1
|
Kirkpatrick M, Mandal G, Elhadidy I, Mariani N, Priestley K, Pariante CM, Borsini A. From placenta to the foetus: a systematic review of in vitro models of stress- and inflammation-induced depression in pregnancy. Mol Psychiatry 2025; 30:1689-1707. [PMID: 39639175 PMCID: PMC11919713 DOI: 10.1038/s41380-024-02866-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Depression in pregnancy can increase vulnerability for psychiatric disorders in the offspring, likely via the transfer of heightened maternal cortisol and cytokines to the in-utero environment. However, the precise cellular and molecular mechanisms, are largely unclear. Animal studies can represent this complex pathophysiology at a systemic level but are expensive and ethically challenging. While simpler, in vitro models offer high-throughput opportunities. Therefore, this systematic review integrates findings of in vitro models relevant to depression in pregnancy, to generate novel hypotheses and targets for intervention. METHODS The systematic analysis covered studies investigating glucocorticoid or cytokine challenges on placental or foetal neural progenitor cells (NPCs), with or without co-treatment with sex hormones. RESULTS Of the 50 included studies, 11 used placental cells and 39 NPCs; surprisingly, only one used a combination of oestrogen and cortisol, and no study combined placental cells and NPCs. In placental cells, cortisol or cytokines decreased nutrient transporter expression and steroidogenic enzyme activity, and increased cytokine production. NPCs exhibited decreases in proliferation and differentiation, via specific molecular pathways, namely, inhibition of hedgehog signalling and activation of kynurenine pathway. In these cells, studies also highlighted epigenetic priming of stress and inflammatory pathways. CONCLUSIONS Overall, results suggest that stress and inflammation not only detrimentally impact placental regulation of nutrients and hormones to the foetus, but also activate downstream pathways through increased inflammation in the placenta, ultimately eliciting adverse effects on foetal neurogenesis. Future research should investigate how sex hormones regulate these mechanisms, with the aim of developing targeted therapeutic approaches for depression in pregnancy.
Collapse
Affiliation(s)
- Madeline Kirkpatrick
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Gargi Mandal
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Ismail Elhadidy
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Nicole Mariani
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Kristi Priestley
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Alessandra Borsini
- Department of Psychological Medicine, Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK.
| |
Collapse
|
2
|
Carver AJ, Fairbairn FM, Taylor RJ, Boggarapu S, Kamau NR, Gajmer A, Stevens HE. Placental Igf1 Overexpression Sex-Specifically Impacts Mouse Placenta Structure, Altering Offspring Striatal Development and Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.644829. [PMID: 40196637 PMCID: PMC11974794 DOI: 10.1101/2025.03.27.644829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Insulin-like growth factor 1 (IGF1) is produced primarily in the placenta in utero and is an essential hormone for neurodevelopment. Specifically, how placental IGF1 production persistently influences the brain is unclear. This study evaluated the effects of placental Igf1 overexpression on embryonic and postnatal brain development, particularly for striatum, a region highly linked to neurodevelopmental disorders. Placental Igf1 was overexpressed via placental-targeted CRISPR manipulation. This overexpression altered placenta structure and function distinctly in females and males. Early differences in placental function altered the trajectory of striatal development, as adult females showed persistent changes in striatal cell composition and striatal dependent behavior while males were less affected in brain and behavior outcomes. Overall, these results demonstrate that placental Igf1 expression alters striatal development and behavior in ways relevant to neurodevelopmental disorders. These findings expand our understanding of placental influence on neurodevelopment and will aid in identifying placental-targeted preventive interventions.
Collapse
Affiliation(s)
- Annemarie J. Carver
- Interdisciplinary Graduate Program in Genetics, University of Iowa, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Faith M. Fairbairn
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Robert J. Taylor
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Shanmukh Boggarapu
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Njenga R. Kamau
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Amrita Gajmer
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Hanna E. Stevens
- Interdisciplinary Graduate Program in Genetics, University of Iowa, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
- Hawk-Intellectual and Developmental Disabilities Research Center, University of Iowa, IA, USA
- Senior author
- Lead contact
| |
Collapse
|
3
|
Demonceaux M, Werner O, Cadeau O, Guerra A, Roy A, Ferchaud-Roucher V, Baruteau AE. Congenital Heart Diseases and Neurodevelopmental Disorders: New Insights Through the DOHaD Hypothesis. JACC Basic Transl Sci 2025:S2452-302X(25)00068-3. [PMID: 40272355 DOI: 10.1016/j.jacbts.2025.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 04/25/2025]
Abstract
Congenital heart disease (CHD) is the primary cause of birth defects, affecting 9 per 1,000 live births. Up to 50% of them will develop neurodevelopmental disorders, two-thirds of which being unexplained by postnatal risk factors. Recent advances suggest a triangular relationship between the placenta and the fetal heart and brain in CHD, consistent with the Developmental Origins of Health and Disease hypothesis, ie, the in utero programming of early and later-in-life noncommunicable cardiometabolic and mental diseases. The current review provides comprehensive evidence of placental, cardiac, and cerebral tissues interactions, and details how placental dysregulation may affect vasculogenesis, angiogenesis and neural tube closure, hemodynamics, energy supply, endocrine function, and epigenetic regulation of the developing heart and brain. Future studies should include placental research, since identifying placental biomarkers would allow early identification of CHD infants at higher risk for neurodevelopmental disorders, leading to targeted preventive personalized interventions.
Collapse
Affiliation(s)
- Marie Demonceaux
- Nantes Université, CHU Nantes, INRAE UMR 1280 PhAN, Nantes, France
| | - Oscar Werner
- Nantes Université, CHU Nantes, Département de Cardiologie Pédiatrique et Chirurgie Cardiaque Pédiatrique, FHU PRECICARE, Nantes, France; Nantes Université, CHU Nantes, INSERM, CIC FEA 1413, Nantes, France
| | - Olivier Cadeau
- Nantes Université, LPPL, SFR Confluences, Université d'Angers, Angers, France; Nantes Université, CHU Nantes, Centre Référent des Troubles d'Apprentissage, Nantes, France
| | - Amanda Guerra
- Nantes Université, LPPL, SFR Confluences, Université d'Angers, Angers, France; Nantes Université, CHU Nantes, Centre Référent des Troubles d'Apprentissage, Nantes, France
| | - Arnaud Roy
- Nantes Université, LPPL, SFR Confluences, Université d'Angers, Angers, France; Nantes Université, CHU Nantes, Centre Référent des Troubles d'Apprentissage, Nantes, France
| | | | - Alban-Elouen Baruteau
- Nantes Université, CHU Nantes, INRAE UMR 1280 PhAN, Nantes, France; Nantes Université, CHU Nantes, Département de Cardiologie Pédiatrique et Chirurgie Cardiaque Pédiatrique, FHU PRECICARE, Nantes, France; Nantes Université, CHU Nantes, INSERM, CIC FEA 1413, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.
| |
Collapse
|
4
|
Hikosaka M, Parvez MSA, Yamawaki Y, Oe S, Liang Y, Wada Y, Hirahara Y, Koike T, Imai H, Oishi N, Schalbetter SM, Kumagai A, Yoshida M, Sakurai T, Kitada M, Meyer U, Narumiya S, Ohtsuki G. Maternal immune activation followed by peripubertal stress combinedly produce reactive microglia and confine cerebellar cognition. Commun Biol 2025; 8:296. [PMID: 40033126 PMCID: PMC11876345 DOI: 10.1038/s42003-025-07566-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/15/2025] [Indexed: 03/05/2025] Open
Abstract
The functional alteration of microglia arises in brains exposed to external stress during early development. Pathophysiological findings of neurodevelopmental disorders such as schizophrenia and autism spectrum disorder suggest cerebellar functional deficits. However, the link between stress-induced microglia reactivity and cerebellar dysfunction is missing. Here, we investigate the developmental immune environment in translational mouse models that combine two risk factors: maternal infection and repeated social defeat stress (2HIT). We find the synergy of inflammatory stress insults, leading to microglial increase specifically in the cerebellum of both sexes. Microglial turnover correlates with the Purkinje neuron loss in 2HIT mice. Highly multiplexed imaging-mass-cytometry identifies a cell transition to TREM2(+) stress-associated microglia in the cerebellum. Single-cell-proteomic clustering reveals IL-6- and TGFβ-signaling association with microglial cell transitions. Reduced excitability of remaining Purkinje cells, cerebellum-involved brain-wide functional dysconnectivity, and behavioral abnormalities indicate cerebellar cognitive dysfunctions in 2HIT animals, which are ameliorated by both systemic and cerebellum-specific microglia replacement.
Collapse
Affiliation(s)
- Momoka Hikosaka
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Md Sorwer Alam Parvez
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yuki Yamawaki
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Souichi Oe
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Yuan Liang
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
- Institute of Basic Theory in Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yayoi Wada
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Yukie Hirahara
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Taro Koike
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Hirohiko Imai
- Department of Systems Science, Kyoto University Graduate School of Informatics, Yoshida-Honmachi, Kyoto, Japan
- Innovation Research Center for Quantum Medicine, Gifu University School of Medicine, Gifu, Japan
| | - Naoya Oishi
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sina M Schalbetter
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | | | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Takeshi Sakurai
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Masaaki Kitada
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Urs Meyer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Gen Ohtsuki
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
5
|
Zhou J, Tong J, Liang C, Wu P, Ouyang J, Cai W, Cheng L, Teng Y, Sheng J, Gao G, Yan S, Tao F, Tong S, Huang K. Prenatal metals and offspring cognitive development: Insights from a large-scale placental bioassay study. ENVIRONMENTAL RESEARCH 2025; 267:120684. [PMID: 39716677 DOI: 10.1016/j.envres.2024.120684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
The developing foetus is particularly sensitive to neurotoxic metals. The placenta is considered an ideal tissue for biomonitoring prenatal cumulative metal exposure. Based on the Ma'anshan Birth Cohort study (MABC) in China, this study investigated associations of non-essential metals and essential metals in placenta, including arsenic (As), cadmium (Cd), mercury (Hg), lead (Pb), cobalt (Co), selenium (Se) and zinc (Zn), with cognitive development in children among 1586 mother-child pairs. Also, we explored potential interactions between the metals and modifying role of the sex. Children's cognitive development was tested at preschool age using the Wechsler Preschool and Primary Scale of Intelligence, Fourth Edition (WPPSI-IV). Analyses used multiple linear regression, Bayesian kernel machine regression (BKMR), the quantile g-computation (Qgcomp), interaction and marginal effects models, and restricted cubic spline in R and STATA. In this study, the geometric means [GMs (SD)] for placental metal concentrations were 8.10 (7.54) ng/g for As, 32.32 (29.20) ng/g for Cd, 11.89 (13.33) ng/g for Hg, 32.21 (28.24) ng/g for Pb, 15.05 (8.91) ng/g for Co, 508.82 (192.35) ng/g for Se, 18481.60 (14030.61) ng/g for Zn. In individual models, placental As levels were negatively associated with the Fluent Reasoning Index (FRI) in the overall sample. Cd levels were negatively associated with the full-scale intelligence quotient (FSIQ), the Verbal Comprehension Index (VCI) and the Visual Spatial Index (VSI). The four metal mixture (As, Cd, Hg and Pb) was negatively associated with FSIQ, VCI, VSI and FRI. Placental Cd and As were the largest contributors to the negative mixture association on the FSIQ. The negative associations of placental As, Cd and Hg with FSIQ in children were gradually attenuated with increasing Zn and Se. After stratifying by sex, the individual and mixture associations between elevated placental non-essential metal exposures and reduced cognitive scores were significant only in boys. Zn and Se were the major contributors to the positive mixture associations on the FSIQ. In summary, prenatal exposure to As, Cd and Hg has sex-specific adverse associations on children's cognitive development. A more accurate assessment of the necessity of prenatal supplementation of micronutrients including Zn and Se is needed.
Collapse
Affiliation(s)
- Jixing Zhou
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China
| | - Juan Tong
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China
| | - Chunmei Liang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China
| | - Penggui Wu
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China
| | - Jiajun Ouyang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China
| | - Wenjin Cai
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China
| | - Lu Cheng
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China
| | - Yuzhu Teng
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China
| | - Jie Sheng
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Scientific Research Center in Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Guopeng Gao
- Maternal and Child Health Care Center of Ma'anshan, No 24 Jiashan Road, Ma'anshan, 243011, Anhui, China
| | - Shuangqin Yan
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Maternal and Child Health Care Center of Ma'anshan, No 24 Jiashan Road, Ma'anshan, 243011, Anhui, China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China
| | - Shilu Tong
- National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; School of Public Health and Social Work, Queensland University of Technology, Brisbane, QLD 4001, Australia
| | - Kun Huang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (AHMU), MOE, Hefei 230032, China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Hefei 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Hefei, 230032, China.
| |
Collapse
|
6
|
Li M, Hu Y, Wu X, Tong J, Tao J, Tang A, Ji Y, Yao Y, Tao F, Liang C. Placental Ferroptosis May Be Involved in Prenatal Arsenic Exposure Induced Cognitive Impairment in Offspring. Biol Trace Elem Res 2025:10.1007/s12011-025-04525-0. [PMID: 39912999 DOI: 10.1007/s12011-025-04525-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025]
Abstract
The association between prenatal arsenic (As) exposure and offspring's cognition is still unclear, and the underlying etiology has also not been elucidated. Based on the Ma'anshan Birth Cohort (MABC) study in China, 1814 mother-child pairs were included in this study, and the association of As levels in cord serum with preschoolers' intelligence scores was explored. To validate the results from population study, in vivo models were adopted to observe the association between prenatal As exposure and spatial learning and memory abilities of mice offsprings. The As-exposure induced ferroptosis in the placenta of human beings as well as C57BL/6 J mice and HTR-8/SVneo cells was explored in order to clarify the potential cause of impairment of offspring's cognition related to As exposure, respectively. In the population study, we observed a significant inverse association between natural logarithm transformed (ln) As levels and preschoolers' intelligence scores, especially for the fluid reasoning index (FRI) [(β (95%CI): - 1.07 (- 1.98, - 0.16)] and working memory index (WMI) [β (95%CI): - 1.51 (- 2.76, - 0.25)]. Meanwhile, the data from in vivo models revealed that the learning and memory abilities of offspring mice decreased after prenatal As exposure. The occurrence of ferroptosis-like characteristics in the placenta and HTR-8/SVneo cells after As exposure was observed, accompanying with evident oxidative stress, iron accumulation, mitochondrial damage, and decreased protein levels of GPX4, xCT, and FTH1 (or FPN1). Notably, the ferroptosis-like alterations induced by NaAsO2 can be effectively alleviated by N-acetylcysteine (NAC) and ferrostatin-1 (Fer-1) treatment in HTR-8/SVneo cells, respectively. In conclusion, prenatal As exposure associates with impairment of offspring's cognition, and placental ferroptosis may be involved in the association. Further studies are needed to confirm the findings.
