1
|
Cainelli E, Vedovelli L, Bisiacchi P. The mother-child interface: A neurobiological metamorphosis. Neuroscience 2024; 561:92-106. [PMID: 39427701 DOI: 10.1016/j.neuroscience.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
From the start of pregnancy, mother and child induce reciprocal neurobiological changes in the brain that will prove critical for neurodevelopment and survival of both. Molecular communication between mother and fetus is constantly active and persists even after the fetus starts to synthesize its hormones in late gestation. Intriguingly, some mother and fetus exchange cells remain in the other's brain and body with long-lasting effects and memories that do not follow the laws of classical genetics but involve complex epigenetic mechanisms. After childbirth, mother and child go through a transitional phase, a sort of limbo in which both will have a peculiar functioning profile, which is adaptive for contingencies but also renders them vulnerable. The interplay between these two "limbo" states allows for an easier transition to the subsequent phases of development. In this review, we will trace mother's and child's path from pregnancy to the months following birth and, in particular, unravel i) the key features of pregnancy and brain development and the reciprocal influences; ii) how a transitory pattern of functioning characterize mother and child, moving them toward more flexible and evolved forms; and iii) how mother and fetus act during childbirth to promote neuroprotection, pain reduction, and neurophysiological changes. Therefore, this review covers a wide range of topics, integrating neuroanatomical, neurological, biochemical, neurophysiological, and psychological studies in a meaningful way, trying to integrate them in a holistic view of the mother-child interface that is usually neglected.
Collapse
Affiliation(s)
- Elisa Cainelli
- Department of General Psychology, University of Padova, 35131 Padova, Italy.
| | - Luca Vedovelli
- Unit of Biostatistics, Epidemiology, and Public Health, Department of Cardiac, Thoracic, Vascular and Public Health Sciences, University of Padova, 35131 Padova, Italy.
| | - Patrizia Bisiacchi
- Department of General Psychology, University of Padova, 35131 Padova, Italy; Padova Neuroscience Center, PNC, 35131 Padova, Italy.
| |
Collapse
|
2
|
Sebők-Welker T, Posta E, Ágrez K, Rádosi A, Zubovics EA, Réthelyi MJ, Ulbert I, Pászthy B, Bunford N. The Association Between Prenatal Maternal Stress and Adolescent Affective Outcomes is Mediated by Childhood Maltreatment and Adolescent Behavioral Inhibition System Sensitivity. Child Psychiatry Hum Dev 2024; 55:1-21. [PMID: 36738426 PMCID: PMC11362206 DOI: 10.1007/s10578-023-01499-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 02/05/2023]
Abstract
Prenatal maternal stress is linked to offspring outcomes; however, there is little research on adolescents, behavioral, transdiagnostic outcomes, or the mechanisms through which relations operate. We examined, in N = 268 adolescents (Mage = 15.31 years; SD = 1.063; 57.8% boys) whether prenatal maternal stress is associated with adolescent affective outcomes; whether this association is mediated, serially, by childhood home atmosphere and adolescent behavioral inhibition system (BIS) sensitivity; and whether mediational effects are moderated by adolescent attention-deficit/hyperactivity disorder or maternal internalizing symptomology. Prenatal maternal daily stress and major life events were associated with adolescent outcomes through childhood negative atmosphere/neglect and BIS sensitivity, with no evidence of moderation. Results have implications regarding the effect of prenatal maternal stress on offspring outcomes and regarding corresponding sensitive periods.
Collapse
Affiliation(s)
- T Sebők-Welker
- Developmental and Translational Neuroscience Research Group Developmental and Translational Neuroscience Research Group, Research Centre for Natural Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
- Doctoral School of Mental Health Sciences, Semmelweis University, Balassa U. 6, Budapest, 1083, Hungary
| | - E Posta
- Developmental and Translational Neuroscience Research Group Developmental and Translational Neuroscience Research Group, Research Centre for Natural Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
| | - K Ágrez
- Developmental and Translational Neuroscience Research Group Developmental and Translational Neuroscience Research Group, Research Centre for Natural Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
| | - A Rádosi
- Developmental and Translational Neuroscience Research Group Developmental and Translational Neuroscience Research Group, Research Centre for Natural Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
- Doctoral School of Mental Health Sciences, Semmelweis University, Balassa U. 6, Budapest, 1083, Hungary
| | - E A Zubovics
- Developmental and Translational Neuroscience Research Group Developmental and Translational Neuroscience Research Group, Research Centre for Natural Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
| | - M J Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa U. 6, Budapest, 1083, Hungary
| | - I Ulbert
- Integrative Neuroscience Research Group, Research Centre for Natural Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter Utca 50/A, Budapest, 1083, Hungary
| | - B Pászthy
- 1st Department of Paediatrics, Semmelweis University, Bókay János U. 53-54, Budapest, 1083, Hungary
| | - N Bunford
- Developmental and Translational Neuroscience Research Group Developmental and Translational Neuroscience Research Group, Research Centre for Natural Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary.
| |
Collapse
|
3
|
Deer LK, Demers CH, Hankin BL, Doom JR, Shields GS, Hoffman MC, Davis EP. Neonatal Hair Cortisol and Birth Outcomes: An Empirical Study and Meta-Analysis. Psychosom Med 2024; 86:720-729. [PMID: 39132972 DOI: 10.1097/psy.0000000000001339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
OBJECTIVE Prenatal stress physiology is often posited as a predictor of birth outcomes, including gestational age at birth and birthweight. However, research has predominantly relied on indicators in the maternal system, with few studies examining hormones of the fetal system. The current study focuses on fetal cortisol in the third trimester, as measured in neonatal hair, as a biological factor that might be associated with birth outcomes (gestational age at birth and birthweight). We report findings from two studies: a longitudinal cohort (Study 1), and a meta-analysis of the existing literature (Study 2). METHODSSTUDY Hair was collected for cortisol analysis from 168 neonates (55.95% female) shortly after birth. Gestational age at birth and birthweight were abstracted from medical records. METHODSSTUDY An exhaustive search of four databases was conducted, yielding 155 total studies for screening. Papers reporting neonatal hair cortisol (collection <2 weeks postpartum) and birth outcomes among human neonates were retained for analysis, including Study 1 results ( k = 9). RESULTSSTUDY Higher neonatal hair cortisol was related to longer gestation ( r = 0.28, p < .001) and higher birthweight, r = 0.16, p = .040. Sex did not moderate either association. RESULTSSTUDY Across the nine studies, higher neonatal hair cortisol predicted both longer gestation ( r = 0.35, p < .001, 95% confidence interval = 0.24-0.45) and higher birthweight ( r = 0.18, p = .001, 95% confidence interval = 0.07-0.28). Neonatal sex did not moderate these associations. CONCLUSIONS Fetal cortisol exposure in the third trimester plays a role in normative maturation of the fetus, and findings reveal that higher cortisol is associated with positive birth outcomes.
Collapse
Affiliation(s)
- LillyBelle K Deer
- From the Department of Psychology (Deer, Doom, Davis), University of Denver, Denver; Department of Psychiatry (Demers, Hoffman), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Psychology (Hankin), University of Illinois Urbana-Champaign, Champaign, Illinois; Department of Psychological Science (Shields), University of Arkansas, Fayetteville, Arkansas; Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology (Hoffman), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and Department of Pediatrics (Davis), University of California, Irvine, Irvine, California
| | | | | | | | | | | | | |
Collapse
|
4
|
Choi MK, Cook A, Mungikar K, Eachus H, Tochwin A, Linke M, Gerber S, Ryu S. Exposure to elevated glucocorticoid during development primes altered transcriptional responses to acute stress in adulthood. iScience 2024; 27:110160. [PMID: 38989456 PMCID: PMC11233911 DOI: 10.1016/j.isci.2024.110160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/02/2024] [Accepted: 05/29/2024] [Indexed: 07/12/2024] Open
Abstract
Early life stress (ELS) is a major risk factor for developing psychiatric disorders, with glucocorticoids (GCs) implicated in mediating its effects in shaping adult phenotypes. In this process, exposure to high levels of developmental GC (hdGC) is thought to induce molecular changes that prime differential adult responses. However, identities of molecules targeted by hdGC exposure are not completely known. Here, we describe lifelong molecular consequences of hdGC exposure using a newly developed zebrafish double-hit stress model, which shows altered behaviors and stress hypersensitivity in adulthood. We identify a set of primed genes displaying altered expression only upon acute stress in hdGC-exposed adult fish brains. Interestingly, this gene set is enriched in risk factors for psychiatric disorders in humans. Lastly, we identify altered epigenetic regulatory elements following hdGC exposure. Thus, our study provides comprehensive datasets delineating potential molecular targets mediating the impact of hdGC exposure on adult responses.
Collapse
Affiliation(s)
- Min-Kyeung Choi
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, EX4 4QD Exeter, UK
| | - Alexander Cook
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Kanak Mungikar
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Helen Eachus
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, EX4 4QD Exeter, UK
| | - Anna Tochwin
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, EX4 4QD Exeter, UK
| | - Matthias Linke
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Soojin Ryu
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, EX4 4QD Exeter, UK
| |
Collapse
|
5
|
Zheng B, Zheng Y, Hu W, Chen Z. Dissecting the networks underlying diverse brain disorders after prenatal glucocorticoid overexposure. Arch Toxicol 2024; 98:1975-1990. [PMID: 38581585 DOI: 10.1007/s00204-024-03733-2] [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: 12/06/2023] [Accepted: 03/07/2024] [Indexed: 04/08/2024]
Abstract
New human life begins in the uterus in a period of both extreme plasticity and sensitivity to environmental disturbances. The fetal stage is also a vital period for central nervous system development, with experiences at this point profoundly and permanently shaping brain structure and function. As such, some brain disorders may originate in utero. Glucocorticoids, a class of essential stress hormones, play indispensable roles in fetal development, but overexposure may have lasting impacts on the brain. In this review, we summarize data from recent clinical and non-clinical studies regarding alterations in fetal brains due to prenatal glucocorticoid overexposure that are associated with nervous system disorders. We discuss relevant changes to brain structure and cellular functions and explore the underlying molecular mechanisms. In addition, we summarize factors that may cause differential outcomes between varying brain regions, and outline clinically feasible intervention strategies that are expected to minimize negative consequences arising from fetal glucocorticoid overexposure. Finally, we highlight the need for experimental evidence aided by new technologies to clearly determine the effects of excessive prenatal glucocorticoid exposure. This review consolidates diverse findings to help researchers better understand the relationship between the prenatal glucocorticoid overexposure and the effects it has on various fetal brain regions, promoting further development of critical intervention strategies.
