confounders such as genetic factors and gene-environment interactions.
Collapse
Affiliation(s)
- Anna Eleftheriades
- Second Department of Obstetrics and Gynaecology, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.E.); (S.K.); (M.E.)
| | - Sevasti Koulouraki
- Second Department of Obstetrics and Gynaecology, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.E.); (S.K.); (M.E.)
| | - Antonios Belegrinos
- Unit of Developmental and Behavioral Paediatrics, First Department of Paediatrics, Agia Sophia Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Makarios Eleftheriades
- Second Department of Obstetrics and Gynaecology, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.E.); (S.K.); (M.E.)
| | - Panagiota Pervanidou
- Unit of Developmental and Behavioral Paediatrics, First Department of Paediatrics, Agia Sophia Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
2
|
Taylor WW, Korobkova L, Bhinderwala N, Dias BG. Towards understanding and halting legacies of trauma. Biol Psychiatry 2025:S0006-3223(25)00108-8. [PMID: 39956254 DOI: 10.1016/j.biopsych.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Echoes of natural and anthropogenic stressors not only reverberate within the physiology, biology, and neurobiology of the generation directly exposed to them but also within the biology of future generations. With the intent of understanding this phenomenon, significant efforts have sought to establish how exposure to psychosocial stress, chemicals, over- and under-nutrition, and chemosensory experiences exert multi-generational influences. From these studies, we are gaining new appreciation for how negative environmental events experienced by one generation impact future generations. This review first outlines the need to operationally define dimensions of negative environmental events in the laboratory and the routes via which the impact of such events are felt through generations. Next, it discusses molecular processes that cause the effects of negative environmental events to be initiated in the exposed generation and then perpetuated across generations. Finally, we discuss how legacies of flourishing can be engineered to halt or reverse multi-generational influences of negative environmental events. In summary, this review synthesizes our current understanding of the concept, causes and consequences of multi-generational echoes of stress and looks to opportunities to halt them.
Collapse
Affiliation(s)
- William Wesley Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA; Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA
| | - Laura Korobkova
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA; Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA
| | - Nabeel Bhinderwala
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA
| | - Brian George Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA; Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA; Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA.
| |
Collapse
|
3
|
Nivins S, Kumar P, Chen X, Gissler M, Lavebratt C. Prenatal exposure to maternal hypertension and higher body mass index and risks of neurodevelopmental and psychiatric disorders during childhood. Acta Obstet Gynecol Scand 2025; 104:319-330. [PMID: 39611242 PMCID: PMC11782093 DOI: 10.1111/aogs.15021] [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: 07/15/2024] [Revised: 10/03/2024] [Accepted: 11/10/2024] [Indexed: 11/30/2024]
Abstract
INTRODUCTION Hypertensive disorders of pregnancy (HDP) or prepregnancy overweight/obesity are independently associated with the risk for certain neurodevelopmental and psychiatric disorders in offspring. These two conditions often co-exist but the risk from combined exposure is unknown. We investigated whether specific subtypes of maternal HDP, along with prepregnancy overweight/obesity, were associated with the distinct risk of neurodevelopmental and psychiatric disorders in offspring during childhood. MATERIAL AND METHODS This prospective, population-based cohort study used data from 652 732 singleton children born alive in Finland between 2004 and 2014 and followed until 2018. The Cox proportional hazards model was used to estimate adjusted hazard ratios (aHR) and 95% confidence intervals (95% CI). RESULTS Children exposed to both chronic hypertension and obesity exhibited a 2.4-3.5-fold higher risk for mood disorders, specific developmental disorder, autism spectrum disorders, and attention-deficit/hyperactivity disorders. Similarly, exposure to both gestational hypertension and overweight increased the risk for anxiety disorders and attention-deficit hyperactivity disorders by 2.4-fold. Meanwhile, combined exposure to preeclampsia and overweight increased the risk of mood and anxiety disorders, specific developmental disorders, and other behavioral disorders, by 1.8-2.2-fold. The effect size of combined exposure to HDP and overweight/obesity was greater than that of the individual exposure to HDP subtypes or overweight/obesity. Furthermore, overweight/obesity synergistically modified these associations between the HDP subtype exposure and offspring mental disorders, except for specific developmental disorders. CONCLUSIONS Our findings suggest that combined exposure to different subtypes of HDP and higher prepregnancy BMI have distinct impacts on the mental health of offspring. Notably, a more pronounced effect was observed in cases where chronic hypertension and obesity coexisted. Future research should focus on exploring dose-related relationships rather than amalgamating maternal HDP for investigating the offspring outcomes.
Collapse
Affiliation(s)
- Samson Nivins
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | - Parvin Kumar
- Center for Molecular MedicineKarolinska University HospitalStockholmSweden
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
| | - Xinxia Chen
- School of Nursing and Rehabilitation, Cheeloo College of MedicineShandong UniversityShandongChina
| | - Mika Gissler
- Center for Molecular MedicineKarolinska University HospitalStockholmSweden
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Department of Data and AnalyticsFinnish Institute for Health and WelfareHelsinkiFinland
| | - Catharina Lavebratt
- Center for Molecular MedicineKarolinska University HospitalStockholmSweden
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
| |
Collapse
|
4
|
Maurer SV, Evans MM, Dukle M, Kundu S, Dennis JL, Ellerbroek RM, Anema SL, Roshko VC, Stevens HE. Threshold effects of prenatal stress on striatal microglia and relevant behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635666. [PMID: 39975105 PMCID: PMC11838387 DOI: 10.1101/2025.01.30.635666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Prenatal stress, a risk factor for neurodevelopmental disorders (NDDs), leads to immune alterations, including offspring neuroimmune cells. Differences in offspring outcomes may arise from whether the extent of prenatal stress crosses "thresholds" for effects on specific outcomes. Therefore, we sought to determine offspring outcomes using models with different extents of prenatal stress. We focused on striatal outcomes, because of their relevance for NDDs. Pregnant CD1 mice were assigned to four groups (each: N=6): no stress ("NoS") or one of the following stressors administered three times daily: i.p. saline injections (low prenatal stress, LoS), Interleukin-6 injections as a component of prenatal stress (immune prenatal stress; ImS), or restraint stress + saline injections (high prenatal stress, HiS), embryonic day 12-18. In adult offspring, HiS altered striatal-dependent behavior across males and females, while ImS induced fewer behavioral changes, and LoS did not affect behavior. Adult striatal microglia morphologies were mostly unchanged across groups, with only HiS leading to altered striatal density of minimally ramified cells. However, embryonic striatal microglia were affected by all models of stress, albeit in distinct ways. The HiS model, and to a lesser extent LoS, also influenced immune components of the maternal-fetal interface: placental macrophages. In conclusion, high and immune stress affected adult striatal-dependent behavior, exceeding the threshold necessary for persistent impacts, but all stress models affected embryonic microglia, suggesting that early neuroimmune outcomes had a lower threshold for impacts. Distinct severities and aspects of prenatal stress may therefore underlie different outcomes relevant to NDDs.
Collapse
|
5
|
Doratt BM, True HE, Sureshchandra S, Qiao Q, Rincon M, Marshall NE, Messaoudi I. The immune landscape of fetal chorionic villous tissue in term placenta. Front Immunol 2025; 15:1506305. [PMID: 39872537 PMCID: PMC11769816 DOI: 10.3389/fimmu.2024.1506305] [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: 10/04/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction The immune compartment within fetal chorionic villi is comprised of fetal Hofbauer cells (HBC) and invading placenta-associated maternal monocytes and macrophages (PAMM). Recent studies have characterized the transcriptional profile of the first trimester (T1) placenta; however, the phenotypic and functional diversity of chorionic villous immune cells at term (T3) remain poorly understood. Methods To address this knowledge gap, immune cells from human chorionic villous tissues obtained from full-term, uncomplicated pregnancies were deeply phenotyped using a combination of flow cytometry, single-cell RNA sequencing (scRNA-seq, CITE-seq) and chromatin accessibility profiling (snATAC-seq). Results Our results indicate that, relative to the first trimester, the frequency of fetal macrophages (HBC, proliferating HBC) is significantly reduced, whereas that of infiltrating maternal monocytes/macrophages (PAMM1b, PAMM1a, PAMM2, MAC_1) increased in T3. PAMM1b and HBCs exhibit the most phagocytic capacity at term highlighting their regulatory role in tissue homeostasis in late pregnancy. The transcriptional profiles of resident villous immune subsets exhibit a heightened activation state relative to the relative to T1, likely to support labor and parturition. Additionally, we provide one of the first insights into the chromatin accessibility profile of villous myeloid cells at term. We next stratified our findings by pre-pregnancy BMI since maternal pregravid obesity is associated with several adverse pregnancy outcomes. Pregravid obesity increased inflammatory gene expression, particularly among HBC and PAMM1a subsets, but dampened the expression of antimicrobial genes, supporting a tolerant-like phenotype of chorionic villous myeloid cells. We report a decline in HBC abundance accompanied by an increase in infiltrating maternal macrophages, which aligns with reports of heightened chorionic villous inflammatory pathologies with pregravid obesity. Finally, given the shared fetal yolk-sac origin of HBCs and microglia, we leveraged an in vitro model of umbilical cord blood-derived microglia to investigate the impact of pregravid obesity on fetal neurodevelopment. Our findings reveal increased expression of activation markers albeit dampened phagocytic capacity in microglia with pregravid obesity. Discussion Overall, our study highlights immune adaptations in the fetal chorionic villous with gestational age and pregravid obesity, as well as insight towards microglia dysfunction possibly underlying poor neurodevelopmental outcomes in offspring of women with pregravid obesity.