Collapse
Affiliation(s)
- Mengzhu Li
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yuan Hu
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Jiulongpo District Center for Disease Control and Prevention, Chongqing, 400039, China
| | - Xiaoyan Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jiajing Tao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Anni Tang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yanli Ji
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Yuyou Yao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Chunmei Liang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
7
|
Vacher CM, Tsompanidis A, Firestein MR, Penn AA. Neuroactive steroid exposure impacts neurodevelopment: Comparison of human and rodent placental contribution. J Neuroendocrinol 2025:e13489. [PMID: 39789736 DOI: 10.1111/jne.13489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
The placenta is a fetal endocrine organ that secretes many neuroactive factors, including steroids, that play critical roles in brain development. The study of the placenta-brain axis and the links between placental function and brain development represents an emerging research area dubbed "neuroplacentology." The placenta drives many circulating fetal steroids to very high levels during gestation. Recent studies have highlighted the critical role of placental steroids in shaping specific brain structures and behaviors. This review uses a cross-species framework to discuss the genomic factors, in-utero environmental changes, and placental conditions that alter placental steroidogenesis, leading to changes in early developmental trajectories relevant for psychiatric conditions such as autism, in a sex-linked manner.
Collapse
Affiliation(s)
- Claire-Marie Vacher
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Morgan R Firestein
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, USA
| | - Anna A Penn
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
- New York Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| |
Collapse
|
8
|
Pavy CL, Shaw JC, Palliser HK, Moloney RA, Hirst JJ. Neurosteroid replacement therapy using tiagabine and zuranolone restores cerebellar neurodevelopment and reduces hyperactive behaviour following preterm birth. J Dev Orig Health Dis 2025; 16:e2. [PMID: 39773606 DOI: 10.1017/s2040174424000394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Preterm birth exposes the neonate to hypoxic-ischaemic and excitotoxic insults that impair neurodevelopment and are magnified by the premature loss of placentally supplied, inhibitory neurosteroids. The cerebellum is a neuronally dense brain region, which undergoes critical periods of development during late gestation, when preterm births frequently occur. We propose that neurosteroid replacement therapy using tiagabine and zuranolone will protect the cerebellum against preterm-associated insults. Guinea pig dams received c-section surgery preterm (gestational age (GA) 64) or at term (GA70) with preterm pups administered tiagabine (2.5 mg/kg/day), zuranolone (1 mg/kg/day) or vehicle (15% β-cyclodextrin) until term equivalent age (GA70). Behavioural testing was performed at corrected postnatal day 8 (PND8) and PND41 with tissue collection occurring at PND42. Neurodevelopmental markers (MBP, OLIG2 and NeuN) were assessed within the cerebellum by immunohistochemistry, whilst GABAergic and glutamatergic pathway expression was quantified using high throughput RT-PCR. Zuranolone and, to a lesser extent, tiagabine were able to protect against hyperactive behaviour at PND8 in males, whilst in females, a less marked hyperactive phenotype was present with neither treatment impacting behaviour further. Both treatments improved MBP staining, whilst tiagabine was found to restore oligodendrocyte maturation in females only. GABAergic and glutamatergic pathway expression was found to be restored by both treatments in females. Overall, this study demonstrates the neuroprotective attributes of neurosteroid replacement therapy using tiagabine and zuranolone, thereby demonstrating their potential to mitigate long-term neurodevelopmental impairments. Furthermore, the sexually dimorphic effects observed suggest future investigations may show increased benefit by using sex-specific treatment regimes.
Collapse
Affiliation(s)
- Carlton L Pavy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, Australia
| | - Julia C Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, Australia
| | - Roisin A Moloney
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
9
|
Zoubovsky SP, Muglia LJ. Transplacental signals involved in the programming effects of prenatal psychosocial stress on neurodevelopment. Neurotoxicol Teratol 2025; 107:107424. [PMID: 39755178 DOI: 10.1016/j.ntt.2025.107424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/03/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Exposure to psychosocial stress during pregnancy has been associated with the emergence of neurodevelopmental and neuropsychiatric disorders in offspring. The placenta is known to orchestrate various functions that are essential for normal fetal development, including the brain. It has therefore been postulated that alterations in such functions, and downstream signaling, have the potential to dramatically affect brain developmental trajectories and contribute to adverse neurodevelopmental outcomes. This review will focus on discussing various placental functions that have been proposed to be affected by exposure to prenatal psychosocial stress and the implications of such disruptions on long-term neurodevelopmental programming.
Collapse
Affiliation(s)
- Sandra P Zoubovsky
- Department of Pediatrics, University of Colorado, Denver, CO, United States
| | - Louis J Muglia
- Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Office of the President, Burroughs Wellcome Fund, Research Triangle Park, Durham, NC, United States.
| |
Collapse
|
10
|
Hirst JJ, Palliser HK, Pavy C, Shaw JC, Moloney RA. Neurosteroid replacement approaches for improving outcomes after compromised pregnancies and preterm birth. Front Neuroendocrinol 2025; 76:101169. [PMID: 39622477 DOI: 10.1016/j.yfrne.2024.101169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/08/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
The levels of the key neurosteroid of pregnancy, allopregnanolone, are very high in the fetal and maternal brain compared to after birth. These levels are maintained by the placenta which forms a placental connection to fetal brain development. Maternal stresses depress placental synthesis resulting in a fall in allopregnanolone levels leading to deficits in myelination that continue into childhood. This contributes to an increased incidence of behavioural disorders. Supplementing neurosteroid action with allopregnanolone analogues or raising endogenous production with mitochondrial translocator protein (TSPO) ligands reverses these deficits. Preterm birth leads to an early dramatic loss of neurosteroid support for brain development leading to marked deficits in myelination and susceptibility to hypoxic-ischaemic injury. Postnatal treatment with the allopregnanolone analogue ganaxolone improves myelination and reduces hyperactive behaviour. TSPO ligands such as emapunil have been shown to improve oligodendrocyte maturation. These findings support the use of allopregnanolone supplementation approaches after pregnancy compromises to improve outcome.
Collapse
Affiliation(s)
- Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia; Hunter Medical Research Institute, Mothers and Babies Research Program, Newcastle, Australia
| | - Carlton Pavy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia; Hunter Medical Research Institute, Mothers and Babies Research Program, Newcastle, Australia
| | - Julia C Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia; Hunter Medical Research Institute, Mothers and Babies Research Program, Newcastle, Australia
| | - Roisin A Moloney
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia; Hunter Medical Research Institute, Mothers and Babies Research Program, Newcastle, Australia
| |
Collapse
|
11
|
Song X, Yan S, Lai S, Zhang Y, Wang Y, He J, Huang D, Zhang J, Lu X, Chen G, Chen P, Zhong Q, Zhang R, Wu Y, Yin J, Zhong S, Jia Y. Gender differences of neurometabolic and neuroendocrine alternations and its lateralization in adolescents with major depressive disorder. BMC Psychiatry 2024; 24:949. [PMID: 39731037 DOI: 10.1186/s12888-024-06428-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 12/19/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND The clinical characteristics of major depressive disorder (MDD) in adolescents show notable gender-related differences, but the cause of these differences is still not understood. The current research concentrates on the changes in neurometabolism and neuroendocrine function, aiming to identify differences in endocrine function and brain metabolism between male and female adolescents with MDD. METHODS A total of 121 teenagers diagnosed with MDD (43 males and 78 females) were enlisted as participants. Measurement was conducted on levels of endocrine hormones, which included free tri-iodothyronine (FT3), total tri-iodothyronine (TT3), free thyroxin (FT4), total thyroxin (TT4), thyroid-stimulating hormone (TSH), cortisol, and adrenocorticotropic hormone (ACTH). Obtained through 1H-MRS, the N-acetyl aspartate (NAA) and choline containing compounds (Cho) to creatine (Cr) ratios were acquired for the prefrontal whiter matter (PWM), anterior cingulate cortex (ACC), basal ganglia (BG), thalamus, and cerebellum. RESULT After adjusting for multiple comparisons, female adolescents with MDD showed lower ACTH levels compared to their male counterparts. An increased lateralization index (LI) was observed in female patients for both the thalamic Cho/Cr ratio and the basal ganglia NAA/Cr ratio. Additionally, an intriguing finding was that in male adolescent patients, TT4 levels were significantly correlated with the Cho/Cr ratio in the left cerebellum. However, no such correlation between hormones and brain metabolism was found in females. CONCLUSIONS Gender differences in endocrine and neurometabolic abnormalities may contribute to the gender-specific pathophysiology of MDD in adolescent patients. Metabolic abnormalities and lateralization changes are observed in different brain regions for male and female MDD patients.
Collapse
Affiliation(s)
- Xiaodong Song
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Shuya Yan
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Shunkai Lai
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Yiliang Zhang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Ying Wang
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Jiali He
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Dong Huang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Jianzhao Zhang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Xiaodan Lu
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Guanmao Chen
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Pan Chen
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Qilin Zhong
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Rongxu Zhang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Yangyu Wu
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Jie Yin
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Shuming Zhong
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China.
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
12
|
J-P NA, Eitoku M, Yamasaki K, Mitsuda N, Fujieda M, Maeda N, Suganuma N. Association between chest-to-head circumference ratio at birth and childhood neurodevelopment: the Japan Environment and Children's Study. J Dev Orig Health Dis 2024; 15:e34. [PMID: 39726347 DOI: 10.1017/s2040174424000412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Children born growth-restricted are well recognized to be at an increased risk of poor neurodevelopmental outcomes. This prospective study examined the influence of chest-to-head circumference ratio at birth on neurodevelopment in the first three years among children enrolled in the Japan Environment and Children's Study. We analyzed information of 84,311 children (43,217 boys, 41,094 girls). Children were divided into low, normal, and high chest-to-head circumference ratio groups. Neurodevelopment was assessed every six months (from 6 months to 3 years) using the Ages and Stages Questionnaire (Japanese translation), with delays defined as scores below 2 standard deviations from the mean. Additionally, we evaluated the contributions of chest and head circumference to the observed association. Linear mixed-effect regression revealed increased risk of delays in communication, gross motor, fine motor, problem-solving, and personal-social skills in the low-ratio group compared to the normal-ratio group. Adjusted risk ratios were in the range of 1.14 - 1.39 in boys and 1.16 - 1.37 in girls, with no such increase observed in the high-ratio group. The heightened risk in the low-ratio group was likely associated with a relatively narrow chest rather than a large head. The area under the ROC curves in predicting any developmental delay at three years for newborn measurements ranged from 0.513 to 0.526 in boys and 0.509 to 0.531 in girls. These findings suggest that a low chest-to-head circumference ratio may indicate children who are at risk for neurodevelopmental deficits. However, the ability to predict poor neurodevelopmental outcomes at three years of age is limited.
Collapse
Affiliation(s)
- Naw Awn J-P
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Keiko Yamasaki
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Naomi Mitsuda
- Department of Pediatrics, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Mikiya Fujieda
- Department of Pediatrics, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Nagamasa Maeda
- Department of Obstetrics and Gynecology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Narufumi Suganuma
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Nankoku, Japan
| |
Collapse
|
13
|
Jansakova K, Hill M, Celusakova H, Repiska G, Bicikova M, Macova L, Polonyiova K, Kopcikova M, Ostatnikova D. Steroidogenic pathway in girls diagnosed with autism spectrum disorders. PLoS One 2024; 19:e0312933. [PMID: 39636905 PMCID: PMC11620458 DOI: 10.1371/journal.pone.0312933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 10/15/2024] [Indexed: 12/07/2024] Open
Abstract
The diagnostic prevalence of autism spectrum disorders (ASD) shows boys to be more affected than girls. Due to this reason, there is a lack of research including and observing ASD girls. Present study was aimed to detect hormones of steroidogenesis pathway in prepubertal girls (n = 16) diagnosed with ASD and sex and age matched neurotypical controls (CTRL, n = 16). Collected plasma served for detection of conjugated and unconjugated steroids using gas chromatography tandem-mass spectrometry. We observed higher levels of steroids modulating ionotropic receptors, especially, GABAergic steroids and pregnenolone sulfate in ASD group. Concentration of many steroids throughout the pathway tend to be higher in ASD girls compared to CTRL. Pregnenolone and its isomers together with polar progestins and androstanes, i.e. sulfated steroids, were found to be higher in ASD group in comparison with CTRL group. Based on steroid product to precursor ratios, ASD group showed higher levels of sulfated/conjugated steroids suggesting higher sulfotransferase or lower steroid sulfatase activity and we also obtained data indicating lower activity of steroid 11β-hydroxylase compared to CTRL group despite higher corticosterone level observed in ASD. These findings need to be generalized in future studies to examine both genders and other age groups.