Collapse
Affiliation(s)
- Baixiu Zheng
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Weiwei Hu
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
6
|
Ngema M, Xulu ND, Ngubane PS, Khathi A. Pregestational Prediabetes Induces Maternal Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysregulation and Results in Adverse Foetal Outcomes. Int J Mol Sci 2024; 25:5431. [PMID: 38791468 PMCID: PMC11122116 DOI: 10.3390/ijms25105431] [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: 03/26/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Maternal type 2 diabetes mellitus (T2DM) has been shown to result in foetal programming of the hypothalamic-pituitary-adrenal (HPA) axis, leading to adverse foetal outcomes. T2DM is preceded by prediabetes and shares similar pathophysiological complications. However, no studies have investigated the effects of maternal prediabetes on foetal HPA axis function and postnatal offspring development. Hence, this study investigated the effects of pregestational prediabetes on maternal HPA axis function and postnatal offspring development. Pre-diabetic (PD) and non-pre-diabetic (NPD) female Sprague Dawley rats were mated with non-prediabetic males. After gestation, male pups born from the PD and NPD groups were collected. Markers of HPA axis function, adrenocorticotropin hormone (ACTH) and corticosterone, were measured in all dams and pups. Glucose tolerance, insulin and gene expressions of mineralocorticoid (MR) and glucocorticoid (GR) receptors were further measured in all pups at birth and their developmental milestones. The results demonstrated increased basal concentrations of ACTH and corticosterone in the dams from the PD group by comparison to NPD. Furthermore, the results show an increase basal ACTH and corticosterone concentrations, disturbed MR and GR gene expression, glucose intolerance and insulin resistance assessed via the Homeostasis Model Assessment (HOMA) indices in the pups born from the PD group compared to NPD group at all developmental milestones. These observations reveal that pregestational prediabetes is associated with maternal dysregulation of the HPA axis, impacting offspring HPA axis development along with impaired glucose handling.
Collapse
Affiliation(s)
| | | | | | - Andile Khathi
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Westville, Private Bag X54001, Durban 4041, KwaZulu Natal, South Africa; (M.N.); (N.D.X.); (P.S.N.)
| |
Collapse
|
7
|
Fernández V, Borrell V. Keep calm and make neurons: The effects of glucocorticoids on human cortical neurogenesis. Neuron 2024; 112:1373-1375. [PMID: 38697018 DOI: 10.1016/j.neuron.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024]
Abstract
Maternal well-being is important for the development of the fetus, with a key influence on its nervous system. In this issue of Neuron, Krontira et al.1 implicate glucocorticoids, the stress hormones, in the regulation of neural stem cell identity and proliferation, with long-lasting consequences on brain architecture and educational attainment.
Collapse
Affiliation(s)
- Virginia Fernández
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain.
| |
Collapse
|
8
|
Krontira AC, Cruceanu C, Dony L, Kyrousi C, Link MH, Rek N, Pöhlchen D, Raimundo C, Penner-Goeke S, Schowe A, Czamara D, Lahti-Pulkkinen M, Sammallahti S, Wolford E, Heinonen K, Roeh S, Sportelli V, Wölfel B, Ködel M, Sauer S, Rex-Haffner M, Räikkönen K, Labeur M, Cappello S, Binder EB. Human cortical neurogenesis is altered via glucocorticoid-mediated regulation of ZBTB16 expression. Neuron 2024; 112:1426-1443.e11. [PMID: 38442714 DOI: 10.1016/j.neuron.2024.02.005] [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: 01/17/2023] [Revised: 08/15/2023] [Accepted: 02/06/2024] [Indexed: 03/07/2024]
Abstract
Glucocorticoids are important for proper organ maturation, and their levels are tightly regulated during development. Here, we use human cerebral organoids and mice to study the cell-type-specific effects of glucocorticoids on neurogenesis. We show that glucocorticoids increase a specific type of basal progenitors (co-expressing PAX6 and EOMES) that has been shown to contribute to cortical expansion in gyrified species. This effect is mediated via the transcription factor ZBTB16 and leads to increased production of neurons. A phenome-wide Mendelian randomization analysis of an enhancer variant that moderates glucocorticoid-induced ZBTB16 levels reveals causal relationships with higher educational attainment and altered brain structure. The relationship with postnatal cognition is also supported by data from a prospective pregnancy cohort study. This work provides a cellular and molecular pathway for the effects of glucocorticoids on human neurogenesis that relates to lasting postnatal phenotypes.
Collapse
Affiliation(s)
- Anthi C Krontira
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany; International Max Planck Research School for Translational Psychiatry, Munich 80804, Germany.
| | - Cristiana Cruceanu
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Leander Dony
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany; International Max Planck Research School for Translational Psychiatry, Munich 80804, Germany; Department for Computational Health, Helmholtz Munich, Neuherberg 85764, Germany; TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising 85354, Germany
| | - Christina Kyrousi
- Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich 80804, Germany; First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens 15784, Greece; University Mental Health, Neurosciences and Precision Medicine Research Institute "Costas Stefanis", Athens 15601, Greece
| | - Marie-Helen Link
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Nils Rek
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany; International Max Planck Research School for Translational Psychiatry, Munich 80804, Germany
| | - Dorothee Pöhlchen
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany; International Max Planck Research School for Translational Psychiatry, Munich 80804, Germany
| | - Catarina Raimundo
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Signe Penner-Goeke
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Alicia Schowe
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany; Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University, Munich 82152, Germany
| | - Darina Czamara
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland; Finnish Institute for Health and Welfare, Helsinki 00271, Finland; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Sara Sammallahti
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, Helsinki 00014, Finland
| | - Elina Wolford
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Kati Heinonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland; Psychology/Welfare, Faculty of Social Sciences, University of Tampere, Tampere 33014, Finland; Lawrence S. Bloomberg Faculty of Nursing, University of Toronto, Toronto, ON M5T 1P8, Canada
| | - Simone Roeh
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Vincenza Sportelli
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Barbara Wölfel
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Maik Ködel
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Susann Sauer
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Monika Rex-Haffner
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Marta Labeur
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Silvia Cappello
- Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich 80804, Germany; Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich 82152, Germany
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich 80804, Germany.
| |
Collapse
|
9
|
Zeng S, Liu H, Li B, Guo X, Chen S, Li X, Liang J, Liang H, Shen T, Long Y, Zhou H, Zhang D. Association of air temperature exposure during pregnancy with risk of preeclampsia in Guangzhou, China. ENVIRONMENT INTERNATIONAL 2024; 186:108646. [PMID: 38615543 DOI: 10.1016/j.envint.2024.108646] [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: 01/25/2024] [Revised: 03/20/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Environmental exposures during pregnancy have been associated with adverse obstetric outcomes. However, limited and inconsistent evidence exists regarding the association between air temperature exposure and the risk of preeclampsia (PE). This study aimed to evaluate the correlation between ambient temperature exposure during pregnancy and PE risk, as well as identify the specific time window of temperature exposure that increases PE risk. A population-based cohort study was conducted from January 2012 to April 2022 in Guangzhou, China. Pregnant women were recruited in early pregnancy and followed until delivery. A total of 3,314 PE patients and 114,201 normal pregnancies were included. Ambient temperature exposures at different gestational weeks were recorded for each participant. Logistic regression models were used to evaluate the correlation between ambient temperature exposure and PE risk. Stratified analyses were conducted based on maternal age and pre-pregnancy BMI. Distributed lag models were employed to identify the time window of temperature exposure related to PE. Exposure to extreme high temperature (aOR = 1.24, 95 % CI 1.12-1.38) and moderate high temperature (aOR = 1.22, 95 % CI 1.10-1.35) during early pregnancy was associated with an increased risk of PE. Furthermore, women with higher pre-pregnancy BMI had a higher risk of developing PE when exposed to high temperature during early pregnancy compared to normal-weight women. The time window of temperature exposure related to PE was identified as pregnancy weeks 1 to 8. This study provides evidence for the association of high temperature exposure during early pregnancy with the risk of PE, as well as identifies the specific time window of temperature exposure related to PE. These findings have implications for developing potential strategies to protect pregnant women, particularly those with higher pre-pregnancy BMI, from the adverse effects of extreme temperatures during early pregnancy.
Collapse
Affiliation(s)
- Shanshui Zeng
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China; Department of Laboratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, People's Republic of China
| | - Haojing Liu
- Department of Health Management, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Bingyu Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Xuanjie Guo
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Shulei Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Xuyu Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Jiarui Liang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Huaaishi Liang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Tingting Shen
- Medicine Laboratory, NanFang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yan Long
- Department of Laboratory Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, People's Republic of China.
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China.
| | - Dongxin Zhang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China.
| |
Collapse
|
10
|
Castro-Quintas Á, Eixarch E, Martin-Gonzalez NS, Daura-Corral M, Marques-Feixa L, Palma-Gudiel H, Rocavert-Barranco M, Miguel-Valero A, Monteserín-García JL, de la Fuente-Tomás L, Crispi F, Arias B, García-Portilla MP, Fañanás L. Diurnal cortisol throughout pregnancy and its association with maternal depressive symptoms and birth outcomes. Psychoneuroendocrinology 2024; 161:106930. [PMID: 38142606 DOI: 10.1016/j.psyneuen.2023.106930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 11/29/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Depression during pregnancy is a common complication that can negatively affect fetal health and birth outcomes. Cortisol is believed to be a key mediator of this association. Although pregnancy entails a natural increase in cortisol levels, preclinical depression could alter its circadian rhythm, producing excessively high overall diurnal cortisol levels that might be harmful for the fetus and future offspring development. OBJECTIVES Using a prospective longitudinal design, we aimed to study (i) trimestral cortisol circadian rhythm and its overall levels throughout pregnancy in healthy women, (ii) the extent to which maternal depressive symptoms influence both cortisol rhythmicity and overall levels, and (iii) the possible adverse consequences of elevated maternal cortisol on the offspring's weight and gestational age at birth. STUDY DESIGN 112 healthy pregnant women from the general Spanish population were recruited before their first pregnancy. To assess cortisol circadian rhythm, participants provided four saliva samples at each trimester of pregnancy (at awakening, 30 min after awakening, before lunch and before going to bed). Overall cortisol levels were calculated with AUCg approximation. Depressive symptoms were evaluated in each trimester and defined according to EPDS cut-off values (1st trimester, EPDS ≥ 11; 2nd and 3rd trimesters, EPDS ≥ 10). At birth, the risk for low weight, prematurity and weight birth percentile was retrieved for 100 infants. Mixed models and simple effects were employed to study changes of maternal cortisol circadian rhythm and overall levels throughout pregnancy and the possible influence of maternal depressive symptoms. Finally, logistic regressions were performed to assess the associations between maternal overall cortisol levels in each trimester of pregnancy and birth anthropometrics. RESULTS Although overall diurnal cortisol levels increase throughout pregnancy, cortisol circadian rhythm is preserved in all trimesters [1st (F(3110)= 92.565, p < .001), 2nd (F(3,85)= 46.828, p < .001) and 3rd (F(3,90)= 65.555, p < .001)]. However, women with depressive symptoms showed a flattened cortisol circadian pattern only during the second trimester, characterized by a blunted awakening peak and reduced evening decline (F(3,85)= 4.136, p = .009), but not during the first (F(3,11)= 1.676, p = .176) or the third (F(3,90)= 1.089, p = .358) trimesters. Additionally, they did not show a cortisol increase from second to third trimester (p = .636). Finally, higher maternal cortisol levels in second and third trimesters seemed to be associated with increased risk of prematurity (adjusted OR -0.371, 95% CI 0.490-0.972, p = .034) and low birth weight percentile (adjusted OR -0.612, 95% CI 0.348-0.846, p = .007) respectively. CONCLUSION Maternal cortisol levels increased throughout pregnancy, although cortisol circadian rhythm was preserved in all trimesters of pregnancy. However, prenatal depressive symptoms were associated with flattened maternal cortisol circadian rhythm in mid-pregnancy. Therefore, it seems that women with depressive symptoms tended to increase less gradually their cortisol levels from mid to late pregnancy. Finally, higher maternal cortisol levels in mid and late-pregnancy seem to be associated with poorer birth anthropometrics Early detection of depressive symptoms in general population could help to prevent putative obstetrical and birth adverse outcomes.