Collapse
Affiliation(s)
- Brianna M. Doratt
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - Heather E. True
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, United States
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
| | - Qi Qiao
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
- Department of Biostatistics, University of Kentucky, Lexington, KY, United States
| | - Monica Rincon
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Nicole E. Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
6
|
Guma E, Chakravarty MM. Immune Alterations in the Intrauterine Environment Shape Offspring Brain Development in a Sex-Specific Manner. Biol Psychiatry 2025; 97:12-27. [PMID: 38679357 PMCID: PMC11511788 DOI: 10.1016/j.biopsych.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Exposure to immune dysregulation in utero or in early life has been shown to increase risk for neuropsychiatric illness. The sources of inflammation can be varied, including acute exposures due to maternal infection or acute stress, or persistent exposures due to chronic stress, obesity, malnutrition, or autoimmune diseases. These exposures may cause subtle alteration in brain development, structure, and function that can become progressively magnified across the lifespan, potentially increasing the likelihood of developing a neuropsychiatric conditions. There is some evidence that males are more susceptible to early-life inflammatory challenges than females. In this review, we discuss the various sources of in utero or early-life immune alteration and the known effects on fetal development with a sex-specific lens. To do so, we leveraged neuroimaging, behavioral, cellular, and neurochemical findings. Gaining clarity about how the intrauterine environment affects offspring development is critically important for informing preventive and early intervention measures that may buffer against the effects of these early-life risk factors.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, Maryland; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Peixoto LFF, Sudário LED, Silva MDGCE, Mascarenhas FNADP, Muniz EH, Zanon RG, Ribeiro DL. Maternal hyperglycemia affects cell proliferation signalling and stromal organization in the prostate of neonatal and juvenile rat offspring. Acta Histochem 2024; 126:152193. [PMID: 39244868 DOI: 10.1016/j.acthis.2024.152193] [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: 04/22/2024] [Revised: 07/22/2024] [Accepted: 08/28/2024] [Indexed: 09/10/2024]
Abstract
Gestational diabetes mellitus is a common medical complication during pregnancy. It creates a hyperglycemic environment and impacts offspring development, increasing the risk of long-term complications, including obesity, impaired glucose metabolism and cardiovascular disease. The impact of gestational diabetes on the prostates of adult offspring has already been described; however, it is not known whether these effects are due only to the maternal condition or whether the offspring develop them throughout life. This investigation evaluated the prostates of neonatal and juvenile offspring of hyperglycemic rats due to diabetes. Diabetes was induced with streptozotocin (50 mg/kg, ip) in pregnant Wistar rats and the prostates of 7- or 30-day-old pups from healthy (PC7, PC30) or diabetic (PD7, PD30) mothers were evaluated. We found reduced body weight in pups of PD7 and PD30 and prostate weight in PD30. Prostate branching was not affected, but a reduction in apoptotic levels was associated with impaired acinar bud canalization in neonates. Additionally, PD7 presented reduced ERK1/2 phosphorylation, cell proliferation and collagen, but fibroblasts were increased. In PD30, there was a reduction in the area of the secretory epithelium and stroma, but the luminal area was increased. Moreover, fibroblasts, smooth muscle cells, collagen and metalloproteinase 2 were decreased in these juvenile pups. These data indicate that maternal hyperglycemia inactivates an important cell proliferation signaling pathway in the prostate in the first postnatal days (which is restored in the juvenile period), but it was not sufficient to avoid epithelial and stromal atrophy. This effect on postnatal gland development may impact the reproductive capacity of the prostate in adult life.
Collapse
Affiliation(s)
- Luiz Felipe Fernandes Peixoto
- Department of Cell Biology, Histology and Embriology, Institute of Biomedical Sciences - ICBIM, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Laura Eduarda Dinato Sudário
- Department of Cell Biology, Histology and Embriology, Institute of Biomedical Sciences - ICBIM, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Marina das Graças Carneiro E Silva
- Department of Cell Biology, Histology and Embriology, Institute of Biomedical Sciences - ICBIM, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | - Elusca Helena Muniz
- Department of Cell Biology, Histology and Embriology, Institute of Biomedical Sciences - ICBIM, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Renata Graciele Zanon
- Department of Anatomy, Institute of Biomedical Sciences - ICBIM, Universidade Federal de Uberlândia, Minas Gerais, Brazil
| | - Daniele Lisboa Ribeiro
- Department of Cell Biology, Histology and Embriology, Institute of Biomedical Sciences - ICBIM, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
8
|
Viola MF, Franco Taveras E, Mass E. Developmental programming of tissue-resident macrophages. Front Immunol 2024; 15:1475369. [PMID: 39575254 PMCID: PMC11578957 DOI: 10.3389/fimmu.2024.1475369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Macrophages are integral components of the innate immune system that colonize organs early in development and persist into adulthood through self-renewal. Their fate, whether they are replaced by monocytes or retain their embryonic origin, depends on tissue type and integrity. Macrophages are influenced by their environment, a phenomenon referred to as developmental programming. This influence extends beyond the local tissue microenvironment and includes soluble factors that can reach the macrophage niche. These factors include metabolites, antibodies, growth factors, and cytokines, which may originate from maternal diet, lifestyle, infections, or other developmental triggers and perturbations. These influences can alter macrophage transcriptional, epigenetic, and metabolic profiles, affecting cell-cell communication and tissue integrity. In addition to their crucial role in tissue immunity, macrophages play vital roles in tissue development and homeostasis. Consequently, developmental programming of these long-lived cells can modulate tissue physiology and pathology throughout life. In this review, we discuss the ontogeny of macrophages, the necessity of developmental programming by the niche for macrophage identity and function, and how developmental perturbations can affect the programming of macrophages and their subtissular niches, thereby influencing disease onset and progression in adulthood. Understanding these effects can inform targeted interventions or preventive strategies against diseases. Finally, understanding the consequences of developmental programming will shed light on how maternal health and disease may impact the well-being of future generations.
Collapse
Affiliation(s)
| | | | - Elvira Mass
- Developmental Biology of the Immune System, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
9
|
Ozarslan N, Robinson JF, Buarpung S, Kim MY, Ansbro MR, Akram J, Montoya DJ, Kamya MR, Kakuru A, Dorsey G, Rosenthal PJ, Cheng G, Feeney ME, Fisher SJ, Gaw SL. Gravidity influences distinct transcriptional profiles of maternal and fetal placental macrophages at term. Front Immunol 2024; 15:1384361. [PMID: 38994356 PMCID: PMC11237841 DOI: 10.3389/fimmu.2024.1384361] [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: 02/09/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Introduction Maternal intervillous monocytes (MIMs) and fetal Hofbauer cells (HBCs) are myeloid-derived immune cells at the maternal-fetal interface. Maternal reproductive history is associated with differential risk of pregnancy complications. The molecular phenotypes and roles of these distinct monocyte/macrophage populations and the influence of gravidity on these phenotypes has not been systematically investigated. Methods Here, we used RNA sequencing to study the transcriptional profiles of MIMs and HBCs in normal term pregnancies. Results Our analyses revealed distinct transcriptomes of MIMs and HBCs. Genes involved in differentiation and cell organization pathways were more highly expressed in MIMs vs. HBCs. In contrast, HBCs had higher expression of genes involved in inflammatory responses and cell surface receptor signaling. Maternal gravidity influenced monocyte programming, as expression of pro-inflammatory molecules was significantly higher in MIMs from multigravidae compared to primigravidae. In HBCs, multigravidae displayed enrichment of gene pathways involved in cell-cell signaling and differentiation. Discussion Our results demonstrated that MIMs and HBCs have highly divergent transcriptional signatures, reflecting their distinct origins, locations, functions, and roles in inflammatory responses. Furthermore, maternal gravidity influences the gene signatures of MIMs and HBCs, potentially modulating the interplay between tolerance and trained immunity. The phenomenon of reproductive immune memory may play a novel role in the differential susceptibility of primigravidae to pregnancy complications.
Collapse
Affiliation(s)
- Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Joshua F. Robinson
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - M. Yvonne Kim
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Megan R. Ansbro
- Obstetrics & Gynecology Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Jason Akram
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Dennis J. Montoya
- Department of Molecular, Cellular & Developmental Biology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Biochemistry and Molecular Medicine, University of California Davis Health, Sacramento, CA, United States
| | - Moses R. Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Department of Medicine, Makerere University, Kampala, Uganda
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Grant Dorsey
- Division of HIV, Global Medicine, and Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Philip J. Rosenthal
- Division of HIV, Global Medicine, and Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Genhong Cheng
- Department of Molecular Immunology and Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Margaret E. Feeney
- Division of Experimental Medicine, Department of Medicine and Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Susan J. Fisher
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| |
Collapse
|
10
|
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
|
11
|
Batorsky R, Ceasrine AM, Shook LL, Kislal S, Bordt EA, Devlin BA, Perlis RH, Slonim DK, Bilbo SD, Edlow AG. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. Cell Rep 2024; 43:114326. [PMID: 38848212 PMCID: PMC11808824 DOI: 10.1016/j.celrep.2024.114326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/25/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Maternal immune activation is associated with adverse offspring neurodevelopmental outcomes, many mediated by in utero microglial programming. As microglia remain inaccessible throughout development, identification of noninvasive biomarkers reflecting fetal brain microglial programming could permit screening and intervention. We used lineage tracing to demonstrate the shared ontogeny between fetal brain macrophages (microglia) and fetal placental macrophages (Hofbauer cells) in a mouse model of maternal diet-induced obesity, and single-cell RNA-seq to demonstrate shared transcriptional programs. Comparison with human datasets demonstrated conservation of placental resident macrophage signatures between mice and humans. Single-cell RNA-seq identified common alterations in fetal microglial and Hofbauer cell gene expression induced by maternal obesity, as well as sex differences in these alterations. We propose that Hofbauer cells, which are easily accessible at birth, provide insights into fetal brain microglial programs and may facilitate the early identification of offspring vulnerable to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rebecca Batorsky
- Data Intensive Studies Center, Tufts University, Medford, MA, USA
| | - Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Roy H Perlis
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Department of Neurobiology, Duke University, Durham, NC, USA; Lurie Center for Autism, Massachusetts General Hospital, Boston, MA, USA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
12
|
Merabova N, Ugartemendia L, Edlow AG, Ibarra C, Darbinian N, Tatevosian G, Goetzl L. Maternal obesity: sex-specific in utero changes in fetal brain autophagy and mTOR. Obesity (Silver Spring) 2024; 32:1136-1143. [PMID: 38644654 DOI: 10.1002/oby.24017] [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: 11/13/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 04/23/2024]
Abstract
OBJECTIVE Maternal obesity affects 39.7% of reproductive-age women in the United States. Emerging research has suggested that in utero exposure to maternal obesity is associated with adverse neurodevelopmental outcomes, but knowledge of underlying mechanisms in human samples is lacking. METHODS A matched case-control study was performed in women with singleton fetuses who were undergoing elective pregnancy termination at gestational ages 15 to 21 weeks. Maternal adiponectin levels from plasma were measured using ELISA kits. RNA was extracted from fetal brain tissue using RNeasy Mini Kit (QIAGEN). mRNA expression from ADIPOR1, ADIPOR2, MTOR, ATG5, ATG7, BECN1, and MAP1LC3B was quantified through the ΔΔCt method and using GAPDH as a housekeeping gene. RESULTS We have identified transcription patterns associated with inhibition of autophagy in male fetal brain tissue exposed to maternal obesity (↑MTOR, ↓ATG5, ↓ATG7, and ↓MAP1LC3B), with female fetuses demonstrating either no change in transcription or nonsignificant changes associated with increased autophagy. There was significant downregulation of the autophagy-associated gene BECN1 in both male and female individuals who were exposed to obesity in utero. CONCLUSIONS We present novel evidence suggesting that in utero exposure to maternal obesity in humans may significantly affect neurodevelopment, especially in male fetuses, through alterations in normal autophagy molecular mechanisms and with adiponectin as a potential mediator.