Collapse
Affiliation(s)
- Katarina Jansakova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Martin Hill
- Department of Steroid Hormones and Proteohormones, Institute of Endocrinology, Prague, Czech Republic
| | - Hana Celusakova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Gabriela Repiska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Marie Bicikova
- Department of Steroid Hormones and Proteohormones, Institute of Endocrinology, Prague, Czech Republic
| | - Ludmila Macova
- Department of Steroid Hormones and Proteohormones, Institute of Endocrinology, Prague, Czech Republic
| | - Katarína Polonyiova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Mária Kopcikova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Daniela Ostatnikova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| |
Collapse
|
14
|
Wang K, Zhang S, Wang Y, Wu X, Wen L, Meng T, Jin X, Li S, Hong Y, Ke J, Xu Y, Yuan H, Hu F. Taprenepag restores maternal-fetal interface homeostasis for the treatment of neurodevelopmental disorders. J Neuroinflammation 2024; 21:307. [PMID: 39609821 PMCID: PMC11603931 DOI: 10.1186/s12974-024-03300-7] [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: 06/12/2024] [Accepted: 11/16/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND AND PURPOSE Neurodevelopmental disorders (NDDs) are characterized by abnormalities in brain development and neurobehaviors, including autism. The maternal-fetal interface (MFI) is a highly specialized tissue through which maternal factors affect fetal brain development. However, limited research exists on restoring and maintaining MFI homeostasis and its potential impact on NDDs. This study explores the role of placental indoleamine 2,3-dioxygenase (IDO-1) in MFI homeostasis and fetal brain development. EXPERIMENTAL APPROACH The maternal-fetal barrier was disrupted by sodium valproate (VPA) in pregnant mice, whose offspring show typical autism-like behaviors. Ultrastructural analysis and flow cytometric analysis were conducted to observe the morphological and immune system changes. Behavioral tests and immunofluorescence staining was used to investigate the ability and mechanism of taprenepag to alleviate the abnormal behaviors of VPA-exposed offspring and normalize the development of serotonergic neurons. KEY RESULTS In VPA-exposed pregnant mice, the downregulation of IDO-1 led to maternal immune overactivation and disruption of maternal-fetal barrier, resulting in excessive 5-HT synthesis in the placenta. This process disrupted the development of the serotonergic neuronal system in the offspring, resulting in impaired development of serotonergic neurons, thalamocortical axons, and NDDs in the progeny. However, a single injection of taprenepag at E13.5 ultimately upregulated placental IDO-1 through amplifying the positive feedback loop COX-2/PGE2/PTGER-2/IDO-1 and abolished these alterations. CONCLUSION Taprenepag improved autism-like behaviors in the offspring of VPA-exposed mice by addressing placental IDO-1 downregulation. This study highlights the potential of targeting IDO-1 to mitigate MFI disruption and NDD development.
Collapse
Affiliation(s)
- Kai Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Shufen Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Xiaomei Wu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Lijuan Wen
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, PR China
| | - Tingting Meng
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Xiangyu Jin
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, PR China
| | - Sufen Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yiling Hong
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Jia Ke
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yichong Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, PR China.
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China.
| |
Collapse
|
15
|
Pavy CL, Shaw JC, Dyson RM, Palliser HK, Moloney RA, Sixtus RP, Berry MJ, Hirst JJ. Ganaxolone Therapy After Preterm Birth Restores Cerebellar Oligodendrocyte Maturation and Myelination in Guinea Pigs. Dev Psychobiol 2024; 66:e22554. [PMID: 39378309 DOI: 10.1002/dev.22554] [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: 03/26/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 10/10/2024]
Abstract
The postnatal environment is challenging for the preterm neonate with exposure to hypoxic and excitotoxic events, amplified by premature loss of placentally derived neurosteroids. Between preterm birth and term equivalent age (TEA), cerebellar development continues despite these challenges. We hypothesize that neurosteroid replacement therapy during this time will support optimal cerebellar development. Guinea pig sows delivered at term (∼69 days gestation) or were induced to deliver preterm (∼62 days), with preterm pups receiving ganaxolone or vehicle until TEA. Postnatal assessments comprised salivary cortisol (corrected postnatal age [CPA] 0, 7, 38), behavioral analysis (CPA7, 38), and tissue collection (CPA0 and CPA40). Neurodevelopmental markers (MBP, Olig2, and NeuN) were assessed in the cerebellum by immunohistochemistry, whereas RT-PCR was utilized to investigate key inhibitory/excitatory pathways and oligodendrocyte lineage markers. Following preterm birth, there was evidence of a hyperactive phenotype, increased salivary cortisol concentrations, and impaired myelination and oligodendrocyte maturation at the protein level. mRNA expressions of key inhibitory/excitatory pathways and myelin stability were also altered following preterm birth. Importantly, we showed that neurosteroid replacement therapy returns cerebellar development and behavior toward a term-like phenotype. Therefore, ganaxolone may reduce the vulnerability of the cerebellum to postnatal challenges arising from preterm birth.
Collapse
Affiliation(s)
- Carlton L Pavy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Julia C Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Rebecca M Dyson
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
- Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Roisin A Moloney
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Ryan P Sixtus
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
- Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Mary J Berry
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
- Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
16
|
Wu Y, Su K, Zhang Y, Liang L, Wang F, Chen S, Gao L, Zheng Q, Li C, Su Y, Mao Y, Zhu S, Chai C, Lan Q, Zhai M, Jin X, Zhang J, Xu X, Zhang Y, Gao Y, Huang H. A spatiotemporal transcriptomic atlas of mouse placentation. Cell Discov 2024; 10:110. [PMID: 39438452 PMCID: PMC11496649 DOI: 10.1038/s41421-024-00740-6] [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: 01/29/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
The placenta, a temporary but essential organ for gestational support, undergoes intricate morphological and functional transformations throughout gestation. However, the spatiotemporal patterns of gene expression underlying placentation remain poorly understood. Utilizing Stereo-seq, we constructed a Mouse Placentation Spatiotemporal Transcriptomic Atlas (MPSTA) spanning from embryonic day (E) 7.5 to E14.5, which includes the transcriptomes of large trophoblast cells that were not captured in previous single-cell atlases. We defined four distinct strata of the ectoplacental cone, an early heterogeneous trophectoderm structure, and elucidated the spatial trajectory of trophoblast differentiation during early postimplantation stages before E9.5. Focusing on the labyrinth region, the interface of nutrient exchange in the mouse placenta, our spatiotemporal ligand-receptor interaction analysis unveiled pivotal modulators essential for trophoblast development and placental angiogenesis. We also found that paternally expressed genes are exclusively enriched in the placenta rather than in the decidual regions, including a cluster of genes enriched in endothelial cells that may function in placental angiogenesis. At the invasion front, we identified interface-specific transcription factor regulons, such as Atf3, Jun, Junb, Stat6, Mxd1, Maff, Fos, and Irf7, involved in gestational maintenance. Additionally, we revealed that maternal high-fat diet exposure preferentially affects this interface, exacerbating inflammatory responses and disrupting angiogenic homeostasis. Collectively, our findings furnish a comprehensive, spatially resolved atlas that offers valuable insights and benchmarks for future explorations into placental morphogenesis and pathology.
Collapse
Affiliation(s)
- Yanting Wu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
| | - Kaizhen Su
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Zhang
- BGI Research, Shenzhen, Guangdong, China
- Shanxi Medical University - BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Langchao Liang
- BGI Research, Qingdao, Shandong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fei Wang
- BGI Research, Shenzhen, Guangdong, China
| | - Siyue Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Ling Gao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Qiutong Zheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Cheng Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yunfei Su
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Simeng Zhu
- Department of Cardiology, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaochao Chai
- BGI Research, Qingdao, Shandong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qing Lan
- BGI Research, Shenzhen, Guangdong, China
| | - Man Zhai
- BGI Research, Shenzhen, Guangdong, China
| | - Xin Jin
- BGI Research, Shenzhen, Guangdong, China
| | - Jinglan Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Xun Xu
- BGI Research, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, Guangdong, China
| | - Yu Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
| | - Ya Gao
- BGI Research, Shenzhen, Guangdong, China.
- Shanxi Medical University - BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
- Shenzhen Engineering Laboratory for Birth Defects Screening, BGI Research, Shenzhen, Guangdong, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
17
|
Scheggi S, Concas L, Corsi S, Carta M, Melis M, Frau R. Expanding the therapeutic potential of neuro(active)steroids: a promising strategy for hyperdopaminergic behavioral phenotypes. Neurosci Biobehav Rev 2024; 164:105842. [PMID: 39103066 DOI: 10.1016/j.neubiorev.2024.105842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024]
Abstract
Imbalances in dopamine activity significantly contribute to the pathophysiology of several neuropsychiatric disorders, including addiction, ADHD, schizophrenia, impulse control disorders, and Parkinson's Disease. Neuro(active)steroids, comprising endogenous steroids that finely modulate neuronal activity, are considered crucial regulators of brain function and behavior, with implications in various physiological processes and pathological conditions. Specifically, subclasses of Neuro(active)steroids belonging to the 5α reductase pathway are prominently involved in brain disorders characterized by dopaminergic signaling imbalances. This review highlights the neuromodulatory effects of Neuro(active)steroids on the dopamine system and related aberrant behavioral phenotypes. We critically appraise the role of pregnenolone, progesterone, and allopregnanolone on dopamine signaling. Additionally, we discuss the impact of pharmacological interventions targeting 5α reductase activity in neuropsychiatric conditions characterized by excessive activation of the dopaminergic system, ranging from psychotic (endo)phenotypes and motor complications to decision-making problems and addiction.
Collapse
Affiliation(s)
- Simona Scheggi
- Dept. of Molecular and Developmental Medicine, University of Siena, Italy
| | - Luca Concas
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Sara Corsi
- Dept. of Developmental and Regenerative Neurobiology, Lund University, Sweden
| | - Manolo Carta
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Miriam Melis
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Roberto Frau
- Dept. Of Biomedical Sciences, University of Cagliari, Italy; Guy Everett Laboratory, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
18
|
Wang D, Zhao D, Wang W, Hu F, Cui M, Liu J, Meng F, Liu C, Qiu C, Liu D, Xu Z, Wang Y, Zhang Y, Li W, Li C. How do lateral septum projections to the ventral CA1 influence sociability? Neural Regen Res 2024; 19:1789-1801. [PMID: 38103246 PMCID: PMC10960288 DOI: 10.4103/1673-5374.389304] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/10/2023] [Accepted: 08/02/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00033/figure1/v/2023-12-16T180322Z/r/image-tiff Social dysfunction is a risk factor for several neuropsychiatric illnesses. Previous studies have shown that the lateral septum (LS)-related pathway plays a critical role in mediating social behaviors. However, the role of the connections between the LS and its downstream brain regions in social behaviors remains unclear. In this study, we conducted a three-chamber test using electrophysiological and chemogenetic approaches in mice to determine how LS projections to ventral CA1 (vCA1) influence sociability. Our results showed that gamma-aminobutyric acid (GABA)-ergic neurons were activated following social experience, and that social behaviors were enhanced by chemogenetic modulation of these neurons. Moreover, LS GABAergic neurons extended their functional neural connections via vCA1 glutamatergic pyramidal neurons, and regulating LSGABA→vCA1Glu neural projections affected social behaviors, which were impeded by suppressing LS-projecting vCA1 neuronal activity or inhibiting GABAA receptors in vCA1. These findings support the hypothesis that LS inputs to the vCA1 can control social preferences and social novelty behaviors. These findings provide new insights regarding the neural circuits that regulate sociability.
Collapse
Affiliation(s)
- Dan Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Di Zhao
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Wentao Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Fengai Hu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Minghu Cui
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Jing Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Fantao Meng
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Cuilan Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Changyun Qiu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Dunjiang Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Zhicheng Xu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Yameng Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Yu Zhang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- College of Nursing, Binzhou Medical University, Binzhou, Shandong Province, China
| | - Wei Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Chen Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| |
Collapse
|
19
|
Gaynor JW, Moldenhauer JS, Zullo EE, Burnham NB, Gerdes M, Bernbaum JC, D’Agostino JA, Linn RL, Klepczynski B, Randazzo I, Gionet G, Choi GH, Karaj A, Russell WW, Zackai EH, Johnson MP, Gebb JS, Soni S, DeBari SE, Szwast AL, Ahrens-Nicklas RC, Drivas TG, Jacobwitz M, Licht DJ, Vossough A, Nicolson SC, Spray TL, Rychik J, Putt ME. Progesterone for Neurodevelopment in Fetuses With Congenital Heart Defects: A Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2412291. [PMID: 38805228 PMCID: PMC11134212 DOI: 10.1001/jamanetworkopen.2024.12291] [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: 01/16/2024] [Accepted: 03/08/2024] [Indexed: 05/29/2024] Open
Abstract
Importance Neurodevelopmental outcomes for children with congenital heart defects (CHD) have improved minimally over the past 20 years. Objectives To assess the feasibility and tolerability of maternal progesterone therapy as well as the magnitude of the effect on neurodevelopment for fetuses with CHD. Design, Setting, and Participants This double-blinded individually randomized parallel-group clinical trial of vaginal natural progesterone therapy vs placebo in participants carrying fetuses with CHD was conducted between July 2014 and November 2021 at a quaternary care children's hospital. Participants included maternal-fetal dyads where the fetus had CHD identified before 28 weeks' gestational age and was likely to need surgery with cardiopulmonary bypass in the neonatal period. Exclusion criteria included a major genetic or extracardiac anomaly other than 22q11 deletion syndrome and known contraindication to progesterone. Statistical analysis was performed June 2022 to April 2024. Intervention Participants were 1:1 block-randomized to vaginal progesterone or placebo by diagnosis: hypoplastic left heart syndrome (HLHS), transposition of the great arteries (TGA), and other CHD diagnoses. Treatment was administered twice daily between 28 and up to 39 weeks' gestational age. Main Outcomes and Measures The primary outcome was the motor score of the Bayley Scales of Infant and Toddler Development-III; secondary outcomes included language and cognitive scales. Exploratory prespecified subgroups included cardiac diagnosis, fetal sex, genetic profile, and maternal fetal environment. Results The 102 enrolled fetuses primarily had HLHS (n = 52 [50.9%]) and TGA (n = 38 [37.3%]), were more frequently male (n = 67 [65.7%]), and without genetic anomalies (n = 61 [59.8%]). The mean motor score differed by 2.5 units (90% CI, -1.9 to 6.9 units; P = .34) for progesterone compared with placebo, a value not statistically different from 0. Exploratory subgroup analyses suggested treatment heterogeneity for the motor score for cardiac diagnosis (P for interaction = .03) and fetal sex (P for interaction = .04), but not genetic profile (P for interaction = .16) or maternal-fetal environment (P for interaction = .70). Conclusions and Relevance In this randomized clinical trial of maternal progesterone therapy, the overall effect was not statistically different from 0. Subgroup analyses suggest heterogeneity of the response to progesterone among CHD diagnosis and fetal sex. Trial Registration ClinicalTrials.gov Identifier: NCT02133573.