Collapse
Affiliation(s)
- Águeda Castro-Quintas
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Elisenda Eixarch
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; Network Centre for Biomedical Research on Rare Diseases (CIBER of Rare Diseases, CIBERER), Institute of Health Carlos III, Madrid, Spain
| | - Nerea San Martin-Gonzalez
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Maria Daura-Corral
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Laia Marques-Feixa
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Helena Palma-Gudiel
- Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain; College of Public Health and Health Professions, Department of Epidemiology, University of Florida, United States
| | | | - Alba Miguel-Valero
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jose Luis Monteserín-García
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Lorena de la Fuente-Tomás
- Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain; Department of Psychiatry, University of Oviedo, Asturias, Spain
| | - Fátima Crispi
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; Network Centre for Biomedical Research on Rare Diseases (CIBER of Rare Diseases, CIBERER), Institute of Health Carlos III, Madrid, Spain
| | - Barbara Arias
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - María Paz García-Portilla
- Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain; Department of Psychiatry, University of Oviedo, Asturias, Spain; Servicio de Salud del Principado de Asturias (SESPA), Oviedo, Spain
| | - Lourdes Fañanás
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
11
|
Gao P, Chang C, Liang J, Du F, Zhang R. Embryonic Amoxicillin Exposure Has Limited Impact on Liver Development but Increases Susceptibility to NAFLD in Zebrafish Larvae. Int J Mol Sci 2024; 25:2744. [PMID: 38473993 DOI: 10.3390/ijms25052744] [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: 01/23/2024] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Amoxicillin is commonly used in clinical settings to target bacterial infection and is frequently prescribed during pregnancy. Investigations into its developmental toxicity and effects on disease susceptibility are not comprehensive. Our present study examined the effects of embryonic amoxicillin exposure on liver development and function, especially the effects on susceptibility to non-alcoholic fatty liver disease (NAFLD) using zebrafish as an animal model. We discovered that embryonic amoxicillin exposure did not compromise liver development, nor did it induce liver toxicity. However, co-treatment of amoxicillin and clavulanic acid diminished BESP expression, caused bile stasis and induced liver toxicity. Embryonic amoxicillin exposure resulted in elevated expression of lipid synthesis genes and exacerbated hepatic steatosis in a fructose-induced NAFLD model, indicating embryonic amoxicillin exposure increased susceptibility to NAFLD in zebrafish larvae. In summary, this research broadens our understanding of the risks of amoxicillin usage during pregnancy and provides evidence for the impact of embryonic amoxicillin exposure on disease susceptibility in offspring.
Collapse
Affiliation(s)
- Peng Gao
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Cheng Chang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Jieling Liang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Fen Du
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Ruilin Zhang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| |
Collapse
|
12
|
Abramova O, Zorkina Y, Pavlov K, Ushakova V, Morozova A, Zubkov E, Pavlova O, Storozheva Z, Gurina O, Chekhonin V. Chronic Ultrasound Prenatal Stress Altered the Brain's Neurochemical Systems in Newborn Rats. Neural Plast 2024; 2024:3829941. [PMID: 39290524 PMCID: PMC11407898 DOI: 10.1155/2024/3829941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/25/2023] [Accepted: 01/27/2024] [Indexed: 09/19/2024] Open
Abstract
Prenatal stress (PS) affects the development and functioning of the central nervous system, but the exact mechanisms underpinning this effect have not been pinpointed yet. A promising model of PS is one based on chronic exposure of pregnant rodents to variable-frequency ultrasound (US PS), as it mimics the PS with a psychic nature that most adequately captures the human stressors in modern society. The aim of this study was to investigate the effects of US PS on the brain neurotransmitter, neuropeptide, and neurotrophic systems of newborn Wistar rats. We determined the concentration of neurotransmitters and their metabolites (serotonin, HIAA, dopamine, DOPAC, and norepinephrine), neuropeptides (α-MSH, β-endorphin, neurotensin, oxytocin, and substance P), and the neurotrophin brain-derived neurotrophic factor (BDNF) in rat brain tissues by HPLC-ED, ELISA, and multiplex ELISA. Correlation analysis and principal component analysis (PCA) were used to get a sense of the relationship between the biochemical parameters of the brain. The results demonstrated that US PS increases the concentration of serotonin (p=0.004) and DOPAC (p=0.04) in the hippocampus has no effect on the neurotransmitter systems of the frontal cortex, reduces the concentration of BDNF in the entirety of the brain of males (p=0.008), and increases the neuropeptides α-MSH (p=0.02), β-endorphin (p=0.01), oxytocin (p=0.008), and substance P (p < 0.001) in the entire brain. A degree of complexity in the neurotransmitter system network in the frontal cortex and network change in the hippocampus after exposure to US PS have been observed. PCA revealed a similar pattern of neurotransmitter system interactions in the frontal cortex and hippocampus in males and females after exposure to US PS. We suggest that US PS can alter neurodevelopment, which is mediated by changes in the studied neurochemical systems that thus affect the behavioral phenotype in animals.
Collapse
Affiliation(s)
- Olga Abramova
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Yana Zorkina
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Konstantin Pavlov
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Valeria Ushakova
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Anna Morozova
- Mental-Health Clinic No. 1 Named After N.A. Alekseev, Zagorodnoe Highway 2, Moscow 115191, Russia
| | - Eugene Zubkov
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Olga Pavlova
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Zinaida Storozheva
- Laboratory of Functional Neurochemistry, P. K. Anokhin Institute of Normal Physiology, Moscow, Russia
| | - Olga Gurina
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Vladimir Chekhonin
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
13
|
Arcego DM, Buschdorf JP, O'Toole N, Wang Z, Barth B, Pokhvisneva I, Rayan NA, Patel S, de Mendonça Filho EJ, Lee P, Tan J, Koh MX, Sim CM, Parent C, de Lima RMS, Clappison A, O'Donnell KJ, Dalmaz C, Arloth J, Provençal N, Binder EB, Diorio J, Silveira PP, Meaney MJ. A Glucocorticoid-Sensitive Hippocampal Gene Network Moderates the Impact of Early-Life Adversity on Mental Health Outcomes. Biol Psychiatry 2024; 95:48-61. [PMID: 37406925 DOI: 10.1016/j.biopsych.2023.06.028] [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: 11/03/2022] [Revised: 04/15/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Early stress increases the risk for psychiatric disorders. Glucocorticoids are stress mediators that regulate transcriptional activity and morphology in the hippocampus, which is implicated in the pathophysiology of multiple psychiatric conditions. We aimed to establish the relevance of hippocampal glucocorticoid-induced transcriptional activity as a mediator of the effects of early life on later psychopathology in humans. METHODS RNA sequencing was performed with anterior and posterior hippocampal dentate gyrus from adult female macaques (n = 12/group) that were chronically treated with betamethasone (glucocorticoid receptor agonist) or vehicle. Coexpression network analysis identified a preserved gene network in the posterior hippocampal dentate gyrus that was strongly associated with glucocorticoid exposure. The single nucleotide polymorphisms in the genes in this network were used to create an expression-based polygenic score in humans. RESULTS The expression-based polygenic score significantly moderated the association between early adversity and psychotic disorders in adulthood (UK Biobank, women, n = 44,519) and on child peer relations (ALSPAC [Avon Longitudinal Study of Parents and Children], girls, n = 1666 for 9-year-olds and n = 1594 for 11-year-olds), an endophenotype for later psychosis. Analyses revealed that this network was enriched for glucocorticoid-induced epigenetic remodeling in human hippocampal cells. We also found a significant association between single nucleotide polymorphisms from the expression-based polygenic score and adult brain gray matter density. CONCLUSIONS We provide an approach for the use of transcriptomic data from animal models together with human data to study the impact of environmental influences on mental health. The results are consistent with the hypothesis that hippocampal glucocorticoid-related transcriptional activity mediates the effects of early adversity on neural mechanisms implicated in psychiatric disorders.
Collapse
Affiliation(s)
- Danusa Mar Arcego
- Douglas Research Centre, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada.
| | - Jan-Paul Buschdorf
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Nicholas O'Toole
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Zihan Wang
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Barbara Barth
- Douglas Research Centre, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Irina Pokhvisneva
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | | | - Sachin Patel
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | | | - Patrick Lee
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Jennifer Tan
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Ming Xuan Koh
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Chu Ming Sim
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Carine Parent
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | | | - Andrew Clappison
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Kieran J O'Donnell
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada; Yale Child Study Center, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Carla Dalmaz
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Janine Arloth
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Nadine Provençal
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Josie Diorio
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Patrícia Pelufo Silveira
- Douglas Research Centre, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
| | - Michael J Meaney
- Douglas Research Centre, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore; Brain Body Initiative, Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
| |
Collapse
|
14
|
Chen Z, Xia LP, Shen L, Xu D, Guo Y, Wang H. Glucocorticoids and intrauterine programming of nonalcoholic fatty liver disease. Metabolism 2024; 150:155713. [PMID: 37914025 DOI: 10.1016/j.metabol.2023.155713] [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: 07/09/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
Accumulating epidemiological and experimental evidence indicates that nonalcoholic fatty liver disease (NAFLD) has an intrauterine origin. Fetuses exposed to adverse prenatal environments (e.g., maternal malnutrition and xenobiotic exposure) are more susceptible to developing NAFLD after birth. Glucocorticoids are crucial triggers of the developmental programming of fetal-origin diseases. Adverse intrauterine environments often lead to fetal overexposure to maternally derived glucocorticoids, which can program fetal hepatic lipid metabolism through epigenetic modifications. Adverse intrauterine environments program the offspring's glucocorticoid-insulin-like growth factor 1 (GC-IGF1) axis, which contributes to postnatal catch-up growth and disturbs glucose and lipid metabolism. These glucocorticoid-driven programming alterations increase susceptibility to NAFLD in the offspring. Notably, after delivery, offspring often face an environment distinct from their in utero life. The mismatch between the intrauterine and postnatal environments can serve as a postnatal hit that further disturbs the programmed endocrine axes, accelerating the onset of NAFLD. In this review, we summarize the current epidemiological and experimental evidence demonstrating that NAFLD has an intrauterine origin and discuss the underlying intrauterine programming mechanisms, focusing on the role of overexposure to maternally derived glucocorticoids. We also briefly discuss potential early life interventions that may be beneficial against fetal-originated NAFLD.