Collapse
Affiliation(s)
- Nana Merabova
- Department of Family Medicine, Medical College of Wisconsin-Prevea, Green Bay, Wisconsin, USA
| | - Lierni Ugartemendia
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, Massachusetts, USA
| | - Claudia Ibarra
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nune Darbinian
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania, USA
| | - Gabriel Tatevosian
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania, USA
| | - Laura Goetzl
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
13
|
Candia AA, Lean SC, Zhang CXW, McKeating DR, Cochrane A, Gulacsi E, Herrera EA, Krause BJ, Sferruzzi-Perri AN. Obesogenic Diet in Mice Leads to Inflammation and Oxidative Stress in the Mother in Association with Sex-Specific Changes in Fetal Development, Inflammatory Markers and Placental Transcriptome. Antioxidants (Basel) 2024; 13:411. [PMID: 38671859 PMCID: PMC11047652 DOI: 10.3390/antiox13040411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Obesity during pregnancy is related to adverse maternal and neonatal outcomes. Factors involved in these outcomes may include increased maternal insulin resistance, inflammation, oxidative stress, and nutrient mishandling. The placenta is the primary determinant of fetal outcomes, and its function can be impacted by maternal obesity. The aim of this study on mice was to determine the effect of obesity on maternal lipid handling, inflammatory and redox state, and placental oxidative stress, inflammatory signaling, and gene expression relative to female and male fetal growth. METHODS Female mice were fed control or obesogenic high-fat/high-sugar diet (HFHS) from 9 weeks prior to, and during, pregnancy. On day 18.5 of pregnancy, maternal plasma, and liver, placenta, and fetal serum were collected to examine the immune and redox states. The placental labyrinth zone (Lz) was dissected for RNA-sequencing analysis of gene expression changes. RESULTS the HFHS diet induced, in the dams, hepatic steatosis, oxidative stress (reduced catalase, elevated protein oxidation) and the activation of pro-inflammatory pathways (p38-MAPK), along with imbalanced circulating cytokine concentrations (increased IL-6 and decreased IL-5 and IL-17A). HFHS fetuses were asymmetrically growth-restricted, showing sex-specific changes in circulating cytokines (GM-CSF, TNF-α, IL-6 and IFN-γ). The morphology of the placenta Lz was modified by an HFHS diet, in association with sex-specific alterations in the expression of genes and proteins implicated in oxidative stress, inflammation, and stress signaling. Placental gene expression changes were comparable to that seen in models of intrauterine inflammation and were related to a transcriptional network involving transcription factors, LYL1 and PLAG1. CONCLUSION This study shows that fetal growth restriction with maternal obesity is related to elevated oxidative stress, inflammatory pathways, and sex-specific placental changes. Our data are important, given the marked consequences and the rising rates of obesity worldwide.
Collapse
Affiliation(s)
- Alejandro A. Candia
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile;
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile;
- Department for the Woman and Newborn Health Promotion, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Samantha C. Lean
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Cindy X. W. Zhang
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Daniel R. McKeating
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Anna Cochrane
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Edina Gulacsi
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Emilio A. Herrera
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile;
| | - Bernardo J. Krause
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile;
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| |
Collapse
|
14
|
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
|
15
|
Williams RP, Lesseur C, Cheng H, Li Q, Deyssenroth M, Molteno CD, Meintjes EM, Jacobson SW, Jacobson JL, Wainwright H, Hao K, Chen J, Carter RC. RNA-seq analysis reveals prenatal alcohol exposure is associated with placental inflammatory cells and gene expression. Gene 2024; 894:147951. [PMID: 37918548 PMCID: PMC11819563 DOI: 10.1016/j.gene.2023.147951] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/21/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Fetal alcohol spectrum disorders (FASD) are the most common preventable cause of birth defects and neurodevelopmental disorders worldwide. The placenta is the crucial interface between mother and fetus. Prenatal alcohol exposure (PAE) has been shown to alter placental structure and expression of genes in bulk placental tissue samples, but prior studies have not examined effects on placental cell-type composition or taken cell-type into consideration in transcriptome analyses. METHODS We leveraged an existent placenta single-cell RNA-seq dataset to perform cell-type deconvolution of bulk placental RNA-seq data from 35 heavy drinking pregnant women and 33 controls in a prospective birth cohort in Cape Town, South Africa. We used bivariate analyses and multivariable adjusted linear regression models to assess the relation of PAE on inferred placental cell-type proportions. We also examined differential expression of inflammatory response genes and PAE, using multivariable adjusted linear models. RESULTS Deconvolution analyses showed heterogeneous placenta cell-type composition in which stromal (27 %), endothelial (26 %) and cytotrophoblasts (18 %) were the predominant cell-types. PAE around conception was associated with a higher proportion of Hofbauer cells (B = 0.51, p = 0.035) in linear models adjusted for maternal age, infant sex, and gestational age. Among the 652 inflammatory genes examined, 35 were differential expressed in alcohol exposed placentas (FDR p < 0.05). CONCLUSIONS Our findings suggest that heavy alcohol exposure during pregnancy can influence the proportion of fetal placental villi macrophages (Hofbauer cells) and increased expression of inflammatory genes. Future studies are needed to further characterize these effects and to assess the potential functional roles of placental inflammation in FASD.
Collapse
Affiliation(s)
- Randy P Williams
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Haoxiang Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qian Li
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maya Deyssenroth
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Christopher D Molteno
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ernesta M Meintjes
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Sandra W Jacobson
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa; Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Joseph L Jacobson
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Helen Wainwright
- Department of Pathology, National Health Laboratory Service, Cape Town, South Africa
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - R Colin Carter
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa; Departments of Emergency Medicine and Pediatrics, Institute of Human Nutrition, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
16
|
Kweon JY, Mun H, Choi MR, Kim HS, Ahn YJ. Maternal obesity induced metabolic disorders in offspring and myeloid reprogramming by epigenetic regulation. Front Endocrinol (Lausanne) 2024; 14:1256075. [PMID: 38292775 PMCID: PMC10824939 DOI: 10.3389/fendo.2023.1256075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/27/2023] [Indexed: 02/01/2024] Open
Abstract
Maternal obesity and gestational diabetes are associated with childhood obesity and increased cardiovascular risk. In this review, we will discuss and summarize extensive clinical and experimental studies that metabolically imbalanced environment exposure in early life plays a critical role in influencing later susceptibility to chronic inflammatory diseases and metabolic syndrome. The effect of maternal obesity and metabolic disorders, including gestational diabetes cause Large-for-gestational-age (LGA) children to link future development of adverse health issues such as obesity, atherosclerosis, hypertension, and non-alcoholic fatty liver disease by immune reprogramming to adverse micro-environment. This review also addresses intrauterine environment-driven myeloid reprogramming by epigenetic regulations and the epigenetic markers as an underlying mechanism. This will facilitate future investigations regarding maternal-to-fetal immune regulation and the epigenetic mechanisms of obesity and cardiovascular diseases.
Collapse
Affiliation(s)
- Joo Young Kweon
- Medical Science and Engineering, Graduate School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hyeonji Mun
- Medical Science and Engineering, Graduate School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Myeong Ryeol Choi
- Medical Science and Engineering, Graduate School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Yong Joo Ahn
- Medical Science and Engineering, Graduate School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department IT Convergence, Pohang University of Science and Technology, Pohang, Republic of Korea
| |
Collapse
|
17
|
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
|
18
|
Batorsky R, Ceasrine AM, Shook LL, Kislal S, Bordt EA, Devlin BA, Perlis RH, Slonim DK, Bilbo SD, Edlow AG. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.16.571680. [PMID: 38187648 PMCID: PMC10769274 DOI: 10.1101/2023.12.16.571680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Maternal immune activation is associated with adverse offspring neurodevelopmental outcomes, many mediated by in utero microglial programming. As microglia remain inaccessible throughout development, identification of noninvasive biomarkers reflecting fetal brain microglial programming could permit screening and intervention. We used lineage tracing to demonstrate the shared ontogeny between fetal brain macrophages (microglia) and fetal placental macrophages (Hofbauer cells) in a mouse model of maternal diet-induced obesity, and single-cell RNA-seq to demonstrate shared transcriptional programs. Comparison with human datasets demonstrated conservation of placental resident macrophage signatures between mice and humans. Single-cell RNA-seq identified common alterations in fetal microglial and Hofbauer cell gene expression induced by maternal obesity, as well as sex differences in these alterations. We propose that Hofbauer cells, which are easily accessible at birth, provide novel insights into fetal brain microglial programs, and may facilitate the early identification of offspring vulnerable to neurodevelopmental disorders in the setting of maternal exposures.
Collapse
Affiliation(s)
| | - Alexis M. Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Lydia L. Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evan A. Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin A. Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Roy H. Perlis
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Donna K. Slonim
- Department of Computer Science, Tufts University, Medford, MA
| | - Staci D. Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, MA
| | - Andrea G. Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Sheng JA, Handa RJ, Tobet SA. Evaluating different models of maternal stress on stress-responsive systems in prepubertal mice. Front Neurosci 2023; 17:1292642. [PMID: 38130695 PMCID: PMC10733493 DOI: 10.3389/fnins.2023.1292642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Maternal adversity during pregnancy influences neurodevelopment in human and model animal offspring. Adversity can result from stressors coming from many different directions ranging from environmental to nutritional and physiological to immune (e.g., infection). Most stressors result in fetal overexposure to glucocorticoids that have been directly linked to long- and short-term negative impacts on neurological health of offspring. Neuropsychiatric diseases postulated to have fetal origins are diverse and include such things cardiovascular disease, obesity, affective disorders, and metabolic and immune disorders. Methods The experiments in the current study compare 3 stressors: prenatal exposure to dexamethasone (DEX), maternal high fat diet (HFD), and maternal caloric restriction (CR). Offspring of mothers with these treatments were examined prepubertally to evaluate stress responsiveness and stress-related behaviors in in male and female mice. Results Prenatal exposure to synthetic glucocorticoid, DEX, resulted in decreased neonatal body weights, reduced social interaction behavior, and hypoactive stress response offspring exposed to maternal DEX. Maternal CR resulted in decreased body weights and social interaction behavior in males and females and increased anxiety-like behavior and acute stress response only in males. HFD resulted in altered body weight gain in both sex offspring with decreased anxiety-like behavior in a female-biased manner. Discussion The idea that glucocorticoid responses to different stressors might serve as a common stimulus across stress paradigms is insufficient, given that different modes of prenatal stress produced differential effects. Opposite nutritional stressors produced similar outcomes for anxiety-like behavior in both sexes, social-like behavior in females, and a hyperactive adrenal stress response in males. One common theme among the three models of maternal stress (DEX, CR, and HFD) was consistent data showing their role in activating the maternal and fetal immune response. By tuning in on the more immediate immunological aspect on the developing fetus (e.g., hormones, cytokines), additional studies may tease out more direct outcomes of maternal stress in rodents and increase their translational value to human studies.