Collapse
Affiliation(s)
- J. William Gaynor
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Julie S. Moldenhauer
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Erin E. Zullo
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Nancy B. Burnham
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Marsha Gerdes
- Department of Psychology, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Judy C. Bernbaum
- Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jo Ann D’Agostino
- Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Rebecca L. Linn
- Division of Anatomic Pathology, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Brenna Klepczynski
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Isabel Randazzo
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Gabrielle Gionet
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Grace H. Choi
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Antoneta Karaj
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - William W. Russell
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Elaine H. Zackai
- Division of Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Mark P. Johnson
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Juliana S. Gebb
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Shelly Soni
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Suzanne E. DeBari
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Anita L. Szwast
- Division of Cardiology, Department of Pediatrics, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rebecca C. Ahrens-Nicklas
- Division of Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Theodore G. Drivas
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Marin Jacobwitz
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel J. Licht
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Arastoo Vossough
- Division of Radiology, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Susan C. Nicolson
- Division of Cardiac Anesthesia, Department of Anesthesia and Critical Medicine, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Thomas L. Spray
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jack Rychik
- Division of Cardiology, Department of Pediatrics, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mary E. Putt
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
20
|
Cáceres ARR, Cardone DA, Sanhueza MDLÁ, Bosch IM, Cuello-Carrión FD, Rodriguez GB, Scotti L, Parborell F, Halperin J, Laconi MR. Local effect of allopregnanolone in rat ovarian steroidogenesis, follicular and corpora lutea development. Sci Rep 2024; 14:6402. [PMID: 38493224 PMCID: PMC10944484 DOI: 10.1038/s41598-024-57102-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/14/2024] [Indexed: 03/18/2024] Open
Abstract
Allopregnanolone (ALLO) is a known neurosteroid and a progesterone metabolite synthesized in the ovary, CNS, PNS, adrenals and placenta. Its role in the neuroendocrine control of ovarian physiology has been studied, but its in situ ovarian effects are still largely unknown. The aims of this work were to characterize the effects of intrabursal ALLO administration on different ovarian parameters, and the probable mechanism of action. ALLO administration increased serum progesterone concentration and ovarian 3β-HSD2 while decreasing 20α-HSD mRNA expression. ALLO increased the number of atretic follicles and the number of positive TUNEL granulosa and theca cells, while decreasing positive PCNA immunostaining. On the other hand, there was an increase in corpora lutea diameter and PCNA immunostaining, whereas the count of TUNEL-positive luteal cells decreased. Ovarian angiogenesis and the immunohistochemical expression of GABAA receptor increased after ALLO treatment. To evaluate if the ovarian GABAA receptor was involved in these effects, we conducted a functional experiment with a specific antagonist, bicuculline. The administration of bicuculline restored the number of atretic follicles and the diameter of corpora lutea to normal values. These results show the actions of ALLO on the ovarian physiology of the female rat during the follicular phase, some of them through the GABAA receptor. Intrabursal ALLO administration alters several processes of the ovarian morpho-physiology of the female rat, related to fertility and oocyte quality.
Collapse
Affiliation(s)
- Antonella Rosario Ramona Cáceres
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Av. Ruiz Leal s/n Parque General San Martín, CP 5500, Mendoza, Argentina
- Facultad de Ingeniería y Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina
| | - Daniela Alejandra Cardone
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Av. Ruiz Leal s/n Parque General San Martín, CP 5500, Mendoza, Argentina
| | - María de Los Ángeles Sanhueza
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Av. Ruiz Leal s/n Parque General San Martín, CP 5500, Mendoza, Argentina
| | | | - Fernando Darío Cuello-Carrión
- Laboratorio de Oncología, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Mendoza, Argentina
| | | | - Leopoldina Scotti
- Ovarian Pathophysiology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Fernanda Parborell
- Ovarian Pathophysiology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Julia Halperin
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
| | - Myriam Raquel Laconi
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Av. Ruiz Leal s/n Parque General San Martín, CP 5500, Mendoza, Argentina.
- Facultad de Ingeniería y Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina.
| |
Collapse
|
21
|
Sze Y, Brunton PJ. How is prenatal stress transmitted from the mother to the fetus? J Exp Biol 2024; 227:jeb246073. [PMID: 38449331 DOI: 10.1242/jeb.246073] [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] [Indexed: 03/08/2024]
Abstract
Prenatal stress programmes long-lasting neuroendocrine and behavioural changes in the offspring. Often this programming is maladaptive and sex specific. For example, using a rat model of maternal social stress in late pregnancy, we have demonstrated that adult prenatally stressed male, but not prenatally stressed female offspring display heightened anxiety-like behaviour, whereas both sexes show hyperactive hypothalamo-pituitary-adrenal (HPA) axis responses to stress. Here, we review the current knowledge of the mechanisms underpinning dysregulated HPA axis responses, including evidence supporting a role for reduced neurosteroid-mediated GABAergic inhibitory signalling in the brains of prenatally stressed offspring. How maternal psychosocial stress is signalled from the mother to the fetuses is unclear. Direct transfer of maternal glucocorticoids to the fetuses is often considered to mediate the programming effects of maternal stress on the offspring. However, protective mechanisms including attenuated maternal stress responses and placental 11β-hydroxysteroid dehydrogenase-2 (which inactivates glucocorticoids) should limit materno-fetal glucocorticoid transfer during pregnancy. Moreover, a lack of correlation between maternal stress, circulating maternal glucocorticoid levels and circulating fetal glucocorticoid levels is reported in several studies and across different species. Therefore, here we interrogate the evidence for a role for maternal glucocorticoids in mediating the effects of maternal stress on the offspring and consider the evidence for alternative mechanisms, including an indirect role for glucocorticoids and the contribution of changes in the placenta in signalling the stress status of the mother to the fetus.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - Paula J Brunton
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
- Zhejiang University-University of Edinburgh Joint Institute, Haining, Zhejiang 314400, P.R. China
| |
Collapse
|
22
|
Jaarsma D, Birkisdóttir MB, van Vossen R, Oomen DWGD, Akhiyat O, Vermeij WP, Koekkoek SKE, De Zeeuw CI, Bosman LWJ. Different Purkinje cell pathologies cause specific patterns of progressive gait ataxia in mice. Neurobiol Dis 2024; 192:106422. [PMID: 38286390 DOI: 10.1016/j.nbd.2024.106422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 01/31/2024] Open
Abstract
Gait ataxia is one of the most common and impactful consequences of cerebellar dysfunction. Purkinje cells, the sole output neurons of the cerebellar cortex, are often involved in the underlying pathology, but their specific functions during locomotor control in health and disease remain obfuscated. We aimed to describe the effect of gradual adult-onset Purkinje cell degeneration on gaiting patterns in mice, and to determine whether two different mechanisms that both lead to Purkinje cell degeneration cause different patterns in the development of gait ataxia. Using the ErasmusLadder together with a newly developed limb detection algorithm and machine learning-based classification, we subjected mice to a challenging locomotor task with detailed analysis of single limb parameters, intralimb coordination and whole-body movement. We tested two Purkinje cell-specific mouse models, one involving stochastic cell death due to impaired DNA repair mechanisms (Pcp2-Ercc1-/-), the other carrying the mutation that causes spinocerebellar ataxia type 1 (Pcp2-ATXN1[82Q]). Both mouse models showed progressive gaiting deficits, but the sequence with which gaiting parameters deteriorated was different between mouse lines. Our longitudinal approach revealed that gradual loss of Purkinje cell function can lead to a complex pattern of loss of function over time, and that this pattern depends on the specifics of the pathological mechanisms involved. We hypothesize that this variability will also be present in disease progression in patients, and that our findings will facilitate the study of therapeutic interventions in mice, as subtle changes in locomotor abilities can be quantified by our methods.
Collapse
Affiliation(s)
- Dick Jaarsma
- Department of Neuroscience, Erasmus MC, 3015 CA, Rotterdam, the Netherlands.
| | - Maria B Birkisdóttir
- Department of Neuroscience, Erasmus MC, 3015 CA, Rotterdam, the Netherlands; Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, the Netherlands
| | - Randy van Vossen
- Department of Neuroscience, Erasmus MC, 3015 CA, Rotterdam, the Netherlands
| | - Demi W G D Oomen
- Department of Neuroscience, Erasmus MC, 3015 CA, Rotterdam, the Netherlands
| | - Oussama Akhiyat
- Department of Neuroscience, Erasmus MC, 3015 CA, Rotterdam, the Netherlands
| | - Wilbert P Vermeij
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, the Netherlands; Oncode Institute, 3521 AL, Utrecht, the Netherlands
| | | | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, 3015 CA, Rotterdam, the Netherlands; Netherlands Institute for Neuroscience, Royal Dutch Academy of Arts & Science, 1105 BA, Amsterdam, the Netherlands
| | - Laurens W J Bosman
- Department of Neuroscience, Erasmus MC, 3015 CA, Rotterdam, the Netherlands.
| |
Collapse
|
23
|
Potenzieri A, Uccella S, Preiti D, Pisoni M, Rosati S, Lavarello C, Bartolucci M, Debellis D, Catalano F, Petretto A, Nobili L, Fellin T, Tucci V, Ramenghi LA, Savardi A, Cancedda L. Early IGF-1 receptor inhibition in mice mimics preterm human brain disorders and reveals a therapeutic target. SCIENCE ADVANCES 2024; 10:eadk8123. [PMID: 38427732 PMCID: PMC10906931 DOI: 10.1126/sciadv.adk8123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Besides recent advances in neonatal care, preterm newborns still develop sex-biased behavioral alterations. Preterms fail to receive placental insulin-like growth factor-1 (IGF-1), a major fetal growth hormone in utero, and low IGF-1 serum levels correlate with preterm poor neurodevelopmental outcomes. Here, we mimicked IGF-1 deficiency of preterm newborns in mice by perinatal administration of an IGF-1 receptor antagonist. This resulted in sex-biased brain microstructural, functional, and behavioral alterations, resembling those of ex-preterm children, which we characterized performing parallel mouse/human behavioral tests. Pharmacological enhancement of GABAergic tonic inhibition by the U.S. Food and Drug Administration-approved drug ganaxolone rescued functional/behavioral alterations in mice. Establishing an unprecedented mouse model of prematurity, our work dissects the mechanisms at the core of abnormal behaviors and identifies a readily translatable therapeutic strategy for preterm brain disorders.
Collapse
Affiliation(s)
- Alberto Potenzieri
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
- Università degli Studi di Genova, via Balbi, 5, 16126 Genoa, Italy
| | - Sara Uccella
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Deborah Preiti
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Matteo Pisoni
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Silvia Rosati
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Chiara Lavarello
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Lino Nobili
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Valter Tucci
- Genetics and Epigenetics of Behavior (GEB) Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Luca A. Ramenghi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| |
Collapse
|
24
|
Zhang J, Zhang L, Gongol B, Hayes J, Borowsky A, Bailey-Serres J, Girke T. spatialHeatmap: visualizing spatial bulk and single-cell assays in anatomical images. NAR Genom Bioinform 2024; 6:lqae006. [PMID: 38312938 PMCID: PMC10836942 DOI: 10.1093/nargab/lqae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/14/2023] [Accepted: 01/18/2024] [Indexed: 02/06/2024] Open
Abstract
Visualizing spatial assay data in anatomical images is vital for understanding biological processes in cell, tissue, and organ organizations. Technologies requiring this functionality include traditional one-at-a-time assays, and bulk and single-cell omics experiments, including RNA-seq and proteomics. The spatialHeatmap software provides a series of powerful new methods for these needs, and allows users to work with adequately formatted anatomical images from public collections or custom images. It colors the spatial features (e.g. tissues) annotated in the images according to the measured or predicted abundance levels of biomolecules (e.g. mRNAs) using a color key. This core functionality of the package is called a spatial heatmap plot. Single-cell data can be co-visualized in composite plots that combine spatial heatmaps with embedding plots of high-dimensional data. The resulting spatial context information is essential for gaining insights into the tissue-level organization of single-cell data, or vice versa. Additional core functionalities include the automated identification of biomolecules with spatially selective abundance patterns and clusters of biomolecules sharing similar abundance profiles. To appeal to both non-expert and computational users, spatialHeatmap provides a graphical and a command-line interface, respectively. It is distributed as a free, open-source Bioconductor package (https://bioconductor.org/packages/spatialHeatmap) that users can install on personal computers, shared servers, or cloud systems.