Collapse
Affiliation(s)
- Ze Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China
| | - Li-Ping Xia
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Lang Shen
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Dan Xu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
15
|
Ronai C, Katlaps I, Kim A, Valent AM, Thornburg KL, Madriago E. Perinatal Stressors and Consequences for Neonates with Critical Congenital Heart Disease. J Cardiovasc Dev Dis 2023; 10:497. [PMID: 38132664 PMCID: PMC10744155 DOI: 10.3390/jcdd10120497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
INTRODUCTION The prenatal diagnosis of congenital heart disease (CHD) is a traumatic event that can cause expectant parents to experience anxiety, depression, and toxic stress. Prenatal exposure to stress may impact neonatal postoperative outcomes. In addition, expectant parents may have other psychosocial stressors that may compound maternal stress. We investigated the relationship between stress in pregnancies complicated by prenatally diagnosed CHD and their neonatal outcomes. METHODS A pilot retrospective cohort study of pregnancies with prenatally diagnosed critical CHD (2019-2021) was performed. The collected data included pregnancy characteristics and neonatal and postoperative outcomes (including the need for exogenous corticosteroid treatment (ECT)). In order to quantify prenatal stressors, a composite prenatal stress score (PSS) was established and utilized. RESULTS In total, 41 maternal-fetal dyads were evaluated. Thirteen (32%) neonates had single-ventricle anatomy. The need for ECT after CHD surgery was associated with higher pregnant patient PSS (p = 0.01). PSS did not correlate with birthweight, infection, or hypoglycemia in the neonatal period. CONCLUSIONS Prenatal stress is multifactorial; higher PSS is correlates with post-bypass ECT, suggesting that a stressful intrauterine environment may be associated with worse neonatal postoperative outcomes.
Collapse
Affiliation(s)
- Christina Ronai
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Isabel Katlaps
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Amanda Kim
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Amy M. Valent
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kent L. Thornburg
- Center for Developmental Health, Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Erin Madriago
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
16
|
Santos AVS, Cardoso DS, Takada SH, Echeverry MB. Prenatal exposition to haloperidol: A preclinical narrative review. Neurosci Biobehav Rev 2023; 155:105470. [PMID: 37984569 DOI: 10.1016/j.neubiorev.2023.105470] [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: 09/04/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Pre-existing maternal mental disorders may affect the early interactions between mother and baby, impacting the child's psychoemotional development. The typical antipsychotic haloperidol can be used during pregnancy, even with some restrictions. Its prescription is not limited to psychotic disorders, but also to other psychiatric conditions of high incidence and prevalence in the woman's fertile period. The present review focused on the preclinical available data regarding the biological and behavioral implications of embryonic exposure to haloperidol. The understanding of the effects of psychotropic drugs during neurodevelopment is important for its clinical aspect since there is limited evidence regarding the risks of antipsychotic drug treatment in pregnant women and their children. Moreover, a better comprehension of the mechanistic events that can be affected by antipsychotic treatment during the critical period of neurodevelopment may offer insights into the pathophysiology of neurodevelopmental disorders. The findings presented in this review converge to the existence of several risks associated with prenatal exposure to such medication and emphasize the need for further studies regarding its dimensions.
Collapse
Affiliation(s)
- Aline Valéria Sousa Santos
- Laboratory of Neuropharmacology and Motor Behavior, Center for Mathematics, Computation, and Cognition, Federal University of ABC, São Bernardo do Campo, SP, Brazil
| | - Débora Sterzeck Cardoso
- Neurogenetics Laboratory, Center for Mathematics, Computation, and Cognition, Federal University of ABC, São Bernardo do Campo, SP, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Center for Mathematics, Computation, and Cognition, Federal University of ABC, São Bernardo do Campo, SP, Brazil
| | - Marcela Bermúdez Echeverry
- Laboratory of Neuropharmacology and Motor Behavior, Center for Mathematics, Computation, and Cognition, Federal University of ABC, São Bernardo do Campo, SP, Brazil; Neuroscience Laboratory, School of Medicine, Universidad de Santander (UDES), Bucaramanga, Santander, Colombia.
| |
Collapse
|
17
|
Wei M, Gao Q, Liu J, Yang Y, Yang J, Fan J, Lv S, Yang S. Development programming: Stress during gestation alters offspring development in sheep. Reprod Domest Anim 2023; 58:1497-1511. [PMID: 37697713 DOI: 10.1111/rda.14465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/05/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023]
Abstract
Inappropriate management practices of domestic animals during pregnancy can be potential stressors, resulting in complex behavioural, physiological and neurological consequences in the developing offspring. Some of these consequences can last into adulthood or propagate to subsequent generations. We systematically summarized the results of different experimental patterns using artificially increased maternal glucocorticoid levels or prenatal maternal physiological stress paradigms, mediators between prenatal maternal stress (PMS) and programming effects in the offspring and the effects of PMS on offspring phenotypes in sheep. PMS can impair birthweight, regulate the development of the hypothalamic-pituitary-adrenal axis, modify behavioural patterns and cognitive abilities and alter gene expression and brain morphology in offspring. Further research should focus on the effects of programming on gene expression, immune function, gut microbiome, sex-specific effects and maternal behaviour of offspring, especially comparative studies of gestational periods when PMS is applied, continual studies of programming effects on offspring and treatment strategies that effectively reverse the detrimental programming effects of prenatal stress.
Collapse
Affiliation(s)
- Mingji Wei
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Qian Gao
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Junjun Liu
- Hebei Agriculture University, Baoding, China
| | - Yan Yang
- Linyi Academy of Agricultural Sciences, Linyi, China
| | - Jinyan Yang
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Jingchang Fan
- Jiaxiang County Sheep Breeding Farm, Jiaxiang, China
| | - Shenjin Lv
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Shengmei Yang
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
| |
Collapse
|
18
|
Xie H, Shih CH, Aldoohan SD, Wall JT, Wang X. Hypothalamus volume mediates the association between adverse childhood experience and PTSD development after adulthood trauma. Transl Psychiatry 2023; 13:274. [PMID: 37542036 PMCID: PMC10403516 DOI: 10.1038/s41398-023-02576-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/06/2023] Open
Abstract
The hypothalamus is critical for regulation of the hypothalamic-pituitary-adrenal (HPA) axis and response to stress. Adverse childhood experience (ACE) can affect brain structure, which may contribute to development of posttraumatic stress disorder (PTSD) after subsequent adult trauma. It is unclear, however, if ACE history is particularly associated with aspects of hypothalamic structure which contribute to development of PTSD. To address this issue, the present study longitudinally assessed hypothalamic volumes and their associations with ACE and early post-trauma stress symptoms in subjects who did or did not develop PTSD during 12 months after adult trauma. 109 subjects (18-60 years, F/M = 75/34) completed the PTSD Checklist (PCL) questionnaire for post-trauma stress symptoms, the Childhood Trauma Questionnaire (CTQ) for ACE assessment, and an initial MRI brain scan for hypothalamic volume measurement, within 2 weeks after adult trauma. At post-trauma 12 months, subjects underwent a subsequent PTSD diagnosis interview using the Clinician-Administered PTSD Scale (CAPS), and a follow-up MRI scan. Left and right hypothalamus volumes at 2 weeks after adult trauma negatively correlated with CTQ scores. Right hypothalamus volume at this early time mediated an association between ACE and PTSD symptoms 12 months later. Right hypothalamus volumes also remained persistently smaller from 2 weeks to 12 months after trauma in survivors who developed PTSD. These results suggest that smaller right hypothalamus volume may be related to ACE history in ways that contribute to PTSD development after trauma in adulthood.
Collapse
Affiliation(s)
- Hong Xie
- Department of Neurosciences, University of Toledo, Toledo, OH, USA.
| | - Chia-Hao Shih
- Department of Emergency Medicine, University of Toledo, Toledo, OH, USA
| | | | - John T Wall
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Xin Wang
- Department of Psychiatry, University of Toledo, Toledo, OH, USA
| |
Collapse
|
19
|
Zhang F, Liu L, Hu J, Fu H, Li H, Chen J, Yang C, Guo Q, Liang X, Wang L, Guo Y, Dai J, Sheng N, Wang J. Accumulation and glucocorticoid signaling suppression by four emerging perfluoroethercarboxylic acids based on animal exposure and cell testing. ENVIRONMENT INTERNATIONAL 2023; 178:108092. [PMID: 37463541 DOI: 10.1016/j.envint.2023.108092] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023]
Abstract
Various perfluoroethercarboxylic acids (PFECA) have emerged as next-generation replacements of legacy per- and polyfluoroalkyl substances (PFAS). However, there is a paucity of information regarding their bioaccumulation ability and hazard characterization. Here, we explored the accumulation and hepatotoxicity of four PFECA compounds (HFPO-DA, HFPO-TA, PFO4DA, and PFO5DoDA) in comparison to perfluorooctanoic acid (PFOA) after chronic low-dose exposure in mice. Except for HFPO-DA, the levels of all tested PFAS in the liver exceeded that in serum. High molecular weight PFECA compounds (PFO5DoDA and HFPO-TA) showed stronger accumulation capacity and longer half-lives (t1/2) than low molecular weight PFECA compounds (HFPO-DA and PFO4DA) and even legacy PFOA. Although hepatomegaly is a common apical end point of PFAS exposure, the differentially expressed gene (DEG) profiles in the liver suggested significant differences between PFOA and the four PFECA compounds. Gene enrichment analysis supported a considerable inhibitory effect of PFECA, but not PFOA, on the glucocorticoid receptor (GR) signaling pathway. Both HFPO-TA and PFO5DoDA demonstrated a more pronounced ability to perturb RNA expression profiles in vivo and to suppress GR signaling in vitro compared to HFPO-DA and PFO4DA. Calculated reference doses (RfDs) emphasized the potential hazard of PFECA to human health. Overall, our findings indicate that PFECA alternatives do not ease the concerns raised from legacy PFAS pollution.