Collapse
Affiliation(s)
- Julietta A. Sheng
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Robert J. Handa
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Stuart A. Tobet
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Psychiatry, Mass General Hospital, Harvard Medical School, Boston, MA, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Innovation Center on Sex Differences in Medicine, Mass General Hospital, Cambridge, MA, United States
| |
Collapse
|
20
|
Volqvartz T, Andersen HHB, Pedersen LH, Larsen A. Obesity in pregnancy-Long-term effects on offspring hypothalamic-pituitary-adrenal axis and associations with placental cortisol metabolism: A systematic review. Eur J Neurosci 2023; 58:4393-4422. [PMID: 37974556 DOI: 10.1111/ejn.16184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Obesity, affecting one in three pregnant women worldwide, is not only a major obstetric risk factor. The resulting low-grade inflammation may have a long-term impact on the offspring's HPA axis through dysregulation of maternal, placental and fetal corticosteroid metabolism, and children born of obese mothers have increased risk of diabetes and cardiovascular disease. The long-term effects of maternal obesity on offspring neurodevelopment are, however, undetermined and could depend on the specific effects on placental and fetal cortisol metabolism. This systematic review evaluates how maternal obesity affects placental cortisol metabolism and the offspring's HPA axis. Pubmed, Embase and Scopus were searched for original studies on maternal BMI, obesity, and cortisol metabolism and transfer. Fifteen studies were included after the screening of 4556 identified records. Studies were small with heterogeneous exposures and outcomes. Two studies found that maternal obesity reduced placental HSD11β2 activity. In one study, umbilical cord blood cortisol levels were affected by maternal BMI. In three studies, an altered cortisol response was consistently seen among offspring in childhood (n = 2) or adulthood (n = 1). Maternal BMI was not associated with placental HSD11β1 or HSD11β2 mRNA expression, or placental HSD11β2 methylation. In conclusion, high maternal BMI is associated with reduced placental HSD11β2 activity and a dampened cortisol level among offspring, but the data is sparse. Further investigations are needed to clarify whether the HPA axis is affected by prenatal factors including maternal obesity and investigate if adverse effects can be ameliorated by optimising the intrauterine environment.
Collapse
Affiliation(s)
- Tabia Volqvartz
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Lars Henning Pedersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Aarhus, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
21
|
Lee JY, Lee HJ, Jang YH, Kim H, Im K, Yang S, Hoh JK, Ahn JH. Maternal pre-pregnancy obesity affects the uncinate fasciculus white matter tract in preterm infants. Front Pediatr 2023; 11:1225960. [PMID: 38034827 PMCID: PMC10684693 DOI: 10.3389/fped.2023.1225960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Background A growing body of evidence suggests an association between a higher maternal pre-pregnancy body mass index (BMI) and adverse long-term neurodevelopmental outcomes for their offspring. Despite recent attention to the effects of maternal obesity on fetal and neonatal brain development, changes in the brain microstructure of preterm infants born to mothers with pre-pregnancy obesity are still not well understood. This study aimed to detect the changes in the brain microstructure of obese mothers in pre-pregnancy and their offspring born as preterm infants using diffusion tensor imaging (DTI). Methods A total of 32 preterm infants (born to 16 mothers with normal BMI and 16 mothers with a high BMI) at <32 weeks of gestation without brain injury underwent brain magnetic resonance imaging at term-equivalent age (TEA). The BMI of all pregnant women was measured within approximately 12 weeks before pregnancy or the first 2 weeks of gestation. We analyzed the brain volume using a morphologically adaptive neonatal tissue segmentation toolbox and calculated the major white matter (WM) tracts using probabilistic maps of the Johns Hopkins University neonatal atlas. We investigated the differences in brain volume and WM microstructure between preterm infants of mothers with normal and high BMI. The DTI parameters were compared among groups using analysis of covariance adjusted for postmenstrual age at scan and multiple comparisons. Results Preterm infants born to mothers with a high BMI showed significantly increased cortical gray matter volume (p = 0.001) and decreased WM volume (p = 0.003) after controlling for postmenstrual age and multiple comparisons. We found a significantly lower axial diffusivity in the uncinate fasciculus (UNC) in mothers with high BMI than that in mothers with normal BMI (1.690 ± 0.066 vs. 1.762 ± 0.101, respectively; p = 0.005). Conclusion Our study is the first to demonstrate that maternal obesity impacts perinatal brain development patterns in preterm infants at TEA, even in the absence of apparent brain injury. These findings provide evidence for the detrimental effects of maternal obesity on brain developmental trajectories in offspring and suggest potential neurodevelopmental outcomes based on an altered UNC WM microstructure, which is known to be critical for language and social-emotional functions.
Collapse
Affiliation(s)
- Joo Young Lee
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Hyun Ju Lee
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Division of Neonatology and Development Medicine, Hanyang University Hospital, Seoul, Republic of Korea
| | - Yong Hun Jang
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Hyuna Kim
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Kiho Im
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
- Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Seung Yang
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Pediatrics, Hanyang University Hospital, Seoul, Republic of Korea
| | - Jeong-Kyu Hoh
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Hanyang University Hospital, Seoul, Republic of Korea
| | - Ja-Hye Ahn
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Division of Neonatology and Development Medicine, Hanyang University Hospital, Seoul, Republic of Korea
| |
Collapse
|
22
|
Chen R, Routh BN, Gaudet AD, Fonken LK. Circadian Regulation of the Neuroimmune Environment Across the Lifespan: From Brain Development to Aging. J Biol Rhythms 2023; 38:419-446. [PMID: 37357738 PMCID: PMC10475217 DOI: 10.1177/07487304231178950] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Circadian clocks confer 24-h periodicity to biological systems, to ultimately maximize energy efficiency and promote survival in a world with regular environmental light cycles. In mammals, circadian rhythms regulate myriad physiological functions, including the immune, endocrine, and central nervous systems. Within the central nervous system, specialized glial cells such as astrocytes and microglia survey and maintain the neuroimmune environment. The contributions of these neuroimmune cells to both homeostatic and pathogenic demands vary greatly across the day. Moreover, the function of these cells changes across the lifespan. In this review, we discuss circadian regulation of the neuroimmune environment across the lifespan, with a focus on microglia and astrocytes. Circadian rhythms emerge in early life concurrent with neuroimmune sculpting of brain circuits and wane late in life alongside increasing immunosenescence and neurodegeneration. Importantly, circadian dysregulation can alter immune function, which may contribute to susceptibility to neurodevelopmental and neurodegenerative diseases. In this review, we highlight circadian neuroimmune interactions across the lifespan and share evidence that circadian dysregulation within the neuroimmune system may be a critical component in human neurodevelopmental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruizhuo Chen
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Brandy N. Routh
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Andrew D. Gaudet
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
- Department of Psychology, The University of Texas at Austin, Austin, Texas
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Laura K. Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
23
|
Ozarslan N, Robinson JF, Buarpung S, Kim MY, Ansbro MR, Akram J, Montoya DJ, Kamya MR, Kakuru A, Dorsey G, Rosenthal PJ, Cheng G, Feeney ME, Fisher SJ, Gaw SL. Distinct transcriptional profiles of maternal and fetal placental macrophages at term are associated with gravidity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559419. [PMID: 37808856 PMCID: PMC10557660 DOI: 10.1101/2023.09.25.559419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Maternal intervillous monocytes (MIMs) and fetal Hofbauer cells (HBCs) are myeloid-derived immune cells at the maternal-fetal interface. Little is known regarding the molecular phenotypes and roles of these distinct monocyte/macrophage populations. Here, we used RNA sequencing to investigate the transcriptional profiles of MIMs and HBCs in six normal term pregnancies. Our analyses revealed distinct transcriptomes of MIMs and HBCs. Genes involved in differentiation and cell organization pathways were more highly expressed in MIMs vs. HBCs. In contrast, HBCs had higher expression of genes involved in inflammatory responses and cell surface receptor signaling. Maternal gravidity influenced monocyte programming, as expression of pro-inflammatory molecules was significantly higher in MIMs from multigravidas compared to primigravidas. In HBCs, multigravidas displayed enrichment of gene pathways involved in cell-cell signaling and differentiation. In summary, our results demonstrated that MIMs and HBCs have highly divergent transcriptional signatures, reflecting their distinct origins, locations, functions, and roles in inflammatory responses. Our data further suggested that maternal gravidity influences the gene signatures of MIMs and HBCs, potentially modulating the interplay between tolerance and trained immunity. The phenomenon of reproductive immune memory may play a novel role in the differential susceptibility of primigravidas to pregnancy complications.
Collapse
|
24
|
Salberg S, Yamakawa GR, Beveridge JK, Noel M, Mychasiuk R. A high-fat high-sugar diet and adversity early in life modulate pain outcomes at the behavioural and molecular level in adolescent rats: The role of sex. Brain Behav Immun 2023; 108:57-79. [PMID: 36403882 DOI: 10.1016/j.bbi.2022.11.006] [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: 06/08/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
Given that adolescence is a significant period of brain plasticity and development, early life factors have the potential to alter long term outcomes. For instance, adversities such as consumption of a high-fat high-sugar (HFHS) diet and adverse childhood experiences (ACEs; e.g., neglect), and their resulting inflammation and microglial activation can influence pain outcomes by priming the neuroimmune system to overrespond to stressors. Chronic pain is highly prevalent amongst the adolescent population, with the prevalence and manifestation being sexually dimorphic. Although clinical studies show that females are twice as likely to report pain problems compared to males, the majority of pre-clinical work uses male rodents. Therefore, our aim was to examine the effects of sex, a HFHS diet, and an ACE on chronic pain outcomes following a stressor in adolescence. Rat dams were randomly assigned to a Standard or HFHS diet, with pups maintained on their respective diets then randomly allocated to a No Stress or ACE paradigm, and a Sham or Injury condition as a stressor. Results showed that early life adversities increased nociceptive sensitivity, inflammation, and microglial activation systemically and within the brain. Behaviourally, pain outcomes were more prominent in females, however the neuroimmune response was exacerbated in males. These results demonstrate the sexual dimorphism of chronic pain outcomes following early life adversities and provide insight into the mechanisms driving these changes, which will inform more targeted and effective treatment strategies for youth living with chronic pain.
Collapse
Affiliation(s)
- Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Jaimie K Beveridge
- Department of Psychology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, Canada
| | - Melanie Noel
- Department of Psychology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Psychology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
25
|
Sureshchandra S, Chan CN, Robino JJ, Parmelee LK, Nash MJ, Wesolowski SR, Pietras EM, Friedman JE, Takahashi D, Shen W, Jiang X, Hennebold JD, Goldman D, Packwood W, Lindner JR, Roberts CT, Burwitz BJ, Messaoudi I, Varlamov O. Maternal Western-style diet remodels the transcriptional landscape of fetal hematopoietic stem and progenitor cells in rhesus macaques. Stem Cell Reports 2022; 17:2595-2609. [PMID: 36332628 PMCID: PMC9768582 DOI: 10.1016/j.stemcr.2022.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022] Open
Abstract
Maternal obesity adversely impacts the in utero metabolic environment, but its effect on fetal hematopoiesis remains incompletely understood. During late development, the fetal bone marrow (FBM) becomes the major site where macrophages and B lymphocytes are produced via differentiation of hematopoietic stem and progenitor cells (HSPCs). Here, we analyzed the transcriptional landscape of FBM HSPCs at single-cell resolution in fetal macaques exposed to a maternal high-fat Western-style diet (WSD) or a low-fat control diet. We demonstrate that maternal WSD induces a proinflammatory response in FBM HSPCs and fetal macrophages. In addition, maternal WSD consumption suppresses the expression of B cell development genes and decreases the frequency of FBM B cells. Finally, maternal WSD leads to poor engraftment of fetal HSPCs in nonlethally irradiated immunodeficient NOD/SCID/IL2rγ-/- mice. Collectively, these data demonstrate for the first time that maternal WSD impairs fetal HSPC differentiation and function in a translationally relevant nonhuman primate model.