Collapse
Affiliation(s)
- Jianhai Zhang
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| | - Le Zhang
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| | - Brendan Gongol
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| | - Jordan Hayes
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| | - Alexander T Borowsky
- Center for Plant Cell Biology, Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Julia Bailey-Serres
- Center for Plant Cell Biology, Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Thomas Girke
- Institute for Integrative Genome Biology, Department of Botany and Plant Sciences, 1207F Genomics Building, University of California, Riverside, CA 92521, USA
| |
Collapse
|
25
|
Newman J, Tong X, Tan A, Yeasky T, De Paiva VN, Presicce P, Kannan PS, Williams K, Damianos A, Tamase Newsam M, Benny MK, Wu S, Young KC, Miller LA, Kallapur SG, Chougnet CA, Jobe AH, Brambilla R, Schmidt AF. Chorioamnionitis accelerates granule cell and oligodendrocyte maturation in the cerebellum of preterm nonhuman primates. J Neuroinflammation 2024; 21:16. [PMID: 38200558 PMCID: PMC10777625 DOI: 10.1186/s12974-024-03012-y] [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: 07/03/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Preterm birth is often associated with chorioamnionitis and leads to increased risk of neurodevelopmental disorders, such as autism. Preterm birth can lead to cerebellar underdevelopment, but the mechanisms of disrupted cerebellar development in preterm infants are not well understood. The cerebellum is consistently affected in people with autism spectrum disorders, showing reduction of Purkinje cells, decreased cerebellar grey matter, and altered connectivity. METHODS Preterm rhesus macaque fetuses were exposed to intra-amniotic LPS (1 mg, E. coli O55:B5) at 127 days (80%) gestation and delivered by c-section 5 days after injections. Maternal and fetal plasma were sampled for cytokine measurements. Chorio-decidua was analyzed for immune cell populations by flow cytometry. Fetal cerebellum was sampled for histology and molecular analysis by single-nuclei RNA-sequencing (snRNA-seq) on a 10× chromium platform. snRNA-seq data were analyzed for differences in cell populations, cell-type specific gene expression, and inferred cellular communications. RESULTS We leveraged snRNA-seq of the cerebellum in a clinically relevant rhesus macaque model of chorioamnionitis and preterm birth, to show that chorioamnionitis leads to Purkinje cell loss and disrupted maturation of granule cells and oligodendrocytes in the fetal cerebellum at late gestation. Purkinje cell loss is accompanied by decreased sonic hedgehog signaling from Purkinje cells to granule cells, which show an accelerated maturation, and to oligodendrocytes, which show accelerated maturation from pre-oligodendrocytes into myelinating oligodendrocytes. CONCLUSION These findings suggest a role of chorioamnionitis on disrupted cerebellar maturation associated with preterm birth and on the pathogenesis of neurodevelopmental disorders among preterm infants.
Collapse
Affiliation(s)
- Josef Newman
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Xiaoying Tong
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - April Tan
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Toni Yeasky
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Vanessa Nunes De Paiva
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Pietro Presicce
- Division of Neonatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, USA
| | - Paranthaman S Kannan
- Division of Neonatology and Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Kevin Williams
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Andreas Damianos
- Division of Neonatology and Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Marione Tamase Newsam
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Merline K Benny
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Shu Wu
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Karen C Young
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA
| | - Lisa A Miller
- California National Primate Research Center, University of California, Davis, USA
| | - Suhas G Kallapur
- Division of Neonatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, USA
| | - Claire A Chougnet
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Alan H Jobe
- Division of Neonatology and Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, USA
| | - Augusto F Schmidt
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children's Hospital, Jackson Health System, Miami, USA.
- Batchelor Children's Research Institute, 1580 NW 10Th Ave, Room 348, Miami, FL, 33146, USA.
| |
Collapse
|
26
|
Baker MR, Lee AS, Rajadhyaksha AM. L-type calcium channels and neuropsychiatric diseases: Insights into genetic risk variant-associated genomic regulation and impact on brain development. Channels (Austin) 2023; 17:2176984. [PMID: 36803254 PMCID: PMC9980663 DOI: 10.1080/19336950.2023.2176984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/01/2023] [Indexed: 02/21/2023] Open
Abstract
Recent human genetic studies have linked a variety of genetic variants in the CACNA1C and CACNA1D genes to neuropsychiatric and neurodevelopmental disorders. This is not surprising given the work from multiple laboratories using cell and animal models that have established that Cav1.2 and Cav1.3 L-type calcium channels (LTCCs), encoded by CACNA1C and CACNA1D, respectively, play a key role in various neuronal processes that are essential for normal brain development, connectivity, and experience-dependent plasticity. Of the multiple genetic aberrations reported, genome-wide association studies (GWASs) have identified multiple single nucleotide polymorphisms (SNPs) in CACNA1C and CACNA1D that are present within introns, in accordance with the growing body of literature establishing that large numbers of SNPs associated with complex diseases, including neuropsychiatric disorders, are present within non-coding regions. How these intronic SNPs affect gene expression has remained a question. Here, we review recent studies that are beginning to shed light on how neuropsychiatric-linked non-coding genetic variants can impact gene expression via regulation at the genomic and chromatin levels. We additionally review recent studies that are uncovering how altered calcium signaling through LTCCs impact some of the neuronal developmental processes, such as neurogenesis, neuron migration, and neuron differentiation. Together, the described changes in genomic regulation and disruptions in neurodevelopment provide possible mechanisms by which genetic variants of LTCC genes contribute to neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Madelyn R. Baker
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, USA
| | - Andrew S. Lee
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Developmental Biology Program, Sloan Kettering Institute, New York, USA
| | - Anjali M. Rajadhyaksha
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, USA
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, USA
| |
Collapse
|
27
|
Jo S, Alejandro EU. RISING STARS: Mechanistic insights into maternal-fetal cross talk and islet beta-cell development. J Endocrinol 2023; 259:e230069. [PMID: 37855321 PMCID: PMC10692651 DOI: 10.1530/joe-23-0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023]
Abstract
The metabolic health trajectory of an individual is shaped as early as prepregnancy, during pregnancy, and lactation period. Both maternal nutrition and metabolic health status are critical factors in the programming of offspring toward an increased propensity to developing type 2 diabetes in adulthood. Pancreatic beta-cells, part of the endocrine islets, which are nutrient-sensitive tissues important for glucose metabolism, are primed early in life (the first 1000 days in humans) with limited plasticity later in life. This suggests the high importance of the developmental window of programming in utero and early in life. This review will focus on how changes to the maternal milieu increase offspring's susceptibility to diabetes through changes in pancreatic beta-cell mass and function and discuss potential mechanisms by which placental-driven nutrient availability, hormones, exosomes, and immune alterations that may impact beta-cell development in utero, thereby affecting susceptibility to type 2 diabetes in adulthood.
Collapse
Affiliation(s)
- Seokwon Jo
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Emilyn U Alejandro
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
28
|
Fitzgerald E, Shen M, Yong HEJ, Wang Z, Pokhvisneva I, Patel S, O'Toole N, Chan SY, Chong YS, Chen H, Gluckman PD, Chan J, Lee PKM, Meaney MJ. Hofbauer cell function in the term placenta associates with adult cardiovascular and depressive outcomes. Nat Commun 2023; 14:7120. [PMID: 37963865 PMCID: PMC10645763 DOI: 10.1038/s41467-023-42300-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 10/05/2023] [Indexed: 11/16/2023] Open
Abstract
Pathological placental inflammation increases the risk for several adult disorders, but these mediators are also expressed under homeostatic conditions, where their contribution to adult health outcomes is unknown. Here we define an inflammation-related expression signature, primarily expressed in Hofbauer cells of the term placenta and use expression quantitative trait loci to create a polygenic score (PGS) predictive of its expression. Using this PGS in the UK Biobank we conduct a phenome-wide association study, followed by Mendelian randomization and identify protective, sex-dependent effects of the placental module on cardiovascular and depressive outcomes. Genes differentially regulated by intra-amniotic infection and preterm birth are over-represented within the module. We also identify aspirin as a putative modulator of this inflammation-related signature. Our data support a model where disruption of placental Hofbauer cell function, due to preterm birth or prenatal infection, contributes to the increased risk of depression and cardiovascular disease observed in these individuals.
Collapse
Affiliation(s)
- Eamon Fitzgerald
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, Canada.
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, Canada.
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Canada.
| | - Mojun Shen
- Singapore Institute for Clinical Sciences, Agency for Science, Technology & Research, Singapore, Singapore
| | - Hannah Ee Juen Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology & Research, Singapore, Singapore
| | - Zihan Wang
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Canada
| | - Irina Pokhvisneva
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Canada
| | - Sachin Patel
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Canada
| | - Nicholas O'Toole
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, Canada
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Canada
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology & Research, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology & Research, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Helen Chen
- KK Women's and Children's Hospital, Singapore, Singapore
- Duke-National University of Singapore, Singapore, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science, Technology & Research, Singapore, Singapore
- The University of Auckland, Auckland, New Zealand
| | - Jerry Chan
- KK Women's and Children's Hospital, Singapore, Singapore
- Duke-National University of Singapore, Singapore, Singapore
| | - Patrick Kia Ming Lee
- Brain - Body Initiative, Agency for Science, Technology & Research, Singapore, Singapore
| | - Michael J Meaney
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, Canada.
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Canada.
- Singapore Institute for Clinical Sciences, Agency for Science, Technology & Research, Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Brain - Body Initiative, Agency for Science, Technology & Research, Singapore, Singapore.
| |
Collapse
|
29
|
Wortinger LA, Shadrin AA, Szabo A, Nerland S, Smelror RE, Jørgensen KN, Barth C, Andreou D, Thoresen M, Andreassen OA, Djurovic S, Ursini G, Agartz I. The impact of placental genomic risk for schizophrenia and birth asphyxia on brain development. Transl Psychiatry 2023; 13:343. [PMID: 37938559 PMCID: PMC10632427 DOI: 10.1038/s41398-023-02639-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
The placenta plays a role in fetal brain development, and pregnancy and birth complications can be signs of placental dysfunction. Birth asphyxia is associated with smaller head size and higher risk of developing schizophrenia (SZ), but whether birth asphyxia and placental genomic risk factors associated with SZ are related and how they might impact brain development is unclear. 433 adult patients with SZ and 870 healthy controls were clinically evaluated and underwent brain magnetic resonance imaging. Pregnancy and birth information were obtained from the Medical Birth Registry of Norway. Polygenic risk scores (PRS) from the latest genome-wide association study in SZ were differentiated into placental PRS (PlacPRS) and non-placental PRS. If the interaction between PRSs and birth asphyxia on case-control status was significant, neonatal head circumference (nHC) and adult intracranial volume (ICV) were further evaluated with these variables using multiple regression. PlacPRS in individuals with a history of birth asphyxia was associated with a higher likelihood of being a patient with SZ (t = 2.10, p = 0.018). We found a significant interaction between PlacPRS and birth asphyxia on nHC in the whole sample (t = -2.43, p = 0.008), with higher placental PRS for SZ associated with lower nHC in those with birth asphyxia. This relationship was specific to males (t = -2.71, p = 0.005) and also found with their adult ICV (t = -1.97, p = 0.028). These findings suggest that placental pathophysiology and birth asphyxia may affect early and late trajectories of brain development, particularly in males with a higher vulnerability to SZ. This knowledge might lead to new strategies of treatment and prevention in SZ.
Collapse
Affiliation(s)
- Laura A Wortinger
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway.
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Alexey A Shadrin
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Attila Szabo
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Stener Nerland
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Runar Elle Smelror
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kjetil Nordbø Jørgensen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry, Telemark Hospital, Skien, Norway
| | - Claudia Barth
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dimitrios Andreou
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Marianne Thoresen
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Neonatal Neuroscience, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Ole A Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gianluca Ursini
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
30
|
Bakalar D, Gavrilova O, Jiang SZ, Zhang HY, Roy S, Williams SK, Liu N, Wisser S, Usdin TB, Eiden LE. Constitutive and conditional deletion reveals distinct phenotypes driven by developmental versus neurotransmitter actions of the neuropeptide PACAP. J Neuroendocrinol 2023; 35:e13286. [PMID: 37309259 PMCID: PMC10620107 DOI: 10.1111/jne.13286] [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: 02/02/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 06/14/2023]
Abstract
Neuropeptides may exert trophic effects during development, and then neurotransmitter roles in the developed nervous system. One way to associate peptide-deficiency phenotypes with either role is first to assess potential phenotypes in so-called constitutive knockout mice, and then proceed to specify, regionally and temporally, where and when neuropeptide expression is required to prevent these phenotypes. We have previously demonstrated that the well-known constellation of behavioral and metabolic phenotypes associated with constitutive pituitary adenylate cyclase-activating peptide (PACAP) knockout mice are accompanied by transcriptomic alterations of two types: those that distinguish the PACAP-null phenotype from wild-type (WT) in otherwise quiescent mice (cPRGs), and gene induction that occurs in response to acute environmental perturbation in WT mice that do not occur in knockout mice (aPRGs). Comparing constitutive PACAP knockout mice to a variety of temporally and regionally specific PACAP knockouts, we show that the prominent hyperlocomotor phenotype is a consequence of early loss of PACAP expression, is associated with Fos overexpression in hippocampus and basal ganglia, and that a thermoregulatory effect previously shown to be mediated by PACAP-expressing neurons of medial preoptic hypothalamus is independent of PACAP expression in those neurons in adult mice. In contrast, PACAP dependence of weight loss/hypophagia triggered by restraint stress, seen in constitutive PACAP knockout mice, is phenocopied in mice in which PACAP is deleted after neuronal differentiation. Our results imply that PACAP has a prominent role as a trophic factor early in development determining global central nervous system characteristics, and in addition a second, discrete set of functions as a neurotransmitter in the fully developed nervous system that support physiological and psychological responses to stress.