Collapse
Affiliation(s)
- Fenghong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Lei Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jianglin Hu
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huayu Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Hongyuan Li
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiamiao Chen
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chunyu Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Qingrong Guo
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Xiaotian Liang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Lin Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yong Guo
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Nan Sheng
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jianshe Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| |
Collapse
|
20
|
Fox MM, Knorr DA, Kwon D, Wiley KS, Parrish MH. How prenatal cortisol levels relate to grandmother-mother relationships among a cohort of Latina women. Am J Hum Biol 2023; 35:e23883. [PMID: 36862026 PMCID: PMC10474942 DOI: 10.1002/ajhb.23883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 03/03/2023] Open
Abstract
INTRODUCTION As part of the human reproductive strategy, mothers receive childcare assistance from others. For kin, allomothers are adaptively incentivized to provide assistance due to inclusive fitness benefits. Previous studies across a broad range of populations identify grandmothers as particularly consistent allomothers. Minimal attention has been paid to the possibility that allomothers may begin investing in offspring quality during the prenatal stage of life. Here, we innovate within the area of grandmother allocare research by examining the prenatal stage of life and biopsychosocial mechanisms by which prenatal grandmother effects may be enacted. METHODS Data derive from the Mothers' Cultural Experiences study, a cohort of 107 pregnant Latina women in Southern California. At <16 weeks' gestation, we administered questionnaires, collected morning urine samples, and measured cortisol by enzyme-linked immunosorbent assay, correcting for specific gravity. We measured the soon-to-be maternal and paternal grandmothers' relationship quality, social support, frequency of seeing each other, communicating, and geographic proximity to pregnant mothers, that is, their daughters and daughters-in-law. These measures were self-reported by the pregnant mothers. We assessed how grandmother constructs related to the pregnant women's depression, stress, anxiety, and cortisol levels. RESULTS We observed benefits conferred by maternal grandmothers for mothers' prenatal mental health and lower cortisol levels. Paternal grandmothers also conferred mental health benefits to pregnant daughters-in-law, but higher cortisol levels. CONCLUSION Our results suggest that grandmothers, especially maternal grandmothers, are able to improve their inclusive fitness by caring for pregnant daughters, and allomother support may positively impact prenatal health. This work extends the traditional cooperative breeding model by identifying a prenatal grandmother effect, and, by examining a maternal biomarker.
Collapse
Affiliation(s)
- Molly M. Fox
- Department of Anthropology, University of California, Los Angeles, Los Angeles, CA, 90095 USA
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, 90095 USA
| | - Delaney A. Knorr
- Department of Anthropology, University of California, Los Angeles, Los Angeles, CA, 90095 USA
| | - Dayoon Kwon
- Department of Epidemiology, UCLA Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, 90095 USA
| | - Kyle S. Wiley
- Department of Anthropology, University of California, Los Angeles, Los Angeles, CA, 90095 USA
| | - Michael H. Parrish
- Department of Psychology, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
21
|
Heynen JP, McHugh RR, Boora NS, Simcock G, Kildea S, Austin MP, Laplante DP, King S, Montina T, Metz GAS. Urinary 1H NMR Metabolomic Analysis of Prenatal Maternal Stress Due to a Natural Disaster Reveals Metabolic Risk Factors for Non-Communicable Diseases: The QF2011 Queensland Flood Study. Metabolites 2023; 13:metabo13040579. [PMID: 37110237 PMCID: PMC10145263 DOI: 10.3390/metabo13040579] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Prenatal stress alters fetal programming, potentially predisposing the ensuing offspring to long-term adverse health outcomes. To gain insight into environmental influences on fetal development, this QF2011 study evaluated the urinary metabolomes of 4-year-old children (n = 89) who were exposed to the 2011 Queensland flood in utero. Proton nuclear magnetic resonance spectroscopy was used to analyze urinary metabolic fingerprints based on maternal levels of objective hardship and subjective distress resulting from the natural disaster. In both males and females, differences were observed between high and low levels of maternal objective hardship and maternal subjective distress groups. Greater prenatal stress exposure was associated with alterations in metabolites associated with protein synthesis, energy metabolism, and carbohydrate metabolism. These alterations suggest profound changes in oxidative and antioxidative pathways that may indicate a higher risk for chronic non-communicable diseases such obesity, insulin resistance, and diabetes, as well as mental illnesses, including depression and schizophrenia. Thus, prenatal stress-associated metabolic biomarkers may provide early predictors of lifetime health trajectories, and potentially serve as prognostic markers for therapeutic strategies in mitigating adverse health outcomes.
Collapse
Affiliation(s)
- Joshua P Heynen
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Rebecca R McHugh
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Naveenjyote S Boora
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Gabrielle Simcock
- Midwifery Research Unit, Mater Research Institute, University of Queensland, Brisbane, QLD 4072, Australia
- School of Psychology, University of Queensland, Brisbane, QLD 4072, Australia
| | - Sue Kildea
- Midwifery Research Unit, Mater Research Institute, University of Queensland, Brisbane, QLD 4072, Australia
- Molly Wardaguga Research Centre, Faculty of Health, Charles Darwin University, Alice Springs, NT 0870, Australia
| | - Marie-Paule Austin
- Perinatal and Woman's Health Unit, University of New South Wales, Sydney, NSW 2052, Australia
| | - David P Laplante
- Centre for Child Development and Mental Health, Lady Davis Institute for Medical Research, Jewish General Hospital, 4335 Chemin de la Côte-Sainte-Catherine, Montreal, QC H3T 1E4, Canada
| | - Suzanne King
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 LaSalle Boulevard, Montreal, QC H4H 1R3, Canada
| | - Tony Montina
- Southern Alberta Genome Sciences Centre, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
22
|
Martín-González NS, Castro-Quintas Á, Marques-Feixa L, Ayesa-Arriola R, López M, Fañanás L. Maternal respiratory viral infections during pregnancy and offspring's neurodevelopmental outcomes: a systematic review. Neurosci Biobehav Rev 2023; 149:105178. [PMID: 37059407 DOI: 10.1016/j.neubiorev.2023.105178] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/18/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
Maternal infections during pregnancy, as cytomegalovirus and zika, have been consistently associated with severe newborn neurodevelopmental conditions, mainly related to vertical transmission and congenital infection. However, little is known about the neurodevelopmental consequences of maternal respiratory viral infections, which are the most prevalent infections during pregnancy. The recent COVID-19 pandemic has increased the interest in understanding the consequences of infections in offspring's development. This systematic review explores whether maternal gestational viral respiratory infections are associated with neurodevelopmental deviations in children below 10 years-old. The search was conducted in Pubmed, PsychInfo and Web of Science databases. 12 articles were revised, including information about maternal infection (Influenza, SARS-CoV-2 and unspecified respiratory infections) and offspring's neurodevelopment (global development, specific functions, temperament and behavioral/emotional aspects). Controversial results were reported regarding maternal respiratory infections during pregnancy and infants' neurodevelopment. Maternal infections seem to be associated with subtle alterations in some offspring's developmental subdomains, as early motor development, and attentional, behavioral/emotional minor problems. Further studies are needed to determine the impact of other psychosocial confounding factors.
Collapse
Affiliation(s)
- Nerea San Martín-González
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBER-SAM), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.
| | - Águeda Castro-Quintas
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBER-SAM), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.
| | - Laia Marques-Feixa
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBER-SAM), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.
| | - Rosa Ayesa-Arriola
- Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBER-SAM), Madrid, Spain; Department of Psychiatry, School of Medicine, University of Cantabria, University Hospital Marqués de Valdecilla, Santander, Spain; IDIVAL, Valdecilla Biomedical Research Institute, Santander, Spain.
| | - Marta López
- Fetal Medicine Research Center, Maternal fetal medicine department, BCNatal-Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; Centre for Biomedical Research on Rare Diseases (CIBER of Rare Diseases, CIBER-ER), Madrid, Spain.
| | - Lourdes Fañanás
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBER-SAM), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
23
|
Kim SA, Jang EH, Lee J, Cho SH. Neonatal Exposure to Valproate Induces Long-Term Alterations in Steroid Hormone Levels in the Brain Cortex of Prepubertal Rats. Int J Mol Sci 2023; 24:ijms24076681. [PMID: 37047656 PMCID: PMC10094755 DOI: 10.3390/ijms24076681] [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: 01/27/2023] [Revised: 03/23/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023] Open
Abstract
Valproic acid (VPA) is a known drug for treating epilepsy and mood disorders; however, it is not recommended for pregnant women because of its possible teratogenicity. VPA affects neurotransmission and gene expression through epigenetic mechanisms by acting as a histone deacetylase inhibitor and has been used to establish animal models of autism spectrum disorder (ASD). However, studies on the long-term effects of early exposure to VPA on glucocorticoid and neurosteroid synthesis in the brain are lacking. Therefore, this study aimed to investigate the long-term changes in metabolic alterations and gene expression regulation according to sex, using metabolic steroid profiling data from cerebral cortex samples of rats four weeks after VPA exposure (400 mg/kg). In neonatal VPA-exposed models, estradiol levels decreased, and cytochrome P450 19A1 gene (Cyp19a1) expression was reduced in the prepubertal male cortex. Progesterone and allopregnanolone levels decreased, and 3β-hydroxysteroid dehydrogenase 1 gene (Hsd3b1) expression was also downregulated in the prepubertal female cortex. Furthermore, cortisol levels increased, and mRNA expression of the nuclear receptor subfamily 3 group C member 1 gene (Nr3c1) was downregulated in the cortices of both sexes. Unlike the neonatal VPA-exposed models, although a decrease in progestin and estradiol levels was observed in females and males, respectively, no differences were observed in cortisol levels in the cortex tissues of 8-week-old adult rats administered VPA for four weeks. These results indicate that early environmental chemical exposure induces long-term neurosteroid metabolic effects in the brain, with differences according to sex.
Collapse
Affiliation(s)
- Soon-Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Eun-Hye Jang
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Jangjae Lee
- Chemical Analysis Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Sung-Hee Cho
- Chemical Analysis Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| |
Collapse
|
24
|
Wei J, Arber C, Wray S, Hardy J, Piers TM, Pocock JM. Human myeloid progenitor glucocorticoid receptor activation causes genomic instability, type 1 IFN- response pathway activation and senescence in differentiated microglia; an early life stress model. Glia 2023; 71:1036-1056. [PMID: 36571248 DOI: 10.1002/glia.24325] [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: 06/09/2022] [Revised: 10/26/2022] [Accepted: 12/09/2022] [Indexed: 12/27/2022]
Abstract
One form of early life stress, prenatal exposure to glucocorticoids (GCs), confers a higher risk of psychiatric and neurodevelopmental disorders in later life. Increasingly, the importance of microglia in these disorders is recognized. Studies on GCs exposure during microglial development have been limited, and there are few, if any, human studies. We established an in vitro model of ELS by continuous pre-exposure of human iPS-microglia to GCs during primitive hematopoiesis (the critical stage of iPS-microglial differentiation) and then examined how this exposure affected the microglial phenotype as they differentiated and matured to microglia, using RNA-seq analyses and functional assays. The iPS-microglia predominantly expressed glucocorticoid receptors over mineralocorticoid receptors, and in particular, the GR-α splice variant. Chronic GCs exposure during primitive hematopoiesis was able to recapitulate in vivo ELS effects. Thus, pre-exposure to prolonged GCs resulted in increased type I interferon signaling, the presence of Cyclic GMP-AMP synthase-positive (cGAS) micronuclei, cellular senescence and reduced proliferation in the matured iPS-microglia. The findings from this in vitro ELS model have ramifications for the responses of microglia in the pathogenesis of GC- mediated ELS-associated disorders such as schizophrenia, attention-deficit hyperactivity disorder and autism spectrum disorder.