Collapse
Affiliation(s)
- Suhas Sureshchandra
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, Institute for Immunology, Center for Virus Research, University of California-Irvine, Irvine, CA 92697, USA
| | - Chi N Chan
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR 97006
| | - Jacob J Robino
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006
| | - Lindsay K Parmelee
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR 97006
| | - Michael J Nash
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephanie R Wesolowski
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Eric M Pietras
- Department of Immunology and Microbiology, Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jacob E Friedman
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Diana Takahashi
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR 97006
| | - Weining Shen
- Department of Statistics, University of California-Irvine, Irvine, CA 92697, USA
| | - Xiwen Jiang
- Department of Statistics, University of California-Irvine, Irvine, CA 92697, USA
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006; Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Devorah Goldman
- Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jonathan R Lindner
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Charles T Roberts
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006; Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Benjamin J Burwitz
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR 97006; Vaccine & Gene Therapy Institute, Beaverton, OR 97006, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, Institute for Immunology, Center for Virus Research, University of California-Irvine, Irvine, CA 92697, USA; Department of Immunology, Microbiology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Oleg Varlamov
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006.
| |
Collapse
|
26
|
Ceasrine AM, Devlin BA, Bolton JL, Green LA, Jo YC, Huynh C, Patrick B, Washington K, Sanchez CL, Joo F, Campos-Salazar AB, Lockshin ER, Kuhn C, Murphy SK, Simmons LA, Bilbo SD. Maternal diet disrupts the placenta-brain axis in a sex-specific manner. Nat Metab 2022; 4:1732-1745. [PMID: 36443520 PMCID: PMC10507630 DOI: 10.1038/s42255-022-00693-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022]
Abstract
High maternal weight is associated with detrimental outcomes in offspring, including increased susceptibility to neurological disorders such as anxiety, depression and communicative disorders. Despite widespread acknowledgement of sex biases in the development of these disorders, few studies have investigated potential sex-biased mechanisms underlying disorder susceptibility. Here, we show that a maternal high-fat diet causes endotoxin accumulation in fetal tissue, and subsequent perinatal inflammation contributes to sex-specific behavioural outcomes in offspring. In male offspring exposed to a maternal high-fat diet, increased macrophage Toll-like receptor 4 signalling results in excess microglial phagocytosis of serotonin (5-HT) neurons in the developing dorsal raphe nucleus, decreasing 5-HT bioavailability in the fetal and adult brains. Bulk sequencing from a large cohort of matched first-trimester human samples reveals sex-specific transcriptome-wide changes in placental and brain tissue in response to maternal triglyceride accumulation (a proxy for dietary fat content). Further, fetal brain 5-HT levels decrease as placental triglycerides increase in male mice and male human samples. These findings uncover a microglia-dependent mechanism through which maternal diet can impact offspring susceptibility for neuropsychiatric disorder development in a sex-specific manner.
Collapse
Affiliation(s)
- Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Jessica L Bolton
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Lauren A Green
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Young Chan Jo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Carolyn Huynh
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Bailey Patrick
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Kamryn Washington
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Cristina L Sanchez
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Faith Joo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | | | - Elana R Lockshin
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Cynthia Kuhn
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Leigh Ann Simmons
- Department of Human Ecology, Perinatal Origins of Disparities Center, University of California, Davis, Davis, CA, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
- Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
27
|
Shook LL, Fourman LT, Edlow AG. Immune Responses to SARS-CoV-2 in Pregnancy: Implications for the Health of the Next Generation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1465-1473. [PMID: 36192115 PMCID: PMC9536183 DOI: 10.4049/jimmunol.2200414] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/08/2022] [Indexed: 11/06/2022]
Abstract
Widespread SARS-CoV-2 infection among pregnant individuals has led to a generation of fetuses exposed in utero, but the long-term impact of such exposure remains unknown. Although fetal infection is rare, children born to mothers with SARS-CoV-2 infection may be at increased risk for adverse neurodevelopmental and cardiometabolic outcomes. Fetal programming effects are likely to be mediated at least in part by maternal immune activation. In this review, we discuss recent evidence regarding the effects of prenatal SARS-CoV-2 infection on the maternal, placental, and fetal immune response, as well as the implications for the long-term health of offspring. Extrapolating from what is known about the impact of maternal immune activation in other contexts (e.g., obesity, HIV, influenza), we review the potential for neurodevelopmental and cardiometabolic morbidity in offspring. Based on available data suggesting potential increased neurodevelopmental risk, we highlight the importance of establishing large cohorts to monitor offspring born to SARS-CoV-2-positive mothers for neurodevelopmental and cardiometabolic sequelae.
Collapse
Affiliation(s)
- Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA; and
| | - Lindsay T Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA;
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA; and
| |
Collapse
|
28
|
Denizli M, Capitano ML, Kua KL. Maternal obesity and the impact of associated early-life inflammation on long-term health of offspring. Front Cell Infect Microbiol 2022; 12:940937. [PMID: 36189369 PMCID: PMC9523142 DOI: 10.3389/fcimb.2022.940937] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
The prevalence of obesity is increasingly common in the United States, with ~25% of women of reproductive age being overweight or obese. Metaflammation, a chronic low grade inflammatory state caused by altered metabolism, is often present in pregnancies complicated by obesity. As a result, the fetuses of mothers who are obese are exposed to an in-utero environment that has altered nutrients and cytokines. Notably, both human and preclinical studies have shown that children born to mothers with obesity have higher risks of developing chronic illnesses affecting various organ systems. In this review, the authors sought to present the role of cytokines and inflammation during healthy pregnancy and determine how maternal obesity changes the inflammatory landscape of the mother, leading to fetal reprogramming. Next, the negative long-term impact on offspring’s health in numerous disease contexts, including offspring’s risk of developing neuropsychiatric disorders (autism, attention deficit and hyperactive disorder), metabolic diseases (obesity, type 2 diabetes), atopy, and malignancies will be discussed along with the potential of altered immune/inflammatory status in offspring as a contributor of these diseases. Finally, the authors will list critical knowledge gaps in the field of developmental programming of health and diseases in the context of offspring of mothers with obesity, particularly the understudied role of hematopoietic stem and progenitor cells.
Collapse
Affiliation(s)
- Merve Denizli
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis IN, United States
| | - Maegan L. Capitano
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis IN, United States
| | - Kok Lim Kua
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis IN, United States
- *Correspondence: Kok Lim Kua,
| |
Collapse
|
29
|
Cirulli F, De Simone R, Musillo C, Ajmone-Cat MA, Berry A. Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies. Nutrients 2022; 14:nu14153150. [PMID: 35956326 PMCID: PMC9370669 DOI: 10.3390/nu14153150] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/18/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a main risk factor for the onset and the precipitation of many non-communicable diseases. This condition, which is associated with low-grade chronic systemic inflammation, is of main concern during pregnancy leading to very serious consequences for the new generations. In addition to the prominent role played by the adipose tissue, dysbiosis of the maternal gut may also sustain the obesity-related inflammatory milieu contributing to create an overall suboptimal intrauterine environment. Such a condition here generically defined as “inflamed womb” may hold long-term detrimental effects on fetal brain development, increasing the vulnerability to mental disorders. In this review, we will examine the hypothesis that maternal obesity-related gut dysbiosis and the associated inflammation might specifically target fetal brain microglia, the resident brain immune macrophages, altering neurodevelopmental trajectories in a sex-dependent fashion. We will also review some of the most promising nutritional strategies capable to prevent or counteract the effects of maternal obesity through the modulation of inflammation and oxidative stress or by targeting the maternal microbiota.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- Correspondence: (F.C.); (A.B.)
| | - Roberta De Simone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.D.S.); (M.A.A.-C.)
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- PhD Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Antonietta Ajmone-Cat
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.D.S.); (M.A.A.-C.)
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- Correspondence: (F.C.); (A.B.)
| |
Collapse
|
30
|
Urbonaite G, Knyzeliene A, Bunn FS, Smalskys A, Neniskyte U. The impact of maternal high-fat diet on offspring neurodevelopment. Front Neurosci 2022; 16:909762. [PMID: 35937892 PMCID: PMC9354026 DOI: 10.3389/fnins.2022.909762] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022] Open
Abstract
A maternal high-fat diet affects offspring neurodevelopment with long-term consequences on their brain health and behavior. During the past three decades, obesity has rapidly increased in the whole human population worldwide, including women of reproductive age. It is known that maternal obesity caused by a high-fat diet may lead to neurodevelopmental disorders in their offspring, such as autism spectrum disorder, attention deficit hyperactivity disorder, anxiety, depression, and schizophrenia. A maternal high-fat diet can affect offspring neurodevelopment due to inflammatory activation of the maternal gut, adipose tissue, and placenta, mirrored by increased levels of pro-inflammatory cytokines in both maternal and fetal circulation. Furthermore, a maternal high fat diet causes gut microbial dysbiosis further contributing to increased inflammatory milieu during pregnancy and lactation, thus disturbing both prenatal and postnatal neurodevelopment of the offspring. In addition, global molecular and cellular changes in the offspring's brain may occur due to epigenetic modifications including the downregulation of brain-derived neurotrophic factor (BDNF) expression and the activation of the endocannabinoid system. These neurodevelopmental aberrations are reflected in behavioral deficits observed in animals, corresponding to behavioral phenotypes of certain neurodevelopmental disorders in humans. Here we reviewed recent findings from rodent models and from human studies to reveal potential mechanisms by which a maternal high-fat diet interferes with the neurodevelopment of the offspring.
Collapse
Affiliation(s)
- Gintare Urbonaite
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agne Knyzeliene
- Centre for Cardiovascular Science, The Queen’s Medical Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Fanny Sophia Bunn
- Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Adomas Smalskys
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Urte Neniskyte
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- VU LSC-EMBL Partnership for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
31
|
Khatib N, Ginsberg Y, Ben David C, Ross MG, Vitner D, Zipori Y, Zamora O, Weiner Z, Beloosesky R. Magnesium sulphate neuroprotection mechanism is placental mediated by inhibition of inflammation, apoptosis and oxidative stress. Placenta 2022; 127:29-36. [DOI: 10.1016/j.placenta.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 07/03/2022] [Accepted: 07/14/2022] [Indexed: 10/17/2022]
|
32
|
Ruyak SL, Noor S, DiDomenico J, Sun MS, Fernandez Oropeza AK, Rodriguez DE, Marquez LE, Milligan ED, Bakhireva LN. Effects of prenatal opioid and alcohol exposures on immune and serotonin factors in human placenta. Exp Neurol 2022; 353:114057. [PMID: 35364108 PMCID: PMC10035581 DOI: 10.1016/j.expneurol.2022.114057] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Opioids and alcohol impact critical serotonin (5-HT) function in the developing placenta and fetus through the actions of immune proinflammatory factors. Yet, possible convergent effects of opioids and alcohol on human placental toll-like receptor 4 (TLR4) activation and subsequent 5-HT homeostasis remain entirely unknown. The purpose of this study was to examine the effect of prenatal exposure to opioids with or without prenatal alcohol exposure (PAE) on the expression of key placental immune and serotonin signaling factors in human placental tissue obtained from a well-characterized prospective cohort. METHODS Data were collected from a subset of participants enrolled in the prospective pre-birth Ethanol, Neurodevelopment, Infant, and Child Health (ENRICH-1) cohort. Women were recruited and classified into four study groups: 1) PAE (n = 20); 2) those taking medications for opioid use disorder (MOUD; n = 28), 3) concurrent PAE and MOUD (n = 20); and 4) controls (HC; n = 20) based on prospective, repeated self-report, and biomarker analysis. Placenta samples underwent tissue processing to identify mRNA for TLR4, nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), serotonin transporter (SERT), tryptophan hydroxylase (TPH1), indoleamine 2,3-Dioxygenase 1 (IDO) as well as protein concentrations of TLR4, IL-1β, TNF-α, SERT. To consider the association between study group and mRNA/protein expression of our targets, multivariable regression models were developed with inclusion of a priori selected covariates. RESULTS There was a significant negative association between PAE and SERT mRNA (β = -0.01; p < 0.01) and a positive association with TPH1 mRNA expression (β = 0.78; p < 0.05). In addition, there was a negative association between MOUD and TNF-α protein expression (β = -0.12; p < 0.05). CONCLUSIONS This study provides the first evidence that PAE may inhibit SERT expression while simultaneously promoting increased TPH1 protein expression in human placenta. This may result in increased 5-HT in fetal circulation known to affect neurodevelopment. Our data suggest that opioids and alcohol may disturb the bidirectional, dynamic interaction between the placental immune and serotonin system. Given the implication for brain development and health across the life-span further investigation of these critical mechanisms in well-defined cohorts is required.