Collapse
Affiliation(s)
- Dana Bakalar
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Kidney Disease- Intramural Research Program, Bethesda, Maryland, USA
| | - Sunny Z Jiang
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Hai-Ying Zhang
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Snehashis Roy
- Systems Neuroscience Imaging Resource, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Sarah K Williams
- Systems Neuroscience Imaging Resource, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Naili Liu
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Kidney Disease- Intramural Research Program, Bethesda, Maryland, USA
| | - Stephen Wisser
- Systems Neuroscience Imaging Resource, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Ted B Usdin
- Systems Neuroscience Imaging Resource, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Chen L, Xiong XY, Yao TT, Gui LN, Luo F, Du Y, Cheng Y. Blood exosome sensing via neuronal insulin-like growth factor-1 regulates autism-related phenotypes. Pharmacol Res 2023; 197:106965. [PMID: 37852341 DOI: 10.1016/j.phrs.2023.106965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023]
Abstract
The development and progression of autism spectrum disorder (ASD) is characterized by multiple complex molecular events, highlighting the importance of the prefrontal brain regions in this process. Exosomes are nanovesicles that play a critical role in intercellular communication. Peripheral systems influence brain function under both physiological and pathological conditions. We investigated whether this influence was mediated by the direct sensing of peripheral blood exosomes by brain cells. Administration of serum exosomes from rats with valproic acid-induced ASD resulted in ASD-related phenotypes in mice, whereas exosomes from normal rats did not exhibit such effects. RNA sequencing and bioinformatics analysis suggested that negative regulation of medial prefrontal cortex (mPFC) insulin-like growth factor 1 (IGF-1) by exosome-derived miR-29b-3p may contribute to these ASD-associated effects. Further evidence showed that miR-29b-3p-enriched exosomes crossed the blood-brain barrier to reach the mPFC, subsequently inducing the suppression of IGF-1 expression in neurons. Optogenetic activation of excitatory neurons in the mPFC improved behavioral abnormalities in exosome-treated mice. The addition of exogenous IGF-1 or inhibition of miR-29b-3p expression in the mPFC also rescued the ASD-related phenotypes in mice. Importantly, administration of miR-29b-3p-enriched serum exosomes from human donors with ASD into the mouse medial prefrontal cortex was sufficient to induce hallmark ASD behaviors. Together, our findings indicate that blood-brain cross-talk is crucial for ASD pathophysiology and that the brain may sense peripheral system changes through exosomes, which could serve as the basis for future neurological therapies.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Xi-Yue Xiong
- NHC Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Tong-Tong Yao
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Lue-Ning Gui
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Fan Luo
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China.
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China; College of Life and Environmental Sciences, Minzu University of China, Beijing, China; Institute of National Security, Minzu University of China, Beijing, China.
| |
Collapse
|
32
|
Ju L, Glastad KM, Sheng L, Gospocic J, Kingwell CJ, Davidson SM, Kocher SD, Bonasio R, Berger SL. Hormonal gatekeeping via the blood-brain barrier governs caste-specific behavior in ants. Cell 2023; 186:4289-4309.e23. [PMID: 37683635 PMCID: PMC10807403 DOI: 10.1016/j.cell.2023.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/10/2023] [Accepted: 08/01/2023] [Indexed: 09/10/2023]
Abstract
Here, we reveal an unanticipated role of the blood-brain barrier (BBB) in regulating complex social behavior in ants. Using scRNA-seq, we find localization in the BBB of a key hormone-degrading enzyme called juvenile hormone esterase (Jhe), and we show that this localization governs the level of juvenile hormone (JH3) entering the brain. Manipulation of the Jhe level reprograms the brain transcriptome between ant castes. Although ant Jhe is retained and functions intracellularly within the BBB, we show that Drosophila Jhe is naturally extracellular. Heterologous expression of ant Jhe into the Drosophila BBB alters behavior in fly to mimic what is seen in ants. Most strikingly, manipulation of Jhe levels in ants reprograms complex behavior between worker castes. Our study thus uncovers a remarkable, potentially conserved role of the BBB serving as a molecular gatekeeper for a neurohormonal pathway that regulates social behavior.
Collapse
Affiliation(s)
- Linyang Ju
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Karl M Glastad
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Lihong Sheng
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Janko Gospocic
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Urology and Institute of Neuropathology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Callum J Kingwell
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA
| | - Shawn M Davidson
- Lewis-Sigler Institute for Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Sarah D Kocher
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Roberto Bonasio
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Shelley L Berger
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Kanellopoulos AK, Costello S, Mainardi F, Koshibu K, Deoni S, Schneider N. Dynamic Interplay between Social Brain Development and Nutrient Intake in Young Children. Nutrients 2023; 15:3754. [PMID: 37686785 PMCID: PMC10490067 DOI: 10.3390/nu15173754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Myelination of the brain structures underlying social behavior in humans is a dynamic process that parallels the emergence of social-emotional development and social skills in early life. Of the many genetic and environmental factors regulating the myelination processes, nutrition is considered as a critical and modifiable early-life factor for establishing healthy social brain networks. However, the impact of nutrition on the longitudinal development of social brain myelination remains to be fully understood. This study examined the interplay between childhood nutrient intake and social brain development across the first 5 years of life. Myelin-sensitive neuroimaging and food-intake data were analyzed in 293 children, 0.5 to 5 years of age, and explored for dynamic patterns of nutrient-social brain myelin associations. We found three data-driven age windows with specific nutrient correlation patterns, 63 individual nutrient-myelin correlations, and six nutrient combinations with a statistically significant predictive value for social brain myelination. These results provide novel insights into the impact of specific nutrient intakes on early brain development, in particular social brain regions, and suggest a critical age-sensitive opportunity to impact these brain regions for potential longer-term improvements in socio-emotional development and related executive-function and critical-thinking skills.
Collapse
Affiliation(s)
- Alexandros K. Kanellopoulos
- Brain Health Department, Nestlé Institute of Health Sciences, Société des Produits Nestlé SA, Vers-Chez-les-Blanc, 1000 Lausanne, Switzerland
| | - Sarah Costello
- Brain Health Department, Nestlé Institute of Health Sciences, Société des Produits Nestlé SA, Vers-Chez-les-Blanc, 1000 Lausanne, Switzerland
| | - Fabio Mainardi
- Data Science Group, Nestlé Institute of Health Sciences, Société des Produits Nestlé SA, Vers-Chez-les-Blanc, 1000 Lausanne, Switzerland
| | - Kyoko Koshibu
- Brain Health Department, Nestlé Institute of Health Sciences, Société des Produits Nestlé SA, Vers-Chez-les-Blanc, 1000 Lausanne, Switzerland
| | - Sean Deoni
- Advanced Baby Imaging Lab, Rhode Island Hospital, 1 Hoppin Street, Providence, RI 20903, USA
- Department of Radiology, Warren Alpert Medical School of Brown University, 222 Richmond St., Providence, RI 02912, USA
- Spinn Neuroscience, Seattle, WA 98275, USA
| | - Nora Schneider
- Brain Health Department, Nestlé Institute of Health Sciences, Société des Produits Nestlé SA, Vers-Chez-les-Blanc, 1000 Lausanne, Switzerland
| |
Collapse
|
34
|
Zhu Y, Zhang Y, Jin Y, Jin H, Huang K, Tong J, Gan H, Rui C, Lv J, Wang X, Wang Q, Tao F. Identification and prediction model of placenta-brain axis genes associated with neurodevelopmental delay in moderate and late preterm children. BMC Med 2023; 21:326. [PMID: 37633927 PMCID: PMC10464496 DOI: 10.1186/s12916-023-03023-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 08/07/2023] [Indexed: 08/28/2023] Open
Abstract
BACKGROUND Moderate and late preterm (MLPT) birth accounts for the vast majority of preterm births, which is a global public health problem. The association between MLPT and neurobehavioral developmental delays in children and the underlying biological mechanisms need to be further revealed. The "placenta-brain axis" (PBA) provides a new perspective for gene regulation and risk prediction of neurodevelopmental delays in MLPT children. METHODS The authors performed multivariate logistic regression models between MLPT and children's neurodevelopmental outcomes, using data from 129 MLPT infants and 3136 full-term controls from the Ma'anshan Birth Cohort (MABC). Furthermore, the authors identified the abnormally regulated PBA-related genes in MLPT placenta by bioinformatics analysis of RNA-seq data and RT-qPCR verification on independent samples. Finally, the authors established the prediction model of neurodevelopmental delay in children with MLPT using multiple machine learning models. RESULTS The authors found an increased risk of neurodevelopmental delay in children with MLPT at 6 months, 18 months, and 48 months, especially in boys. Further verification showed that APOE and CST3 genes were significantly correlated with the developmental levels of gross-motor domain, fine-motor domain, and personal social domain in 6-month-old male MLPT children. CONCLUSIONS These findings suggested that there was a sex-specific association between MLPT and neurodevelopmental delays. Moreover, APOE and CST3 were identified as placental biomarkers. The results provided guidance for the etiology investigation, risk prediction, and early intervention of neurodevelopmental delays in children with MLPT.
Collapse
Affiliation(s)
- Yumin Zhu
- Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China.
| | - Yimin Zhang
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Yunfan Jin
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Heyue Jin
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Kun Huang
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Juan Tong
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Hong Gan
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Chen Rui
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Jia Lv
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xianyan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Qu'nan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| | - Fangbiao Tao
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
35
|
Anamthathmakula P, Shallie PD, Nayak N, Dhal S, Vivian JL, Mor G, Soares MJ, Nayak NR. Variable Cre Recombination Efficiency in Placentas of Cyp19-Cre ROSA mT/mG Transgenic Mice. Cells 2023; 12:2096. [PMID: 37626906 PMCID: PMC10453067 DOI: 10.3390/cells12162096] [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] [Received: 06/28/2023] [Revised: 08/06/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The aromatase-Cre recombinase (Cyp19-Cre) transgenic mouse model has been extensively used for placenta-specific gene inactivation. In a pilot study, we observed unexpected phenotypes using this mouse strain, which prompted an extensive characterization of Cyp19-Cre placental phenotypes using ROSAmT/mG transgenic reporter mice. The two strains were mated to generate bi-transgenic Cyp19-Cre;ROSAmT/mG mice following a standard transgenic breeding scheme, and placental and fetal tissues were analyzed on embryonic day 17.5. Both maternal and paternal Cre inheritance were analyzed by mating the respective Cyp19-Cre and ROSAmT/mG males and females. The genotype results showed the expected percentage of Cyp19-Cre;ROSAmT/mG fetuses (73%) and Cre mRNA was expressed in all of the Cyp19-Cre placentas. However, surprisingly, only about 50% of the Cyp19-Cre;ROSAmT/mG placentas showed Cre-mediated recombinase activity as demonstrated by placental enhanced green fluorescent protein (EGFP) expression. Further genetic excision analysis of the placentas revealed consistent results showing the absence of excision of the tdTomato in all of the Cyp19-Cre;ROSAmT/mG placentas lacking EGFP expression. Moreover, among the EGFP-expressing placentas, there was wide variability in recombination efficiency, even in placentas from the same litter, leading to a mosaic pattern of EGFP expression in different zones and cell types of the placentas. In addition, we observed a significantly higher percentage of Cre recombination activity in placentas with maternal Cre inheritance. Our results show frequent mosaicism, inconsistent recombination activity, and parent-of-origin effects in placentas from Cyp19-Cre;ROSAmT/mG mice, suggesting that tail-biopsy genotype results may not necessarily indicate the excision of floxed genes in Cyp19-Cre positive placentas. Thus, placenta-specific mutagenesis studies using the Cyp19-Cre model require extensive characterization and careful interpretation of the placental phenotypes for each floxed allele.
Collapse
Affiliation(s)
- Prashanth Anamthathmakula
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
- Department of Surgery, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Philemon D. Shallie
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
- Department of Surgery, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Neha Nayak
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Sabita Dhal
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Jay L. Vivian
- Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Michael J. Soares
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
| | - Nihar R. Nayak
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| |
Collapse
|
36
|
Boueid MJ, El-Hage O, Schumacher M, Degerny C, Tawk M. Zebrafish as an emerging model to study estrogen receptors in neural development. Front Endocrinol (Lausanne) 2023; 14:1240018. [PMID: 37664862 PMCID: PMC10469878 DOI: 10.3389/fendo.2023.1240018] [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: 06/14/2023] [Accepted: 07/27/2023] [Indexed: 09/05/2023] Open
Abstract
Estrogens induce several regulatory signals in the nervous system that are mainly mediated through estrogen receptors (ERs). ERs are largely expressed in the nervous system, yet the importance of ERs to neural development has only been elucidated over the last decades. Accumulating evidence shows a fundamental role for estrogens in the development of the central and peripheral nervous systems, hence, the contribution of ERs to neural function is now a growing area of research. The conservation of the structure of the ERs and their response to estrogens make the zebrafish an interesting model to dissect the role of estrogens in the nervous system. In this review, we highlight major findings of ER signaling in embryonic zebrafish neural development and compare the similarities and differences to research in rodents. We also discuss how the recent generation of zebrafish ER mutants, coupled with the availability of several transgenic reporter lines, its amenability to pharmacological studies and in vivo live imaging, could help us explore ER function in embryonic neural development.
Collapse
Affiliation(s)
| | | | | | | | - Marcel Tawk
- *Correspondence: Cindy Degerny, ; Marcel Tawk,
| |
Collapse
|
37
|
Kruger L, Lapehn S, Paquette A, Singh DK, MacDonald J, Bammler TK, Enquobahrie DA, Zhao Q, Mozhui K, Sathyanarayana S, Prasad B. Characterization of Xenobiotic and Steroid Disposition Potential of Human Placental Tissue and Cell Lines (BeWo, JEG-3, JAR, and HTR-8/SVneo) by Quantitative Proteomics. Drug Metab Dispos 2023; 51:1053-1063. [PMID: 37164652 PMCID: PMC10353074 DOI: 10.1124/dmd.123.001345] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/04/2023] [Accepted: 04/05/2023] [Indexed: 05/12/2023] Open
Abstract
The placenta is a fetal organ that performs critical functions to maintain pregnancy and support fetal development, including metabolism and transport of xenobiotics and steroids between the maternal-fetal unit. In vitro placenta models are used to study xenobiotic and steroid disposition, but how well these models recapitulate the human placenta is not well understood. We first characterized the abundance of proteins involved in xenobiotic and steroid disposition in human placental tissue. In pooled human placenta, the following xenobiotic and steroid disposition proteins were detected (highest to lowest), 1) enzymes: glutathione S-transferase P, carbonyl reductase 1, aldo-keto reductase 1B1, hydroxysteroid dehydrogenases (HSD3B1 and HSD11B1), aromatase, epoxide hydrolase 1 (EPHX1) and steryl-sulfatase, and 2) transporters: monocarboxylate transporters (MCT1 and 4), organic anion transporting polypeptide 2B1, organic anion transporter 4, and breast cancer resistance protein (BCRP). Then, the tissue proteomics data were compared with four placental cell lines (BeWo, JEG-3, JAR, and HTR-8/SVneo). The differential global proteomics analysis revealed that the tissue and cell lines shared 1420 cytosolic and 1186 membrane proteins. Although extravillous trophoblast and cytotrophoblast marker proteins were detected in all cell lines, only BeWo and JEG-3 cells expressed the syncytiotrophoblast marker, chorionic somatomammotropin hormone 1. BeWo and JEG-3 cells expressed most target proteins including aromatase, HSDs, EPHX1, MCT1, and BCRP. JEG-3 cells treated with commonly detected phthalates in human biofluids showed dysregulation of steroid pathways. The data presented here show that BeWo and JEG-3 cells are closer to the placental tissue for studying xenobiotic and steroid disposition. SIGNIFICANCE STATEMENT: This is the first study to compare proteomics data of human placental tissue and cell lines (BeWo, JAR, JEG-3, and HTR-8/SVneo). The placental cell line and tissue proteomes are vastly different, but BeWo and JEG-3 cells showed greater resemblance to the tissue in the expression of xenobiotic and steroid disposition proteins. These data will assist researchers to select an optimum cell model for mechanistic investigations on xenobiotic and steroid disposition in the placenta.