Collapse
Affiliation(s)
- Jingzhang Wei
- Department of Neuroinflammation, University College London Institute of Neurology, London, UK
| | - Charles Arber
- Department of Molecular Neuroscience, University College London Institute of Neurology, London, UK
| | - Selina Wray
- Department of Molecular Neuroscience, University College London Institute of Neurology, London, UK
| | - John Hardy
- Department of Molecular Neuroscience, University College London Institute of Neurology, London, UK
| | - Thomas M Piers
- Department of Neuroinflammation, University College London Institute of Neurology, London, UK
| | - Jennifer M Pocock
- Department of Neuroinflammation, University College London Institute of Neurology, London, UK
| |
Collapse
|
25
|
Li P, Ru X, Teng Y, Han Y, Liu Z, Tao F, Huang K. Interaction between isolated maternal hypothyroxinemia and pregnancy-related anxiety on preschooler's internalizing and externalizing problems: A birth cohort study. Psychoneuroendocrinology 2023; 152:106102. [PMID: 37018881 DOI: 10.1016/j.psyneuen.2023.106102] [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: 10/29/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Isolated maternal hypothyroxinemia (IMH) and pregnancy-related anxiety may increase the risk of offspring's emotional and behavioral problems, but little is known about their potential interactive effect on preschoolers' internalizing and externalizing problems. METHODS We conducted a large prospective cohort study in Ma'anshan Maternal and Child Health Hospital between May 2013 and September 2014. There were a total of 1372 mother-child pairs from the Ma'anshan birth cohort (MABC) included in this study. IMH was defined as the thyroid-stimulating hormone (TSH) level within the normal reference range (2.5-97.5th percentile) and the free thyroxine (FT4) level below the 2.5th percentile, and negative TPOAb. The pregnancy-related anxiety questionnaire (PRAQ) was used to assess women's pregnancy-related anxiety status in the first (1-13 weeks), second (14-27 weeks) and third (after 28 weeks) trimesters of pregnancy. The Achenbach Child Behavior Checklist (CBCL/1.5-5) was used to assess preschoolers' internalizing and externalizing problems. RESULTS Preschoolers born of mothers with IMH and anxiety had an increased risk of anxious/depressed (OR = 6.40, 95% CI 1.89-21.68), somatic complaints (OR = 2.69, 95% CI 1.01-7.20), attention problems (OR = 2.95, 95% CI 1.00-8.69) and total problems (OR = 3.40, 95% CI 1.60-7.21). Particularly, mothers with IMH and anxiety was associated with an increased risk of preschool girls' anxious/depressed (OR = 8.14, 95% CI 1.74-38.08), withdrawn (OR = 7.03, 95% CI 2.25-21.92), internalizing problems (OR = 2.66, 95% CI 1.00-7.08), and total problems (OR = 5.50, 95% CI 2.00-15.10). CONCLUSIONS IMH and pregnancy-related anxiety during pregnancy may synergistically increase the risk of internalizing and externalizing problems in preschooler children. This interaction is distinct in internalizing problems of preschool girls.
Collapse
Affiliation(s)
- Peixuan Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xue Ru
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yuzhu Teng
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yan Han
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Zijian Liu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
26
|
Bassil K, Krontira AC, Leroy T, Escoto AIH, Snijders C, Pernia CD, Pasterkamp RJ, de Nijs L, van den Hove D, Kenis G, Boks MP, Vadodaria K, Daskalakis NP, Binder EB, Rutten BPF. In vitro modeling of the neurobiological effects of glucocorticoids: A review. Neurobiol Stress 2023; 23:100530. [PMID: 36891528 PMCID: PMC9986648 DOI: 10.1016/j.ynstr.2023.100530] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Hypothalamic-pituitary adrenal (HPA)axis dysregulation has long been implicated in stress-related disorders such as major depression and post-traumatic stress disorder. Glucocorticoids (GCs) are released from the adrenal glands as a result of HPA-axis activation. The release of GCs is implicated with several neurobiological changes that are associated with negative consequences of chronic stress and the onset and course of psychiatric disorders. Investigating the underlying neurobiological effects of GCs may help to better understand the pathophysiology of stress-related psychiatric disorders. GCs impact a plethora of neuronal processes at the genetic, epigenetic, cellular, and molecular levels. Given the scarcity and difficulty in accessing human brain samples, 2D and 3D in vitro neuronal cultures are becoming increasingly useful in studying GC effects. In this review, we provide an overview of in vitro studies investigating the effects of GCs on key neuronal processes such as proliferation and survival of progenitor cells, neurogenesis, synaptic plasticity, neuronal activity, inflammation, genetic vulnerability, and epigenetic alterations. Finally, we discuss the challenges in the field and offer suggestions for improving the use of in vitro models to investigate GC effects.
Collapse
Affiliation(s)
- Katherine Bassil
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Anthi C Krontira
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Thomas Leroy
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Alana I H Escoto
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Clara Snijders
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Cameron D Pernia
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center (UMC) Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Laurence de Nijs
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Daniel van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Marco P Boks
- Psychiatry, UMC Utrecht Brain Center, Utrecht, the Netherlands
| | - Krishna Vadodaria
- Salk Institute for Biological Studies, La Jolla, San Diego, United States
| | | | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
27
|
Li YS, Lee HC, Huang JP, Lin YZ, Au HK, Lo YC, Chien LC, Chao HJ, Estinfort W, Chen YH. Adverse effects of inadequate sleep duration patterns during pregnancy on toddlers suspected developmental delay: A longitudinal study. Sleep Med 2023; 105:68-77. [PMID: 36966578 DOI: 10.1016/j.sleep.2023.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/30/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
BACKGROUND Although studies have reported the effects of inadequate sleep on maternal health, few have examined the relationships of maternal sleep patterns with fetal health and early childhood development. This study investigated maternal sleep duration patterns from early pregnancy to 3-years postpartum and their effects on birth outcomes and child development. METHODS This study recruited pregnant women and their partners during prenatal visits at five selected hospitals in the Taipei area; follow-up lasted from July 2011 to April 2021. A total of 1178 parents completed self-reported assessments from early pregnancy until childbirth and 544 completed eight assessments up to 3-years postpartum. Generalized estimated equation models were used for analyses. RESULTS Group-based trajectory modeling was used to identify four trajectories of sleep duration patterns. Although maternal sleep duration was not associated with birth outcomes, maternal "short decreasing" and "stably short" sleep patterns were associated with a higher risk of suspected overall developmental delay and language developmental delay, respectively. Furthermore, an "extremely long decreasing" pattern was associated with a higher risk of suspected overall developmental delay, [adjusted odds ratio (aOR) = 2.97, 95% confidence interval (CI):1.39-6.36)], gross motor delay, (aOR = 3.14, 95% CI: 1.42-6.99) and language developmental delay (aOR = 4.59, 95% CI:1.62-13.00). The results were significant for the children of multiparous women. CONCLUSIONS We identified a U-shaped distribution of risk between offspring developmental delay and maternal prenatal sleep duration, with the highest risk levels on both ends of the maternal prenatal sleep duration pattern. Interventions for maternal sleep are relatively straightforward to implement and should thus be a key part of standard prenatal care.
Collapse
|
28
|
Petrican R, Paine AL, Escott-Price V, Shelton KH. Overlapping brain correlates of superior cognition among children at genetic risk for Alzheimer's disease and/or major depressive disorder. Sci Rep 2023; 13:984. [PMID: 36653486 PMCID: PMC9849214 DOI: 10.1038/s41598-023-28057-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Early life adversity (ELA) tends to accelerate neurobiological ageing, which, in turn, is thought to heighten vulnerability to both major depressive disorder (MDD) and Alzheimer's disease (AD). The two conditions are putatively related, with MDD representing either a risk factor or early symptom of AD. Given the substantial environmental susceptibility of both disorders, timely identification of their neurocognitive markers could facilitate interventions to prevent clinical onset. To this end, we analysed multimodal data from the Adolescent Brain and Cognitive Development study (ages 9-10 years). To disentangle genetic from correlated genetic-environmental influences, while also probing gene-adversity interactions, we compared adoptees, a group generally exposed to substantial ELA, with children raised by their biological families via genetic risk scores (GRS) from genome-wide association studies. AD and MDD GRSs predicted overlapping and widespread neurodevelopmental alterations associated with superior fluid cognition. Specifically, among adoptees only, greater AD GRS were related to accelerated structural maturation (i.e., cortical thinning) and higher MDD GRS were linked to delayed functional neurodevelopment, as reflected in compensatory brain activation on an inhibitory control task. Our study identifies compensatory mechanisms linked to MDD risk and highlights the potential cognitive benefits of accelerated maturation linked to AD vulnerability in late childhood.
Collapse
Affiliation(s)
- Raluca Petrican
- Institute of Population Health, Department of Psychology, University of Liverpool, Bedford Street South, Liverpool, L69 7ZA, UK.
| | - Amy L Paine
- School of Psychology, Cardiff University, 70 Park Place, Cardiff, CF10 3AT, UK
| | - Valentina Escott-Price
- Division of Neuroscience and Mental Health, School of Medicine, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Katherine H Shelton
- School of Psychology, Cardiff University, 70 Park Place, Cardiff, CF10 3AT, UK
| |
Collapse
|
29
|
Yao D, Lu Y, Li L, Wang S, Mu Y, Ding C, Zhao J, Liu M, Xu M, Wu H, Dou C, Zhu Z, Li H. Prolactin and glucocorticoid receptors in the prefrontal cortex are associated with anxiety-like behavior in prenatally stressed adolescent offspring rats. J Neuroendocrinol 2023; 35:e13231. [PMID: 36683309 DOI: 10.1111/jne.13231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 11/29/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022]
Abstract
Prenatal stress (PS) causes anxiety in mothers and their offspring and chewing is a commonly observed behavior during maternal stress. Prolactin (PRL) is an anti-anxiety factor that suppresses the hypothalamic-pituitary-adrenal axis. Here, we studied the roles of PRL, corticosterone (CORT), and their receptors in PS-induced anxiety-like behavior in dams and their offspring. We further investigated whether chewing during maternal stress could prevent PS-induced harmful consequences. Pregnant rats were randomly divided into PS, PS + chewing, and control groups. Anxiety-like behaviors of dams and their adolescent offspring were assessed using the open field test and elevated plus maze. Serum levels of PRL and CORT were measured by ELISA. Expression of mRNA and protein of PRLR and glucocorticoid receptor (GR) in the prefrontal cortex (PFC) were evaluated by qRT-PCR and western blotting, respectively. Compared to the control rats, dams and their female offspring, but not male offspring, in the PS group showed increased anxiety-like behaviors. The PS-affected rats had a lower serum PRL level and increased PRLR expression in the PFC. In contrast, these rats had a higher serum CORT level and decreased GR expression in the PFC. Chewing ameliorated anxiety-like behaviors and counteracted stress-induced changes in serum PRL and CORT, as well as the expression of their receptors in the PFC. Conclusion: PS-induced anxiety-like behavior is associated with changes in the serum levels of PRL and CORT and expression of their receptors in the PFC. Moreover, chewing blunts the hormonal and receptor changes and may serve as an effective stress-coping method for preventing PS-induced anxiety-like behavior.