Collapse
Affiliation(s)
- Sharon L Ruyak
- College of Nursing, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America; College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America.
| | - Shahani Noor
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States of America
| | - Jared DiDomenico
- College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Melody S Sun
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States of America
| | - Annette K Fernandez Oropeza
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States of America
| | - Dominique E Rodriguez
- College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Lidia Enriquez Marquez
- College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Erin D Milligan
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States of America
| | - Ludmila N Bakhireva
- College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| |
Collapse
|
33
|
Musillo C, Berry A, Cirulli F. Prenatal psychological or metabolic stress increases the risk for psychiatric disorders: the "funnel effect" model. Neurosci Biobehav Rev 2022; 136:104624. [PMID: 35304226 DOI: 10.1016/j.neubiorev.2022.104624] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 12/16/2022]
Abstract
Adverse stressful experiences in utero can redirect fetal brain development, ultimately leading to increased risk for psychiatric disorders. Obesity during pregnancy can have similar effects as maternal stress, affecting mental health in the offspring. In order to explain how similar outcomes may originate from different prenatal conditions, we propose a "funnel effect" model whereby maternal psychological or metabolic stress triggers the same evolutionarily conserved response pathways, increasing vulnerability for psychopathology. In this context, the placenta, which is the main mother-fetus interface, appears to facilitate such convergence, re-directing "stress" signals to the fetus. Characterizing converging pathways activated by different adverse environmental conditions is fundamental to assess the emergence of risk signatures of major psychiatric disorders, which might enable preventive measures in risk populations, and open up new diagnostics, and potentially therapeutic approaches for disease prevention and health promotion already during pregnancy.
Collapse
Affiliation(s)
- Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; PhD Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
34
|
True H, Blanton M, Sureshchandra S, Messaoudi I. Monocytes and macrophages in pregnancy: The good, the bad, and the ugly. Immunol Rev 2022; 308:77-92. [PMID: 35451089 DOI: 10.1111/imr.13080] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022]
Abstract
A successful human pregnancy requires precisely timed adaptations by the maternal immune system to support fetal growth while simultaneously protecting mother and fetus against microbial challenges. The first trimester of pregnancy is characterized by a robust increase in innate immune activity that promotes successful implantation of the blastocyst and placental development. Moreover, early pregnancy is also a state of increased vulnerability to vertically transmitted pathogens notably, human immunodeficiency virus (HIV), Zika virus (ZIKV), SARS-CoV-2, and Listeria monocytogenes. As gestation progresses, the second trimester is marked by the establishment of an immunosuppressive environment that promotes fetal tolerance and growth while preventing preterm birth, spontaneous abortion, and other gestational complications. Finally, the period leading up to labor and parturition is characterized by the reinstatement of an inflammatory milieu triggering childbirth. These dynamic waves of carefully orchestrated changes have been dubbed the "immune clock of pregnancy." Monocytes in maternal circulation and tissue-resident macrophages at the maternal-fetal interface play a critical role in this delicate balance. This review will summarize the current data describing the longitudinal changes in the phenotype and function of monocyte and macrophage populations in healthy and complicated pregnancies.
Collapse
Affiliation(s)
- Heather True
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA.,Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Madison Blanton
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA.,Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | | | - Ilhem Messaoudi
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
35
|
Magalhaes MS, Potter HG, Ahlback A, Gentek R. Developmental programming of macrophages by early life adversity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:213-259. [PMID: 35636928 DOI: 10.1016/bs.ircmb.2022.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Macrophages are central elements of all organs, where they have a multitude of physiological and pathological functions. The first macrophages are produced during fetal development, and most adult organs retain populations of fetal-derived macrophages that self-maintain without major input of hematopoietic stem cell-derived monocytes. Their developmental origins make macrophages highly susceptible to environmental perturbations experienced in early life, in particular the fetal period. It is now well recognized that such adverse developmental conditions contribute to a wide range of diseases later in life. This chapter explores the notion that macrophages are key targets of environmental adversities during development, and mediators of their long-term impact on health and disease. We first briefly summarize our current understanding of macrophage ontogeny and their biology in tissues and consider potential mechanisms by which environmental stressors may mediate fetal programming. We then review evidence for programming of macrophages by adversities ranging from maternal immune activation and diet to environmental pollutants and toxins, which have disease relevance for different organ systems. Throughout this chapter, we contemplate appropriate experimental strategies to study macrophage programming. We conclude by discussing how our current knowledge of macrophage programming could be conceptualized, and finally highlight open questions in the field and approaches to address them.
Collapse
Affiliation(s)
- Marlene S Magalhaes
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Harry G Potter
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Ahlback
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
36
|
Ceasrine AM, Bilbo SD. Dietary fat: a potent microglial influencer. Trends Endocrinol Metab 2022; 33:196-205. [PMID: 35078706 PMCID: PMC8881786 DOI: 10.1016/j.tem.2021.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 12/25/2022]
Abstract
Poor nutrition, lack of exercise, and genetic predisposition all contribute to the growing epidemic of obesity. Overweight/obesity create an environment of chronic inflammation that leads to negative physiological and neurological outcomes, such as diabetes, cardiovascular disease, and anxiety/depression. While the whole body contributes to metabolic homeostasis, the neuroimmune system has recently emerged as a key regulator of metabolism. Microglia, the resident immune cells of the brain, respond both directly and indirectly to dietary fat, and the environment in which microglia develop contributes to their responsiveness later in life. Thus, high maternal weight during pregnancy may have consequences for microglial function in offspring. Here, we discuss the most recent findings on microglia signaling in overweight/obesity with a focus on perinatal programming.
Collapse
Affiliation(s)
- Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27710, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27710, USA; Department of Neurobiology, Duke University, Durham, NC 27710, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
37
|
Han VX, Jones HF, Patel S, Mohammad SS, Hofer MJ, Alshammery S, Maple-Brown E, Gold W, Brilot F, Dale RC. Emerging evidence of Toll-like receptors as a putative pathway linking maternal inflammation and neurodevelopmental disorders in human offspring: A systematic review. Brain Behav Immun 2022; 99:91-105. [PMID: 34562595 DOI: 10.1016/j.bbi.2021.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/21/2021] [Accepted: 09/18/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation is increasingly recognised to play a major role in gene-environment interactions in neurodevelopmental disorders (NDDs). The effects of aberrant immune responses to environmental stimuli in the mother and in the child can affect neuroimmune signalling that is central to brain development. Toll-like receptors (TLR) are the best known innate immune pattern and danger recognition sensors to various environmental threats. In animal models, maternal immune activation (MIA), secondary to inflammatory factors including maternal gestational infection, obesity, diabetes, and stress activate the TLR pathway in maternal blood, placenta, and fetal brain, which correlate with offspring neurobehavioral abnormalities. Given the central role of TLR activation in animal MIA models, we systematically reviewed the human evidence for TLR activation and response to stimulation across the maternal-fetal interface. Firstly, we included 59 TLR studies performed in peripheral blood of adults in general population (outside of pregnancy) with six chronic inflammatory factors which have epidemiological evidence for increased risk of offspring NDDs, namely, obesity, diabetes mellitus, depression, low socio-economic status, autoimmune diseases, and asthma. Secondly, eight TLR studies done in human pregnancies with chronic inflammatory factors, involving maternal blood, placenta, and cord blood, were reviewed. Lastly, ten TLR studies performed in peripheral blood of individuals with NDDs were included. Despite these studies, there were no studies which examined TLR function in both the pregnant mother and their offspring. Increased TLR2 and TLR4 mRNA and/or protein levels in peripheral blood were common in obesity, diabetes mellitus, depression, autoimmune thyroid disease, and rheumatoid arthritis. To a lesser degree, TLR 3, 7, 8, and 9 activation were found in peripheral blood of humans with autoimmune diseases and depression. In pregnancy, increased TLR4 mRNA levels were found in the peripheral blood of women with diabetes mellitus and systemic lupus erythematosus. Placental TLR activation was found in mothers with obesity or diabetes. Postnatally, dysregulated TLR response to stimulation was found in peripheral blood of individuals with NDDs. This systematic review found emerging evidence that TLR activation may represent a mechanistic link between maternal inflammation and offspring NDD, however the literature is incomplete and longitudinal outcome studies are lacking. Identification of pathogenic mechanisms in MIA could create preventive and therapeutic opportunities to mitigate NDD prevalence and severity.