Collapse
Affiliation(s)
- Laken Kruger
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Samantha Lapehn
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Alison Paquette
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Dilip Kumar Singh
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - James MacDonald
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Theo K Bammler
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Daniel A Enquobahrie
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Qi Zhao
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Khyobeni Mozhui
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Sheela Sathyanarayana
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (L.K., D.K.S., B.P.); Centers for Developmental Biology and Regenerative Medicine (S.L., A.P.) and Child Health, Behavior and Development (S.S.), Seattle Children's Research Institute, Seattle, Washington; Departments of Pediatrics (A.P., S.S.), Environmental and Occupational Health Sciences (J.M., T.K.B., S.S.), and Epidemiology (D.A.E.), University of Washington, Seattle, Washington; and Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee (K.M., Q.Z.)
| |
Collapse
|
38
|
Vacher CM, Bonnin A, Mir IN, Penn AA. Editorial: Advances and perspectives in neuroplacentology. Front Endocrinol (Lausanne) 2023; 14:1206072. [PMID: 37274324 PMCID: PMC10236794 DOI: 10.3389/fendo.2023.1206072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Affiliation(s)
- Claire-Marie Vacher
- Department of Pediatrics, NewYork Presbyterian Hospital, New York, NY, United States
- Columbia University Irving Medical Center, Columbia University, New York, NY, United States
| | - Alexandre Bonnin
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Imran N. Mir
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Anna A. Penn
- Department of Pediatrics, NewYork Presbyterian Hospital, New York, NY, United States
- Columbia University Irving Medical Center, Columbia University, New York, NY, United States
| |
Collapse
|
39
|
Bruce MR, Couch ACM, Grant S, McLellan J, Ku K, Chang C, Bachman A, Matson M, Berman RF, Maddock RJ, Rowland D, Kim E, Ponzini MD, Harvey D, Taylor SL, Vernon AC, Bauman MD, Van de Water J. Altered behavior, brain structure, and neurometabolites in a rat model of autism-specific maternal autoantibody exposure. Mol Psychiatry 2023; 28:2136-2147. [PMID: 36973347 PMCID: PMC10575787 DOI: 10.1038/s41380-023-02020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023]
Abstract
Maternal immune dysregulation is a prenatal risk factor for autism spectrum disorder (ASD). Importantly, a clinically relevant connection exists between inflammation and metabolic stress that can result in aberrant cytokine signaling and autoimmunity. In this study we examined the potential for maternal autoantibodies (aAbs) to disrupt metabolic signaling and induce neuroanatomical changes in the brains of exposed offspring. To accomplish this, we developed a model of maternal aAb exposure in rats based on the clinical phenomenon of maternal autoantibody-related ASD (MAR-ASD). Following confirmation of aAb production in rat dams and antigen-specific immunoglobulin G (IgG) transfer to offspring, we assessed offspring behavior and brain structure longitudinally. MAR-ASD rat offspring displayed a reduction in pup ultrasonic vocalizations and a pronounced deficit in social play behavior when allowed to freely interact with a novel partner. Additionally, longitudinal in vivo structural magnetic resonance imaging (sMRI) at postnatal day 30 (PND30) and PND70, conducted in a separate cohort of animals, revealed sex-specific differences in total and regional brain volume. Treatment-specific effects by region appeared to converge on midbrain and cerebellar structures in MAR-ASD offspring. Simultaneously, in vivo 1H magnetic resonance spectroscopy (1H-MRS) data were collected to examine brain metabolite levels in the medial prefrontal cortex. Results showed that MAR-ASD offspring displayed decreased levels of choline-containing compounds and glutathione, accompanied by increased taurine compared to control animals. Overall, we found that rats exposed to MAR-ASD aAbs present with alterations in behavior, brain structure, and neurometabolites; reminiscent of findings observed in clinical ASD.
Collapse
Affiliation(s)
- Matthew R Bruce
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Amalie C M Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Simone Grant
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Janna McLellan
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Katherine Ku
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Christina Chang
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Angelica Bachman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Matthew Matson
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Robert F Berman
- Department of Neurological Surgery, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Douglas Rowland
- Center for Molecular and Genomic Imaging, University of California, Davis, CA, USA
| | - Eugene Kim
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Matthew D Ponzini
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Danielle Harvey
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Sandra L Taylor
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA.
- MIND Institute, University of California, Davis, CA, USA.
| |
Collapse
|
40
|
Tsompanidis A, Hampton S, Aydin E, Allison C, Holt R, Baron-Cohen S. Mini-puberty testosterone and infant autistic traits. Front Endocrinol (Lausanne) 2023; 14:1126023. [PMID: 37091846 PMCID: PMC10113441 DOI: 10.3389/fendo.2023.1126023] [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: 12/16/2022] [Accepted: 02/24/2023] [Indexed: 04/08/2023] Open
Abstract
Background Levels of steroid hormones in the first three months of life, a period referred to as 'mini-puberty', are one of the earliest physiological differences between typical males and females postnatally. Autistic traits also show consistent typical sex differences in later infancy, after the 18th month of life. Previous studies have shown prenatal testosterone is associated with later levels of autistic traits. Studies testing if postnatal testosterone levels are associated with autistic traits have reported null results. No studies to date have investigated mini-puberty longitudinally or tested for interactions with baseline sex differences or familial likelihood of autism. Methods The 'Cambridge Human Imaging and Longitudinal Development Study' (CHILD) is a prospective enriched cohort study in Cambridge, UK. It includes physiological measurements in early infancy, as well as neurodevelopmental follow-ups over the first two years of life. A subset of the cohort also includes children with a family history of autism (a diagnosed parent or sibling). Salivary testosterone levels were assessed at two time-points, just after the 2nd and 6th month of life. Autistic traits were measured using the Quantitative Checklist of Autism in Toddlers (Q-CHAT) when the children were 18 months of age. Results Salivary testosterone levels were significantly higher during 'mini-puberty' in the 2nd and 3rd month of life, compared to after the 6th month of life, in both males and females. There was no significant sex difference at either time-point. Log-transformed testosterone levels were not associated with autistic traits (Q-CHAT). There was no interaction effect with infant sex, autism family history or baseline testosterone levels after mini-puberty (at >6 months of age). Conclusion Both male and female infants have elevated levels of salivary testosterone during mini-puberty but in this relatively small sample this was not associated with their later autistic traits at 18 months or their family history of autism. This suggests that prenatal rather than postnatal testosterone levels are more relevant for understanding the causes of autism. Future studies should test these relationships in larger samples.
Collapse
Affiliation(s)
- Alex Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Hampton
- York Trials Unit, University of York, York, United Kingdom
| | - Ezra Aydin
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Carrie Allison
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Rosemary Holt
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
41
|
Andescavage N, Bullen T, Liggett M, Barnett SD, Kapse A, Kapse K, Ahmadzia H, Vezina G, Quistorff J, Lopez C, duPlessis A, Limperopoulos C. Impaired in vivo feto-placental development is associated with neonatal neurobehavioral outcomes. Pediatr Res 2023; 93:1276-1284. [PMID: 36335267 PMCID: PMC10147575 DOI: 10.1038/s41390-022-02340-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Fetal growth restriction (FGR) is a risk factor for neurodevelopmental problems, yet remains poorly understood. We sought to examine the relationship between intrauterine development and neonatal neurobehavior in pregnancies diagnosed with antenatal FGR. METHODS We recruited women with singleton pregnancies diagnosed with FGR and measured placental and fetal brain volumes using MRI. NICU Network Neurobehavioral Scale (NNNS) assessments were performed at term equivalent age. Associations between intrauterine volumes and neurobehavioral outcomes were assessed using generalized estimating equation models. RESULTS We enrolled 44 women diagnosed with FGR who underwent fetal MRI and 28 infants underwent NNNS assessments. Placental volumes were associated with increased self-regulation and decreased excitability; total brain, brainstem, cortical and subcortical gray matter (SCGM) volumes were positively associated with higher self-regulation; SCGM also was positively associated with higher quality of movement; increasing cerebellar volumes were positively associated with attention, decreased lethargy, non-optimal reflexes and need for special handling; brainstem volumes also were associated with decreased lethargy and non-optimal reflexes; cerebral and cortical white matter volumes were positively associated with hypotonicity. CONCLUSION Disrupted intrauterine growth in pregnancies complicated by antenatally diagnosed FGR is associated with altered neonatal neurobehavior. Further work to determine long-term neurodevelopmental impacts is warranted. IMPACT Fetal growth restriction is a risk factor for adverse neurodevelopment, but remains difficult to accurately identify. Intrauterine brain volumes are associated with infant neurobehavior. The antenatal diagnosis of fetal growth restriction is a risk factor for abnormal infant neurobehavior.
Collapse
Affiliation(s)
- Nickie Andescavage
- Division of Neonatology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
- Prenatal Pediatric Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Theresa Bullen
- School of Medicine, George Washington University, Washington, DC, USA
| | - Melissa Liggett
- Division of Psychology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Scott D Barnett
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Anushree Kapse
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Kushal Kapse
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Homa Ahmadzia
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, George Washington University, 2300 Eye St. NW, Washington, DC, 20037, USA
| | - Gilbert Vezina
- Division of Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
- Department of Radiology, George Washington University, 2300 Eye St. NW, Washington, DC, 20037, USA
| | - Jessica Quistorff
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Catherine Lopez
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Adre duPlessis
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
- Prenatal Pediatric Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
- Department of Pediatrics, George Washington University, 2300 Eye St. NW, Washington, DC, 20037, USA
| | - Catherine Limperopoulos
- Developing Brain Institute, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA.
- Department of Radiology, George Washington University, 2300 Eye St. NW, Washington, DC, 20037, USA.
- Department of Pediatrics, George Washington University, 2300 Eye St. NW, Washington, DC, 20037, USA.
| |
Collapse
|
42
|
Yang Y, Shen Y, Lin J, Dai S, Lu X, Xun G, Li Y, Wu R, Xia K, Luo X, Zhao J, Ou J. Association between history of miscarriage and autism spectrum disorder. Eur Arch Psychiatry Clin Neurosci 2023; 273:687-697. [PMID: 36251093 DOI: 10.1007/s00406-022-01494-6] [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: 02/22/2022] [Accepted: 09/13/2022] [Indexed: 11/03/2022]
Abstract
This case-control study was designed to examine the association between different types of miscarriage history and autism spectrum disorder (ASD), and determine whether the number of miscarriage history affects the risk of ASD. All of 2274 children with ASD and 1086 healthy controls were recruited. Sociodemographic and prenatal, perinatal, and neonatal characteristics were compared between the two groups. Multivariable logistic regression analyses were applied to investigate association between miscarriage history and ASD. Stratified analyses based on sex and types of miscarriages were similarly performed. History of miscarriage was potential risk factors for ASD ([aOR] = 2.919; 95% [CI] = 2.327-3.517). Stratified analyses revealed that induced ([aOR] = 2.763, 95% [CI] = 2.259-3.379) and spontaneous miscarriage history ([aOR] = 3.341, 95% [CI] = 1.939-4.820) were associated with high risk of ASD, respectively. A sex-biased ratio in the risk of ASD was observed between females ([aOR] = 3.049, 95% [CI] = 2.153-4.137) and males ([aOR] = 2.538, 95% [CI] = 1.978-3.251). Stratified analysis of induced miscarriage history revealed that only iatrogenic miscarriage history was associated with an increased risk ASD ([aOR] = 2.843, 95% [CI] = 1.534-4.268). Also, multiple spontaneous miscarriage histories ([aOR] = 1.836, 95% [CI] = 1.252-2.693) were associated with higher autism risk than one spontaneous miscarriages history ([aOR] = 3.016, 95% [CI] = 1.894-4.174). In conclusion, miscarriage history is related to an increased risk for ASD in offspring, which is affected by the types of miscarriage and sex of the fetus.