Collapse
Affiliation(s)
- Dan Yao
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Yong Lu
- Central Laboratory, Heze Medical College, Heze, Shandong, China
| | - Li Li
- Central Laboratory, Heze Medical College, Heze, Shandong, China
| | - Shan Wang
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Yingjun Mu
- Central Laboratory, Heze Medical College, Heze, Shandong, China
| | - Chenxi Ding
- School of Public Health, Xi'an Jiaotong University Health Science Center, Shaanxi, China
| | - Jing Zhao
- Central Laboratory, Heze Medical College, Heze, Shandong, China
| | - Mingzhe Liu
- Central Laboratory, Heze Medical College, Heze, Shandong, China
| | - Meina Xu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Shaanxi, China
| | - Haoyue Wu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Shaanxi, China
| | - Chengyin Dou
- School of Public Health, Xi'an Jiaotong University Health Science Center, Shaanxi, China
| | - Zhongliang Zhu
- Maternal and Infant Health Research Institute, Northwest University, Shaanxi, China
| | - Hui Li
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
- Department of Neonatology, The Affiliated Children Hospital of Xi'an Jiaotong University, Shaanxi, China
| |
Collapse
|
30
|
Wu T, Zhou K, Hua Y, Zhang W, Li Y. The molecular mechanisms in prenatal drug exposure-induced fetal programmed adult cardiovascular disease. Front Pharmacol 2023; 14:1164487. [PMID: 37153765 PMCID: PMC10157035 DOI: 10.3389/fphar.2023.1164487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/03/2023] [Indexed: 05/10/2023] Open
Abstract
The "developmental origins of health and disease" (DOHaD) hypothesis posits that early-life environmental exposures have a lasting impact on individual's health and permanently shape growth, structure, and metabolism. This reprogramming, which results from fetal stress, is believed to contribute to the development of adulthood cardiovascular diseases such as hypertension, coronary artery disease, heart failure, and increased susceptibility to ischemic injuries. Recent studies have shown that prenatal exposure to drugs, such as glucocorticoids, antibiotics, antidepressants, antiepileptics, and other toxins, increases the risk of adult-onset cardiovascular diseases. In addition, observational and animal experimental studies have demonstrated the association between prenatal drug exposure and the programming of cardiovascular disease in the offspring. The molecular mechanisms underlying these effects are still being explored but are thought to involve metabolism dysregulation. This review summarizes the current evidence on the relationship between prenatal drug exposure and the risk of adult cardiovascular disorders. Additionally, we present the latest insights into the molecular mechanisms that lead to programmed cardiovascular phenotypes after prenatal drug exposure.
Collapse
Affiliation(s)
- Ting Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Ultrasonic Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Ultrasonic Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Ultrasonic Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wen Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Ultrasonic Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Wen Zhang, ; Yifei Li,
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Wen Zhang, ; Yifei Li,
| |
Collapse
|
31
|
Kim AW, Said Mohamed R, Norris SA, Richter LM, Kuzawa CW. Psychological legacies of intergenerational trauma under South African apartheid: Prenatal stress predicts greater vulnerability to the psychological impacts of future stress exposure during late adolescence and early adulthood in Soweto, South Africa. J Child Psychol Psychiatry 2023; 64:110-124. [PMID: 35853622 PMCID: PMC10083984 DOI: 10.1111/jcpp.13672] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND South Africa's rates of psychiatric morbidity are among the highest in sub-Saharan Africa and are foregrounded by the country's long history of political violence during apartheid. Growing evidence suggests that in utero stress exposure is a potent developmental risk factor for future mental illness risk, yet the extent to which the psychiatric effects of prenatal stress impact the next generation are unknown. We evaluate the intergenerational effects of prenatal stress experienced during apartheid on psychiatric morbidity among children at ages 17-18 and also assess the moderating effects of maternal age, social support, and past household adversity. METHODS Participants come from Birth-to-Twenty, a longitudinal birth cohort study in Soweto-Johannesburg, South Africa's largest peri-urban township which was the epicentre of violent repression and resistance during the final years of the apartheid regime. Pregnant women were prospectively enrolled in 1990 and completed questionnaires assessing social experiences, and their children's psychiatric morbidity were assessed at ages 17-18. RESULTS Full data were available from 304 mother-child pairs in 2007-8. Maternal prenatal stress in 1990 was not directly associated greater psychiatric morbidity during at ages 17-18. Maternal age and past household adversity moderated the intergenerational mental health effects of prenatal stress such that children born to younger mothers and late adolescent/young adult children experiencing greater household adversity exhibited worse psychiatric morbidity at ages 17-18. Social support did not buffer against the long-term psychiatric impacts of prenatal stress. CONCLUSIONS Greater prenatal stress from apartheid predicted adverse psychiatric outcomes among children born to younger mothers and adolescents/young adults who experienced greater concurrent stress. Our findings suggest that prenatal stress may affect adolescent mental health, have stress-sensitising effects, and represent possible intergenerational effects of trauma experienced under apartheid in this sample.
Collapse
Affiliation(s)
- Andrew Wooyoung Kim
- Department of Anthropology, University of California, Berkeley, Berkeley, CA, USA.,SAMRC/Wits Developmental Pathways for Health Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rihlat Said Mohamed
- SAMRC/Wits Developmental Pathways for Health Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shane A Norris
- SAMRC/Wits Developmental Pathways for Health Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Global Health Research Institute, School of Human Development and Health, University of Southampton, Southampton, UK.,DSI-NRF Centre of Excellence in Human Development, University of the Witwatersrand, Johannesburg, South Africa
| | - Linda M Richter
- DSI-NRF Centre of Excellence in Human Development, University of the Witwatersrand, Johannesburg, South Africa
| | - Christopher W Kuzawa
- Department of Anthropology, Northwestern University, Evanston, IL, USA.,Institute for Policy Research, Northwestern University, Evanston, IL, USA
| |
Collapse
|
32
|
Bassil K, De Nijs L, Rutten BPF, Van Den Hove DLA, Kenis G. In vitro modeling of glucocorticoid mechanisms in stress-related mental disorders: Current challenges and future perspectives. Front Cell Dev Biol 2022; 10:1046357. [DOI: 10.3389/fcell.2022.1046357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
In the last decade, in vitro models has been attracting a great deal of attention for the investigation of a number of mechanisms underlying neurological and mental disorders, including stress-related disorders, for which human brain material has rarely been available. Neuronal cultures have been extensively used to investigate the neurobiological effects of stress hormones, in particular glucocorticoids. Despite great advancements in this area, several challenges and limitations of studies attempting to model and investigate stress-related mechanisms in vitro exist. Such experiments often come along with non-standardized definitions stress paradigms in vitro, variations in cell models and cell types investigated, protocols with differing glucocorticoid concentrations and exposure times, and variability in the assessment of glucocorticoid-induced phenotypes, among others. Hence, drawing consensus conclusions from in-vitro stress studies is challenging. Addressing these limitations and aligning methodological aspects will be the first step towards an improved and standardized way of conducting in vitro studies into stress-related disorders, and is indispensable to reach the full potential of in vitro neuronal models. Here, we consider the most important challenges that need to be overcome and provide initial guidelines to achieve improved use of in vitro neuronal models for investigating mechanisms underlying the development of stress-related mental disorders.
Collapse
|
33
|
The Potential Role of PPARs in the Fetal Origins of Adult Disease. Cells 2022; 11:cells11213474. [PMID: 36359869 PMCID: PMC9653757 DOI: 10.3390/cells11213474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
The fetal origins of adult disease (FOAD) hypothesis holds that events during early development have a profound impact on one’s risk for the development of future adult disease. Studies from humans and animals have demonstrated that many diseases can begin in childhood and are caused by a variety of early life traumas, including maternal malnutrition, maternal disease conditions, lifestyle changes, exposure to toxins/chemicals, improper medication during pregnancy, and so on. Recently, the roles of Peroxisome proliferator-activated receptors (PPARs) in FOAD have been increasingly appreciated due to their wide variety of biological actions. PPARs are members of the nuclear hormone receptor subfamily, consisting of three distinct subtypes: PPARα, β/δ, and γ, highly expressed in the reproductive tissues. By controlling the maturation of the oocyte, ovulation, implantation of the embryo, development of the placenta, and male fertility, the PPARs play a crucial role in the transition from embryo to fetus in developing mammals. Exposure to adverse events in early life exerts a profound influence on the methylation pattern of PPARs in offspring organs, which can affect development and health throughout the life course, and even across generations. In this review, we summarize the latest research on PPARs in the area of FOAD, highlight the important role of PPARs in FOAD, and provide a potential strategy for early prevention of FOAD.
Collapse
|
34
|
Microglia involvement in sex-dependent behaviors and schizophrenia occurrence in offspring with maternal dexamethasone exposure. SCHIZOPHRENIA 2022; 8:71. [PMID: 36075925 PMCID: PMC9458670 DOI: 10.1038/s41537-022-00280-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/27/2022] [Indexed: 12/30/2022]
Abstract
Fetal microglia that are particularly sensitive cells to the changes in utero environment might be involved in the sex-biased onset and vulnerability to psychiatric disorders. To address this issue, we administered a 50 µg/kg dexamethasone (DEX) to dams subcutaneously from gestational days 16 to 18 and a series of behavioral assessments were performed in the offspring. Prenatal exposure to dexamethasone (PN-DEX) induced schizophrenia (SCZ)-relevant behaviors in male mice and depressive-like behavior in female mice. SCZ-relevant behavioral patterns occurred in 10-week-old (10 W) male mice but not in 4-week-old (4 W) male mice. Microglia in the medial prefrontal cortex (mPFC) and the striatum (STR) of 10 W males prenatally treated with dexamethasone (10 W PN-DEX-M) showed hyper-ramified morphology and dramatically reduced spine density in mPFC. Immunofluorescence studies indicated that microglia in the mPFC of the 10 W PN-DEX-M group interacted with pre-synaptic Bassoon and post-synaptic density 95 (PSD95) puncta. PN-DEX-M also showed significantly changed dopamine system proteins. However, a testosterone surge during adolescence was not a trigger on SCZ-relevant behavior occurrence in 10 W PN-DEX-M. Furthermore, females prenatally treated with dexamethasone (PN-DEX-F) displayed depressive-like behavior, in addition to HPA-axis activation and inflammatory microglial phenotypes in their hippocampus (HPC). We propose that altered microglial function, such as increased synaptic pruning, may be involved in the occurrence of SCZ-relevant behavior in PN-DEX-M and sex-biased abnormal behavior in the PN-DEX model.