Collapse
Affiliation(s)
- Velda X Han
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; Khoo-Teck Puat-National University Children's Medical Institute, National University Health System, Singapore; School of Medical Sciences, The University of Sydney, Sydney, Australia
| | - Hannah F Jones
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; Department of Neuroservices, Starship Children's Hospital, Auckland, New Zealand
| | - Shrujna Patel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Shekeeb S Mohammad
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Markus J Hofer
- School of Life and Environmental Sciences and Charles Perkins Centre, The University of Sydney, Sydney, Australia; The Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Sarah Alshammery
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; School of Medical Sciences, The University of Sydney, Sydney, Australia; Molecular Neurobiology Research Laboratory, Kids Research, Children's Hospital at Westmead, and The Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Emma Maple-Brown
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; School of Medical Sciences, The University of Sydney, Sydney, Australia; Molecular Neurobiology Research Laboratory, Kids Research, Children's Hospital at Westmead, and The Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Wendy Gold
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; Molecular Neurobiology Research Laboratory, Kids Research, Children's Hospital at Westmead, and The Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Fabienne Brilot
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; School of Medical Sciences, The University of Sydney, Sydney, Australia; The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; Molecular Neurobiology Research Laboratory, Kids Research, Children's Hospital at Westmead, and The Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Russell C Dale
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; The Brain and Mind Centre, The University of Sydney, Sydney, Australia.
| |
Collapse
|
38
|
Waleczek FJG, Sansonetti M, Xiao K, Jung M, Mitzka S, Dendorfer A, Weber N, Perbellini F, Thum T. Chemical and mechanical activation of resident cardiac macrophages in the living myocardial slice ex vivo model. Basic Res Cardiol 2022; 117:63. [PMID: 36449104 PMCID: PMC9712328 DOI: 10.1007/s00395-022-00971-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022]
Abstract
Resident cardiac macrophages (rcMACs) are among the most abundant immune cells in the heart. Plasticity and activation are hallmarks of rcMACs in response to changes in the microenvironment, which is essential for in vitro experimentation. The in vivo investigation is confounded by the infiltration of other cells hindering direct studies of rcMACs. As a tool to investigate rcMACs, we applied the ex vivo model of living myocardial slices (LMS). LMS are ultrathin ex vivo multicellular cardiac preparations in which the circulatory network is interrupted. The absence of infiltration in this model enables the investigation of the rcMACs response to immunomodulatory and mechanical stimulations. Such conditions were generated by applying interferon-gamma (IFN-γ) or interleukine-4 (IL-4) and altering the preload of cultured LMS, respectively. The immunomodulatory stimulation of the LMS induced alterations of the gene expression pattern without affecting tissue contractility. Following 24 h culture, low input RNA sequencing of rcMACs isolated from LMS was used for gene ontology analysis. Reducing the tissue stretch (unloading) of LMS altered the gene ontology clusters of isolated rcMACs with intermediate semantic similarity to IFN-γ triggered reaction. Through the overlap of genes affected by IFN-γ and unloading, we identified Allograft inflammatory factor 1 (AIF-1) as a potential marker gene for inflammation of rcMACs as significantly altered in whole immunomodulated LMS. MicroRNAs associated with the transcriptomic changes of rcMACs in unloaded LMS were identified in silico. Here, we demonstrate the approach of LMS to understand load-triggered cardiac inflammation and, thus, identify potential translationally important therapeutic targets.
Collapse
Affiliation(s)
- F. J. G. Waleczek
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - M. Sansonetti
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - K. Xiao
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany ,grid.4561.60000 0000 9261 3939Fraunhofer Institute ITEM, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - M. Jung
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - S. Mitzka
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany ,grid.4561.60000 0000 9261 3939Fraunhofer Institute ITEM, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - A. Dendorfer
- grid.5252.00000 0004 1936 973XWalter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University München, Marchioninistraße 27, 81377 Munich, Germany
| | - N. Weber
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - F. Perbellini
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - T. Thum
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany ,grid.4561.60000 0000 9261 3939Fraunhofer Institute ITEM, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
39
|
Smith BL. Improving translational relevance: The need for combined exposure models for studying prenatal adversity. Brain Behav Immun Health 2021; 16:100294. [PMID: 34589787 PMCID: PMC8474200 DOI: 10.1016/j.bbih.2021.100294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Prenatal environmental adversity is a risk factor for neurodevelopmental disorders (NDDs), with the neuroimmune environment proposed to play a role in this risk. Adverse maternal exposures are associated with cognitive consequences in the offspring that are characteristics of NDDs and simultaneous neuroimmune changes that may underlie NDD risk. In both animal models and human studies the association between prenatal environmental exposure and NDD risk has been shown to be complex. Maternal overnutrition/obesity and opioid use are two different examples of complex exposure epidemics, each with their own unique comorbidities. This review will examine maternal obesity and maternal opioid use separately, illustrating the pervasive comorbidities with each exposure to argue a need for animal models of compound prenatal exposures. Many of these comorbidities can impact neuroimmune function, warranting systematic investigation of combined exposures to begin to understand this complexity. While traditional approaches in animal models have focused on modeling a single prenatal exposure or second exposure later in life, a translational approach would begin to incorporate the most prevalent co-occurring prenatal exposures. Long term follow-up in humans is extremely challenging, so animal models can provide timely insight into neurodevelopmental consequences of complex prenatal exposures. Animal models that represent this translational context of comorbid exposures behind maternal obesity or comorbid exposures behind maternal opioid use may reveal potential synergistic neuroimmune interactions that contribute to cognitive consequences and NDD risk. Finally, translational co-exposure models can identify concerning exposure combinations to guide treatment in complex cases, and identify high risk children starting in the prenatal period where early interventions improve prognosis.
Collapse
Affiliation(s)
- Brittany L. Smith
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
40
|
Rajasilta O, Häkkinen S, Björnsdotter M, Scheinin NM, Lehtola SJ, Saunavaara J, Parkkola R, Lähdesmäki T, Karlsson L, Karlsson H, Tuulari JJ. Maternal pre-pregnancy BMI associates with neonate local and distal functional connectivity of the left superior frontal gyrus. Sci Rep 2021; 11:19182. [PMID: 34584134 PMCID: PMC8478954 DOI: 10.1038/s41598-021-98574-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 09/06/2021] [Indexed: 11/09/2022] Open
Abstract
Maternal obesity/overweight during pregnancy has reached epidemic proportions and has been linked with adverse outcomes for the offspring, including cognitive impairment and increased risk for neuropsychiatric disorders. Prior neuroimaging investigations have reported widespread aberrant functional connectivity and white matter tract abnormalities in neonates born to obese mothers. Here we explored whether maternal pre-pregnancy adiposity is associated with alterations in local neuronal synchrony and distal connectivity in the neonate brain. 21 healthy mother-neonate dyads from uncomplicated pregnancies were included in this study (age at scanning 26.14 ± 6.28 days, 12 male). The neonates were scanned with a 6-min resting-state functional magnetic resonance imaging (rs-fMRI) during natural sleep. Regional homogeneity (ReHo) maps were computed from obtained rs-fMRI data. Multiple regression analysis was performed to assess the association of pre-pregnancy maternal body-mass-index (BMI) and ReHo. Seed-based connectivity analysis with multiple regression was subsequently performed with seed-ROI derived from ReHo analysis. Maternal adiposity measured by pre-pregnancy BMI was positively associated with neonate ReHo values within the left superior frontal gyrus (SFG) (FWE-corrected p < 0.005). Additionally, we found both positive and negative associations (p < 0.05, FWE-corrected) for maternal pre-pregnancy BMI and seed-based connectivity between left SFG and prefrontal, amygdalae, basal ganglia and insular regions. Our results imply that maternal pre-pregnancy BMI associates with local and distal functional connectivity within the neonate left superior frontal gyrus. These findings add to the evidence that increased maternal pre-pregnancy BMI has a programming influence on the developing neonate brain functional networks.
Collapse
Affiliation(s)
- Olli Rajasilta
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.
| | - Suvi Häkkinen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.,Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Malin Björnsdotter
- Department of Psychiatry for Affective Disorders, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Noora M Scheinin
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.,Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | - Satu J Lehtola
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland
| | - Jani Saunavaara
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Tuire Lähdesmäki
- Department of Pediatric Neurology, Turku University Hospital and University of Turku, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.,Center for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.,Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland.,Center for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Jetro J Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.,Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland.,Department of Psychiatry, University of Oxford, UK (Sigrid Juselius Fellowship), Oxford, UK.,Turku Collegium for Science, Medicine and Technology, University of Turku, Turku, Finland
| |
Collapse
|
41
|
Coley EJL, Hsiao EY. Malnutrition and the microbiome as modifiers of early neurodevelopment. Trends Neurosci 2021; 44:753-764. [PMID: 34303552 DOI: 10.1016/j.tins.2021.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/23/2021] [Accepted: 06/11/2021] [Indexed: 01/16/2023]
Abstract
Malnutrition refers to a dearth, excess, or altered differential ratios of calories, macronutrients, or micronutrients. Malnutrition, particularly during early life, is a pressing global health and socioeconomic burden that is increasingly associated with neurodevelopmental impairments. Understanding how perinatal malnutrition influences brain development is crucial to uncovering fundamental mechanisms for establishing behavioral neurocircuits, with the potential to inform public policy and clinical interventions for neurodevelopmental conditions. Recent studies reveal that the gut microbiome can mediate dietary effects on host physiology and that the microbiome modulates the development and function of the nervous system. This review discusses evidence that perinatal malnutrition alters brain development and examines the maternal and neonatal microbiome as a potential contributing factor.
Collapse
Affiliation(s)
- Elena J L Coley
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
42
|
Clarkson-Townsend DA, Bales KL, Hermetz KE, Burt AA, Pardue MT, Marsit CJ. Developmental chronodisruption alters placental signaling in mice. PLoS One 2021; 16:e0255296. [PMID: 34370755 PMCID: PMC8351967 DOI: 10.1371/journal.pone.0255296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/13/2021] [Indexed: 11/18/2022] Open
Abstract
Chronodisruption has been largely overlooked as a developmental exposure. The placenta, a conduit between the maternal and fetal environments, may relay circadian cues to the fetus. We have previously shown that developmental chronodisruption causes visual impairment and increased retinal microglial and macrophage marker expression. Here, we investigated the impacts of environmental chronodisruption on fetal and placental outcomes in a C57BL/6J mouse (Mus musculus) model. Developmental chronodisruption had no effect on embryo count, placental weight, or fetal sex ratio. When measured with RNAseq, mice exposed to developmental chronodisruption (CD) had differential placental expression of several transcripts including Serpinf1, which encodes pigment epithelium-derived factor (PEDF). Immunofluorescence of microglia/macrophage markers, Iba1 and CD11b, also revealed significant upregulation of immune cell markers in CD-exposed placenta. Our results suggest that in utero chronodisruption enhances placental immune cell expression, potentially programming a pro-inflammatory tissue environment.
Collapse
Affiliation(s)
- Danielle A. Clarkson-Townsend
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States of America
| | - Katie L. Bales
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States of America
- Department of Ophthalmology, Emory University, Atlanta, GA, United States of America
| | - Karen E. Hermetz
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Amber A. Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Machelle T. Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States of America
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Carmen J. Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| |
Collapse
|
43
|
Placental Macrophages Demonstrate Sex-Specific Response to Intrauterine Inflammation and May Serve as a Marker of Perinatal Neuroinflammation. J Reprod Immunol 2021; 147:103360. [PMID: 34390899 DOI: 10.1016/j.jri.2021.103360] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 07/06/2021] [Accepted: 07/26/2021] [Indexed: 12/23/2022]
Abstract
Preterm birth (PTB) is considered to be one of the most frequent causes of neonatal death. Prompt and effective measures to predict adverse fetal outcome following PTB are urgently needed. Placenta macrophages are a critical immune cell population during pregnancy, phenotypically divided into M1 and M2 subsets. An established mouse model of intrauterine inflammation (IUI) was applied. Placenta (labyrinth) and corresponding fetal brain were harvested within 24 hours post injection (hpi). Flow cytometry, Western blot, real-time qPCR, and regular histology were utilized to examine the cytokines, macrophage polarization, and sex-specificity. Placental exposure to LPS led to significantly reduced labyrinth thickness compared to PBS-exposed controls as early as 3 hpi, accompanied by apoptosis and necrosis. Pro-inflammatory M1 markers, Il-1β, and iNOS, and anti-inflammatory M2 marker Il-10 increased significantly in placentas exposed to IUI. Analysis of flow cytometry revealed that fetal macrophages (Hofbauer cell, HBCs) were mostly M1-like and that maternal inter-labyrinth macrophages (MIM) were M2-like in their features in IUI. Male fetuses displayed significantly decreased M2-like features in HBCs at 3 and 6 hpi, while female fetuses showed significant increase in M2-like features in MIM at 3 and 6 hpi. Furthermore, there was a significant correlation between the frequency of HBCs and corresponding microglial marker expression at 3 and 6 hpi. Placental macrophages demonstrated sex-specific features in response to IUI. Specifically, HBCs may be a potential biomarker for fetal brain injury at preterm birth.