Collapse
Affiliation(s)
- Ye Yang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Yidong Shen
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jingjing Lin
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Si Dai
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Xiaozi Lu
- Qingdao Mental Health Center, Qingdao, 266034, Shandong, China
| | - Guanglei Xun
- Shandong Mental Health Center, 49 East Wenhua Road, Jinan, 250014, Shandong, China
| | - Yamin Li
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Renrong Wu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Kun Xia
- Center for Medical Genetics and School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Xuerong Luo
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jingping Zhao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jianjun Ou
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
43
|
"Prenatal programming of the brain: The maternal-fetal interface takes center stage". Brain Behav Immun 2023; 109:219-220. [PMID: 36736542 DOI: 10.1016/j.bbi.2023.01.019] [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: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
|
44
|
Ravaei A, Emanuele M, Nazzaro G, Fadiga L, Rubini M. Placental DNA methylation profile as predicting marker for autism spectrum disorder (ASD). Mol Med 2023; 29:8. [PMID: 36647002 PMCID: PMC9843962 DOI: 10.1186/s10020-022-00593-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that impairs normal brain development and socio-cognitive abilities. The pathogenesis of this condition points out the involvement of genetic and environmental factors during in-utero life. Placenta, as an interface tissue between mother and fetus, provides developing fetus requirements and exposes it to maternal environment as well. Therefore, the alteration of DNA methylation as epigenetic consequence of gene-environmental interaction in the placenta could shed light on ASD pathogenesis. In this study, we reviewed the current findings on placental methylation status and its association with ASD. Differentially methylated regions (DMRs) in ASD-developing placenta were found to be mainly enriched in ASD gene loci affecting synaptogenesis, microtubule dynamics, neurogenesis and neuritogenesis. In addition, non-genic DMRs in ASD-placenta proposes an alternative contributing mechanism for ASD development. Our study highlights the importance of placental DNA methylation signature as a biomarker for ASD prediction.
Collapse
Affiliation(s)
- Amin Ravaei
- grid.8484.00000 0004 1757 2064Medical Genetics Laboratory, Department of Neuroscience and Rehabilitation, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy
| | - Marco Emanuele
- grid.8484.00000 0004 1757 2064Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy ,grid.25786.3e0000 0004 1764 2907IIT@UniFe Center for Translational Neurophysiology of Speech and Communication (CTNSC), Istituto Italiano di Tecnologia, Ferrara, Italy
| | - Giovanni Nazzaro
- grid.8484.00000 0004 1757 2064Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy ,grid.25786.3e0000 0004 1764 2907IIT@UniFe Center for Translational Neurophysiology of Speech and Communication (CTNSC), Istituto Italiano di Tecnologia, Ferrara, Italy
| | - Luciano Fadiga
- grid.8484.00000 0004 1757 2064Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy ,grid.25786.3e0000 0004 1764 2907IIT@UniFe Center for Translational Neurophysiology of Speech and Communication (CTNSC), Istituto Italiano di Tecnologia, Ferrara, Italy
| | - Michele Rubini
- grid.8484.00000 0004 1757 2064Medical Genetics Laboratory, Department of Neuroscience and Rehabilitation, University of Ferrara, Via Fossato di Mortara 74, 44121 Ferrara, Italy
| |
Collapse
|
45
|
Maternal Dexamethasone Exposure Induces Sex-Specific Changes in Histomorphology and Redox Homeostasis of Rat Placenta. Int J Mol Sci 2022; 24:ijms24010540. [PMID: 36613982 PMCID: PMC9820254 DOI: 10.3390/ijms24010540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
As the mediator between the mother and fetus, the placenta allows the most appropriate environment and optimal fetal growth. The placenta of one sex sometimes has a greater ability over the other to respond to and protect against possible maternal insults. Here, we characterized sex differences in the placenta’s morphological features and antioxidant status following dexamethasone (Dx) exposure. Pregnant rats were exposed to Dx or saline. The placenta was histologically and stereologically analyzed. The activity of the antioxidant enzymes, lipid peroxides (TBARS), superoxide anion and nitric oxide (NO) was measured. The decrease in placental zone volumes was more pronounced (p < 0.05) in female placentas. The volume density of PCNA-immunopositive nuclei was reduced (p < 0.05) in both sexes. The reduced (p < 0.05) antioxidant enzyme activities, enhanced TBARS and NO concentration indicate that Dx exposure triggered oxidative stress in the placenta of both fetal sexes, albeit stronger in the placenta of female fetuses. In conclusion, maternal Dx treatment reduced the size and volume of placental zones, altered placental histomorphology, decreased cell proliferation and triggered oxidative stress; however, the placentas of female fetuses exerted more significant responses to the treatment effects. The reduced placental size most probably reduced the transport of nutrients and oxygen, thus resulting in the reduced weight of fetuses, similar in both sexes. The lesser ability of the male placenta to detect and react to maternal exposure to environmental challenges may lead to long-standing health effects.
Collapse
|
46
|
Marable CA, Roell K, Kuban K, O’Shea TM, Fry RC. Placental transcriptional signatures associated with cerebral white matter damage in the neonate. Front Neurosci 2022; 16:1017953. [PMID: 36389237 PMCID: PMC9650394 DOI: 10.3389/fnins.2022.1017953] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/04/2022] [Indexed: 09/10/2023] Open
Abstract
Cerebral white matter is the most common anatomic location of neonatal brain injury in preterm newborns. Factors that predispose preterm newborns to white matter damage are understudied. In relation to studies of the placenta-brain-axis, dysregulated placental gene expression may play a role in preterm brain damage given its implication in programming early life origins of disease, including neurological disorders. There is a critical need to investigate the relationships between the placental transcriptome and white matter damage in the neonate. In a cohort of extremely low gestational age newborns (ELGANs), we aimed to investigate the relationship between the placental transcriptome and white matter damage as assessed by neonatal cranial ultrasound studies (echolucency and/or ventriculomegaly). We hypothesized that genes involved in inflammatory processes would be more highly expressed in placentas of ELGANs who developed ultrasound-defined indicators of white matter damage. Relative to either form of white matter damage, 659 placental genes displayed altered transcriptional profiles. Of these white matter damage-associated genes, largely distinct patterns of gene expression were observed in the study (n = 415/659 genes). Specifically, 381 genes were unique to echolucency and 34 genes were unique to ventriculomegaly. Pathways involved in hormone disruption and metabolism were identified among the unique echolucency or ventriculomegaly genes. Interestingly, a common set of 244 genes or 37% of all genes was similarly dysregulated in the placenta relative to both echolucency and ventriculomegaly. For this common set of white matter damage-related genes, pathways involved in inflammation, immune response and apoptosis, were enriched. Among the white matter damage-associated genes are genes known to be involved in Autism Spectrum Disorder (ASD) and endocrine system disorders. These data highlight differential mRNA expression patterning in the placenta and provide insight into potential etiologic factors that may predispose preterm newborns to white matter damage. Future studies will build upon this work to include functional measures of neurodevelopment as well as measures of brain volume later in life.
Collapse
Affiliation(s)
- Carmen Amelia Marable
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kyle Roell
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Institute for Environmental Health Solutions, Gilling School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Karl Kuban
- Division of Pediatric Neurology, Department of Pediatrics, Boston University Medical Center, Boston, MA, United States
| | - T. Michael O’Shea
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Institute for Environmental Health Solutions, Gilling School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
47
|
Vidal MS, Lintao RCV, Severino MEL, Tantengco OAG, Menon R. Spontaneous preterm birth: Involvement of multiple feto-maternal tissues and organ systems, differing mechanisms, and pathways. Front Endocrinol (Lausanne) 2022; 13:1015622. [PMID: 36313741 PMCID: PMC9606232 DOI: 10.3389/fendo.2022.1015622] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Survivors of preterm birth struggle with multitudes of disabilities due to improper in utero programming of various tissues and organ systems contributing to adult-onset diseases at a very early stage of their lives. Therefore, the persistent rates of low birth weight (birth weight < 2,500 grams), as well as rates of neonatal and maternal morbidities and mortalities, need to be addressed. Active research throughout the years has provided us with multiple theories regarding the risk factors, initiators, biomarkers, and clinical manifestations of spontaneous preterm birth. Fetal organs, like the placenta and fetal membranes, and maternal tissues and organs, like the decidua, myometrium, and cervix, have all been shown to uniquely respond to specific exogenous or endogenous risk factors. These uniquely contribute to dynamic changes at the molecular and cellular levels to effect preterm labor pathways leading to delivery. Multiple intervention targets in these different tissues and organs have been successfully tested in preclinical trials to reduce the individual impacts on promoting preterm birth. However, these preclinical trial data have not been effectively translated into developing biomarkers of high-risk individuals for an early diagnosis of the disease. This becomes more evident when examining the current global rate of preterm birth, which remains staggeringly high despite years of research. We postulate that studying each tissue and organ in silos, as how the majority of research has been conducted in the past years, is unlikely to address the network interaction between various systems leading to a synchronized activity during either term or preterm labor and delivery. To address current limitations, this review proposes an integrated approach to studying various tissues and organs involved in the maintenance of normal pregnancy, promotion of normal parturition, and more importantly, contributions towards preterm birth. We also stress the need for biological models that allows for concomitant observation and analysis of interactions, rather than focusing on these tissues and organ in silos.
Collapse
Affiliation(s)
- Manuel S. Vidal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ryan C. V. Lintao
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Mary Elise L. Severino
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ourlad Alzeus G. Tantengco
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
48
|
Bakalar D, O’Reilly JJ, Lacaille H, Salzbank J, Ellegood J, Lerch JP, Sasaki T, Imamura Y, Hashimoto-Torii K, Vacher CM, Penn AA. Lack of placental neurosteroid alters cortical development and female somatosensory function. Front Endocrinol (Lausanne) 2022; 13:972033. [PMID: 36313771 PMCID: PMC9606442 DOI: 10.3389/fendo.2022.972033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/28/2022] [Indexed: 01/24/2023] Open
Abstract
Placental endocrine function is essential to fetal brain development. Placental hormones include neurosteroids such as allopregnanolone (ALLO), a regulator of neurodevelopmental processes via positive allosteric modulation of the GABAA receptor (GABAA-R). Using a mouse model (plKO) in which the gene encoding the ALLO synthesis enzyme is specifically deleted in trophoblasts, we previously showed that placental ALLO insufficiency alters cerebellar white matter development and leads to male-specific autistic-like behavior. We now demonstrate that the lack of placental ALLO causes female-predominant alterations of cortical development and function. Placental ALLO insufficiency disrupts cell proliferation in the primary somatosensory cortex (S1) in a sex-linked manner. Early changes are seen in plKO embryos of both sexes, but persist primarily in female offspring after birth. Adolescent plKO females show significant reduction in pyramidal neuron density, as well as somatosensory behavioral deficits as compared with plKO males and control littermates. Assessment of layer-specific markers in human postmortem cortices suggests that preterm infants may also have female-biased abnormalities in cortical layer specification as compared with term infants. This study establishes a novel and fundamental link between placental function and sex-linked long-term neurological outcomes, emphasizing the importance of the growing field of neuroplacentology.
Collapse
Affiliation(s)
- Dana Bakalar
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Health System, Washington, DC, United States
| | - Jiaqi J. O’Reilly
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Helene Lacaille
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Jacquelyn Salzbank
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, ON, Canada
| | - Jason P. Lerch
- Wellcome Centre for Integrative Neuroimaging (WIN), Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Toru Sasaki
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Health System, Washington, DC, United States
| | - Yuka Imamura
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Health System, Washington, DC, United States
| | - Claire-Marie Vacher
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Anna A. Penn
- Division of Neonatology, Department of Pediatrics, NewYork-Presbyterian Morgan Stanley Children’s Hospital, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
49
|
Choi J, Kim YS, Kim MH, Kim HJ, Yoon BE. Maternal lead exposure induces sex-dependent cerebellar glial alterations and repetitive behaviors. Front Cell Neurosci 2022; 16:954807. [PMID: 36072563 PMCID: PMC9442054 DOI: 10.3389/fncel.2022.954807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Lead (Pb) is one of the most prevalent heavy metals we encounter daily. Although there are many reports regarding their toxic effects on humans, the effects of exposure to low lead concentrations throughout the pregnancy period on the offspring are not fully elucidated yet. This study aimed to investigate the cellular mechanisms that occur in response to lead exposure. To this end, we administered lead-containing water to pregnant mice from the day of conception till delivery or till day 28 postnatally. Furthermore, we performed neurodevelopmental disorder-related behavior tests and RNA-sequencing analysis. We used both genders for all experiments because neurodevelopmental disorders usually show several sex-dependent differences. The results revealed increased levels of gliosis in the cerebella of lead-exposed pups compared to those in littermates belonging to the control group. Additionally, we observed altered behaviors of male mice in the autism spectrum disorder-related tests. RNA-sequencing results revealed changes in gamma-aminobutyric acid (GABA) signaling in the lead-exposed mouse model. Specifically, the lead-exposed male mice showed decreased monoamine oxidase B and increased levels of diamine oxidase enzyme, which is related to the synthesis of GABA in astrocytes. These findings demonstrate sex-dependent basal developmental changes in glial cells and an increased prevalence of autistic-like behaviors in the young pups of mothers exposed to lead during pregnancy.
Collapse
Affiliation(s)
- Juwon Choi
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
| | - Yoo Sung Kim
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
| | - Mi-Hye Kim
- Department of Physiology, College of Medicine, Dankook University, Cheonan, South Korea
- Department of Medical Laser, Graduate School, Dankook University, Cheonan, South Korea
| | - Hee Jung Kim
- Department of Physiology, College of Medicine, Dankook University, Cheonan, South Korea
| | - Bo-Eun Yoon
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
- *Correspondence: Bo-Eun Yoon,
| |
Collapse
|
50
|
Sze Y, Brunton PJ. Neurosteroids and early-life programming: An updated perspective. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 25:100367. [PMID: 36561280 PMCID: PMC7613978 DOI: 10.1016/j.coemr.2022.100367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Early-life stress can lead to detrimental offspring outcomes, including an increased risk for mood disorders and hypothalamic-pituitary-adrenal axis dysregulation. Neurosteroids bind to ligand-gated neurotransmitter receptors, rapidly modulating neuronal excitability and promoting termination of stress responses. Reduced neurosteroidogenesis underlies some of the aberrant neuroendocrine and behavioural phenotypes observed in adult prenatally stressed rodents. During development, disruptions in neurosteroid generation and action also lead to long-term programming effects on the off-spring's brain and behaviour. Here, we review recent advances in the field, focusing on the interaction between neurosteroids and early-life stress outcomes in adulthood and in the perinatal period. We also discuss the direction of future research, with emphasis on quantification methods, sex differences, and neurosteroids as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, Scotland, UK
| | - Paula J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, Scotland, UK
| |
Collapse
|