Collapse
|
44
|
The impact of maternal obesity on childhood neurodevelopment. J Perinatol 2021; 41:928-939. [PMID: 33249428 DOI: 10.1038/s41372-020-00871-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 10/10/2020] [Accepted: 11/04/2020] [Indexed: 02/08/2023]
Abstract
There is growing clinical and experimental evidence to suggest that maternal obesity increases children's susceptibility to neurodevelopmental and neuropsychiatric disorders. Given the worldwide obesity epidemic, it is crucial that we acquire a thorough understanding of the available evidence, identify gaps in knowledge, and develop an agenda for intervention. This review synthesizes human and animal studies investigating the association between maternal obesity and offspring brain health. It also highlights key mechanisms underlying these effects, including maternal and fetal inflammation, alterations to the microbiome, epigenetic modifications of neurotrophic genes, and impaired dopaminergic and serotonergic signaling. Lastly, this review highlights several proposed interventions and priorities for future investigation.
Collapse
|
45
|
Davis J, Mire E. Maternal obesity and developmental programming of neuropsychiatric disorders: An inflammatory hypothesis. Brain Neurosci Adv 2021; 5:23982128211003484. [PMID: 33889757 PMCID: PMC8040564 DOI: 10.1177/23982128211003484] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
Maternal obesity is associated with the development of a variety of neuropsychiatric disorders; however, the mechanisms behind this association are not fully understood. Comparison between maternal immune activation and maternal obesity reveals similarities in associated impairments and maternal cytokine profile. Here, we present a summary of recent evidence describing how inflammatory processes contribute towards the development of neuropsychiatric disorders in the offspring of obese mothers. This includes discussion on how maternal cytokine levels, fatty acids and placental inflammation may interact with foetal neurodevelopment through changes to microglial behaviour and epigenetic modification. We also propose an exosome-mediated mechanism for the disruption of brain development under maternal obesity and discuss potential intervention strategies.
Collapse
Affiliation(s)
- Jonathan Davis
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Erik Mire
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
46
|
Sheridan SD, Thanos JM, De Guzman RM, McCrea LT, Horng JE, Fu T, Sellgren CM, Perlis RH, Edlow AG. Umbilical cord blood-derived microglia-like cells to model COVID-19 exposure. Transl Psychiatry 2021; 11:179. [PMID: 33741894 PMCID: PMC7976669 DOI: 10.1038/s41398-021-01287-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 02/02/2023] Open
Abstract
Microglia, the resident brain immune cells, play a critical role in normal brain development, and are impacted by the intrauterine environment, including maternal immune activation and inflammatory exposures. The COVID-19 pandemic presents a potential developmental immune challenge to the fetal brain, in the setting of maternal SARS-CoV-2 infection with its attendant potential for cytokine production and, in severe cases, cytokine storming. There is currently no biomarker or model for in utero microglial priming and function that might aid in identifying the neonates and children most vulnerable to neurodevelopmental morbidity, as microglia remain inaccessible in fetal life and after birth. This study aimed to generate patient-derived microglial-like cell models unique to each neonate from reprogrammed umbilical cord blood mononuclear cells, adapting and extending a novel methodology previously validated for adult peripheral blood mononuclear cells. We demonstrate that umbilical cord blood mononuclear cells can be used to create microglial-like cell models morphologically and functionally similar to microglia observed in vivo. We illustrate the application of this approach by generating microglia from cells exposed and unexposed to maternal SARS-CoV-2 infection. Our ability to create personalized neonatal models of fetal brain immune programming enables non-invasive insights into fetal brain development and potential childhood neurodevelopmental vulnerabilities for a range of maternal exposures, including COVID-19.
Collapse
Affiliation(s)
- Steven D Sheridan
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica M Thanos
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Rose M De Guzman
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Liam T McCrea
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Joy E Horng
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Ting Fu
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Roy H Perlis
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | - Andrea G Edlow
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA.
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
47
|
Salberg S, Yamakawa GR, Griep Y, Bain J, Beveridge JK, Sun M, McDonald SJ, Shultz SR, Brady RD, Wright DK, Noel M, Mychasiuk R. Pain in the Developing Brain: Early Life Factors Alter Nociception and Neurobiological Function in Adolescent Rats. Cereb Cortex Commun 2021; 2:tgab014. [PMID: 34296160 PMCID: PMC8152853 DOI: 10.1093/texcom/tgab014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 01/09/2023] Open
Abstract
Although adverse early experiences prime individuals to be at increased risk for chronic pain, little research has examined the trauma–pain relationship in early life or the underlying mechanisms that drive pathology over time. Given that early experiences can potentiate the nociceptive response, this study aimed to examine the effects of a high-fat, high-sugar (HFHS) diet and early life stress (maternal separation [MS]) on pain outcomes in male and female adolescent rats. Half of the rats also underwent a plantar-incision surgery to investigate how the pain system responded to a mildly painful stimuli in adolescence. Compared with controls, animals that were on the HFHS diet, experienced MS, or had exposure to both, exhibited increased anxiety-like behavior and altered thermal and mechanical nociception at baseline and following the surgery. Advanced magnetic resonance imaging demonstrated that the HFHS diet and MS altered the maturation of the brain, leading to changes in brain volume and diffusivity within the anterior cingulate, amygdala, corpus callosum, nucleus accumbens, and thalamus, while also modifying the integrity of the corticospinal tracts. The effects of MS and HFHS diet were often cumulative, producing exacerbated pain sensitivity and increased neurobiological change. As early experiences are modifiable, understanding their role in pain may provide targets for early intervention/prevention.
Collapse
Affiliation(s)
- Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Yannick Griep
- Behavioural Science Institute, Radboud University, Nijmegen 6525 GD, the Netherlands.,Division of Epidemiology, Stress Research Institute, Stockholm University, Stockholm 114 19, Sweden
| | - Jesse Bain
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Jaimie K Beveridge
- Department of Psychology, University of Calgary, Calgary T2N 1N4, Canada
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne 3004, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne 3086, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Melanie Noel
- Department of Psychology, University of Calgary, Calgary T2N 1N4, Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne 3004, Australia.,Department of Psychology, University of Calgary, Calgary T2N 1N4, Canada
| |
Collapse
|
48
|
Pekala M, Doliwa M, Kalita K. Impact of maternal immune activation on dendritic spine development. Dev Neurobiol 2021; 81:524-545. [PMID: 33382515 DOI: 10.1002/dneu.22804] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/26/2020] [Accepted: 12/28/2020] [Indexed: 01/08/2023]
Abstract
Dendritic spines are small dendritic protrusions that harbor most excitatory synapses in the brain. The proper generation and maturation of dendritic spines are crucial for the regulation of synaptic transmission and formation of neuronal circuits. Abnormalities in dendritic spine density and morphology are common pathologies in autism and schizophrenia. According to epidemiological studies, one risk factor for these neurodevelopmental disorders is maternal infection during pregnancy. This review discusses spine alterations in animal models of maternal immune activation in the context of neurodevelopmental disorders. We describe potential mechanisms that might be responsible for prenatal infection-induced changes in the dendritic spine phenotype and behavior in offspring.
Collapse
Affiliation(s)
- Martyna Pekala
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Doliwa
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kalita
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
49
|
Reddaway J, Brydges NM. Enduring neuroimmunological consequences of developmental experiences: From vulnerability to resilience. Mol Cell Neurosci 2020; 109:103567. [PMID: 33068720 PMCID: PMC7556274 DOI: 10.1016/j.mcn.2020.103567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/14/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
The immune system is crucial for normal neuronal development and function (neuroimmune system). Both immune and neuronal systems undergo significant postnatal development and are sensitive to developmental programming by environmental experiences. Negative experiences from infection to psychological stress at a range of different time points (in utero to adolescence) can permanently alter the function of the neuroimmune system: given its prominent role in normal brain development and function this dysregulation may increase vulnerability to psychiatric illness. In contrast, positive experiences such as exercise and environmental enrichment are protective and can promote resilience, even restoring the detrimental effects of negative experiences on the neuroimmune system. This suggests the neuroimmune system is a viable therapeutic target for treatment and prevention of psychiatric illnesses, especially those related to stress. In this review we will summarise the main cells, molecules and functions of the immune system in general and with specific reference to central nervous system development and function. We will then discuss the effects of negative and positive environmental experiences, especially during development, in programming the long-term functioning of the neuroimmune system. Finally, we will review the sparse but growing literature on sex differences in neuroimmune development and response to environmental experiences. The immune system is essential for development and function of the central nervous system (neuroimmune system) Environmental experiences can permanently alter neuroimmune function and associated brain development Altered neuroimmune function following negative developmental experiences may play a role in psychiatric illnesses Positive experiences can promote resilience and rescue the effects of negative experiences on the neuroimmune system The neuroimmune system is therefore a viable therapeutic target for preventing and treating psychiatric illnesses
Collapse
Affiliation(s)
- Jack Reddaway
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Nichola M Brydges
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK.
| |
Collapse
|
50
|
Sheridan SD, Thanos JM, De Guzman RM, McCrea LT, Horng J, Fu T, Sellgren CM, Perlis RH, Edlow AG. Umbilical cord blood derived microglia-like cells to model COVID-19 exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.10.07.329748. [PMID: 33052344 PMCID: PMC7553171 DOI: 10.1101/2020.10.07.329748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Microglia, the resident brain immune cells, play a critical role in normal brain development, and are impacted by the intrauterine environment, including maternal immune activation and inflammatory exposures. The COVID-19 pandemic presents a potential developmental immune challenge to the fetal brain, in the setting of maternal SARS-CoV-2 infection with its attendant potential for cytokine production and, in severe cases, cytokine storming. There is currently no biomarker or model for in utero microglial priming and function that might aid in identifying the neonates and children most vulnerable to neurodevelopmental morbidity, as microglia remain inaccessible in fetal life and after birth. This study aimed to generate patient-derived microglial-like cell models unique to each neonate from reprogrammed umbilical cord blood mononuclear cells, adapting and extending a novel methodology previously validated for adult peripheral blood mononuclear cells. We demonstrate that umbilical cord blood mononuclear cells can be used to create microglial-like cell models morphologically and functionally similar to microglia observed in vivo . We illustrate the application of this approach by generating microglia from cells exposed and unexposed to maternal SARS-CoV-2 infection. Our ability to create personalized neonatal models of fetal brain immune programming enables non-invasive insights into fetal brain development and potential childhood neurodevelopmental vulnerabilities for a range of maternal exposures, including COVID-19.
Collapse
|