Collapse
|
35
|
Yoshino Y, Kumon H, Shimokawa T, Yano H, Ochi S, Funahashi Y, Iga JI, Matsuda S, Tanaka J, Ueno SI. Impact of Gestational Haloperidol Exposure on miR-137-3p and Nr3c1 mRNA Expression in Hippocampus of Offspring Mice. Int J Neuropsychopharmacol 2022; 25:853-862. [PMID: 35859315 PMCID: PMC9593222 DOI: 10.1093/ijnp/pyac044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/11/2022] [Accepted: 07/19/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Schizophrenia is a mental disorder caused by both environmental and genetic factors. Prenatal exposure to antipsychotics, an environmental factor for the fetal brain, induces apoptotic neurodegeneration and cognitive impairment of offspring similar to schizophrenia. The aim was to investigate molecular biological changes in the fetal hippocampus exposed to haloperidol (HAL) by RNA expression as a model of the disorder. METHODS HAL (1 mg/kg/d) was administered to pregnant mice. Upregulated and downregulated gene expressions in the hippocampus of offspring were studied with RNA-sequencing and validated with the qPCR method, and micro-RNA (miR) regulating mRNA expressional changes was predicted by in silico analysis. An in vitro experiment was used to identify the miRNA using a dual-luciferase assay. RESULTS There were significant gene expressional changes (1370 upregulated and 1260 downregulated genes) in the HAL group compared with the control group on RNA-sequencing analysis (P < .05 and q < 0.05). Of them, the increase of Nr3c1 mRNA expression was successfully validated, and in silico analysis predicted that microRNA-137-3p (miR-137-3p) possibly regulates that gene's expression. The expression of miR-137-3p in the hippocampus of offspring was significantly decreased in the first generation, but it increased in the second generation. In vitro experiments with Neuro2a cells showed that miR-137-3p inversely regulated Nr3c1 mRNA expression, which was upregulated in the HAL group. CONCLUSIONS These findings will be key for understanding the impact of the molecular biological effects of antipsychotics on the fetal brain.
Collapse
Affiliation(s)
- Yuta Yoshino
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan
| | - Hiroshi Kumon
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan
| | - Tetsuya Shimokawa
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Shinichiro Ochi
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan
| | - Yu Funahashi
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan
| | - Jun-ichi Iga
- Correspondence: Jun-ichi Iga, MD, PhD, Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan ()
| | - Seiji Matsuda
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Shu-ichi Ueno
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan
| |
Collapse
|
36
|
Gene Dysregulation in the Adult Rat Paraventricular Nucleus and Amygdala by Prenatal Exposure to Dexamethasone. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071077. [PMID: 35888164 PMCID: PMC9316520 DOI: 10.3390/life12071077] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/24/2022] [Accepted: 07/15/2022] [Indexed: 12/02/2022]
Abstract
Fetal programming is the concept that maternal stressors during critical periods of fetal development can alter offspring phenotypes postnatally. Excess glucocorticoids can interact with the fetus to effect genetic and epigenetic changes implicated in adverse developmental outcomes. The present study investigates how chronic exposure to the synthetic glucocorticoid dexamethasone during late gestation alters the expression of genes related to behavior in brain areas relevant to the regulation and function of the hypothalamic–pituitary–adrenal axis. Pregnant Wistar Kyoto rats received subcutaneous injections of dexamethasone (100 μg/kg) daily from gestational day 15–21 or vehicle only as sham controls. The amygdala and paraventricular nucleus (PVN) were micro-punched to extract mRNA for reverse transcription and quantitative polymerase chain reaction for the analysis of the expression of specific genes. In the PVN, the expression of the glucocorticoid receptor NR3C1 was downregulated in female rats in response to programming. The expression of CACNA1C encoding the Cav1.2 pore subunit of L-type voltage-gated calcium channels was downregulated in male and female rats prenatally exposed to dexamethasone. Collectively, the results suggest that prenatal exposure to elevated levels of glucocorticoids plays a role in the dysregulation of the hypothalamic–pituitary–adrenal axis and potentially learning and memory by altering the expression of specific genes within the amygdala and PVN.
Collapse
|
37
|
Deutsch AR, Vargas MC, Lucchini M, Brink LT, Odendaal HJ, Elliott AJ. Effect of individual or comorbid antenatal depression and anxiety on birth outcomes and moderation by maternal traumatic experiences and resilience. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2022; 9:100365. [PMID: 35966253 PMCID: PMC9373828 DOI: 10.1016/j.jadr.2022.100365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background Although antenatal depression and anxiety (e.g., negative antenatal mental health; NAMH) are individually associated with preterm birth (PTB) and infant neurological impairment, few studies account for comorbidity. Understanding how NAMH impacts PTB and infant neurological functioning by either singular (depression or anxiety) or comorbid status, as well as the way in which these effects can be moderated by additional risk or protective factors (traumatic experiences and trait resiliency) can contribute further understanding of NAMH effects on birth outcomes. Methods The sample included 3042 mother-infant dyads from U.S. and South Africa cohorts of the Safe Passage Study (N = 3042). A four-category NAMH variable was created to categorize depression-only, anxiety-only, comorbid, or no NAMH statuses. Results There were no NAMH main effects on PTB, however, anxiety-only and comorbid NAMH increased odds of PTB for mothers with higher rates of traumatic life experiences. Anxiety-only and comorbid NAMH were associated with increased odds of newborn neurological impairment, and the effect of comorbid NAMH was stronger for mothers with higher rates of traumatic experiences. Resiliency decreased odds of neurological impairment for mothers who reported depression-only or anxiety-only NAMH. Limitations Limitations included potential artefacts of two cohorts that differed in rates of almost all variables, a single time point for measuring NAMH, and lack of pregnancy-specific NAMH measures. Conclusions Especially when compared to mothers with no NAMH, comorbidity or singular-condition NAMH statuses associate with negative birth outcomes in nuanced ways, especially when considering additional contexts that may foster or protect against NAMH.
Collapse
Affiliation(s)
- Arielle R. Deutsch
- Avera Research Institute
- University of South Dakota School of Medicine, Department of Pediatrics
| | | | - Maristella Lucchini
- Columbia University Irving Medical Center, Department of Psychiatry
- New York State Psychiatric Institute, Division of Developmental Neuroscience
| | - Lucy T. Brink
- Stellenbosch University, School of Medicine and Health Science, Department of Obstetrics and Gynaecology
| | - Hein J. Odendaal
- Stellenbosch University, School of Medicine and Health Science, Department of Obstetrics and Gynaecology
| | - Amy J. Elliott
- Avera Research Institute
- University of South Dakota School of Medicine, Department of Pediatrics
| |
Collapse
|
38
|
Fowden AL, Vaughan OR, Murray AJ, Forhead AJ. Metabolic Consequences of Glucocorticoid Exposure before Birth. Nutrients 2022; 14:nu14112304. [PMID: 35684104 PMCID: PMC9182938 DOI: 10.3390/nu14112304] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 02/07/2023] Open
Abstract
Glucocorticoids have an important role in development of the metabolic phenotype in utero. They act as environmental and maturational signals in adapting feto-placental metabolism to maximize the chances of survival both before and at birth. They influence placental nutrient handling and fetal metabolic processes to support fetal growth, fuel storage and energy production with respect to nutrient availability. More specifically, they regulate the transport, utilization and production of a range of nutrients by the feto-placental tissues that enables greater metabolic flexibility in utero while minimizing any further drain on maternal resources during periods of stress. Near term, the natural rise in fetal glucocorticoid concentrations also stimulates key metabolic adaptations that prepare tissues for the new energy demanding functions after birth. Glucocorticoids, therefore, have a central role in the metabolic communication between the mother, placenta and fetus that optimizes offspring metabolic phenotype for survival to reproductive age. This review discusses the effects of maternal and fetal glucocorticoids on the supply and utilization of nutrients by the feto-placental tissues with particular emphasis on studies using quantitative methods to assess metabolism in rodents and sheep in vivo during late pregnancy. It considers the routes of glucocorticoid overexposure in utero, including experimental administration of synthetic glucocorticoids, and the mechanisms by which these hormones control feto-placental metabolism at the molecular, cellular and systems levels. It also briefly examines the consequences of intrauterine glucocorticoid overexposure for postnatal metabolic health and the generational inheritance of metabolic phenotype.
Collapse
Affiliation(s)
- Abigail L. Fowden
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.J.M.); (A.J.F.)
- Correspondence:
| | - Owen R. Vaughan
- EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK;
| | - Andrew J. Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.J.M.); (A.J.F.)
| | - Alison J. Forhead
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.J.M.); (A.J.F.)
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
| |
Collapse
|
39
|
Cruceanu C, Dony L, Krontira AC, Fischer DS, Roeh S, Di Giaimo R, Kyrousi C, Kaspar L, Arloth J, Czamara D, Gerstner N, Martinelli S, Wehner S, Breen MS, Koedel M, Sauer S, Sportelli V, Rex-Haffner M, Cappello S, Theis FJ, Binder EB. Cell-Type-Specific Impact of Glucocorticoid Receptor Activation on the Developing Brain: A Cerebral Organoid Study. Am J Psychiatry 2022; 179:375-387. [PMID: 34698522 DOI: 10.1176/appi.ajp.2021.21010095] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE A fine-tuned balance of glucocorticoid receptor (GR) activation is essential for organ formation, with disturbances influencing many health outcomes. In utero, glucocorticoids have been linked to brain-related negative outcomes, with unclear underlying mechanisms, especially regarding cell-type-specific effects. An in vitro model of fetal human brain development, induced human pluripotent stem cell (hiPSC)-derived cerebral organoids, was used to test whether cerebral organoids are suitable for studying the impact of prenatal glucocorticoid exposure on the developing brain. METHODS The GR was activated with the synthetic glucocorticoid dexamethasone, and the effects were mapped using single-cell transcriptomics across development. RESULTS The GR was expressed in all cell types, with increasing expression levels through development. Not only did its activation elicit translocation to the nucleus and the expected effects on known GR-regulated pathways, but also neurons and progenitor cells showed targeted regulation of differentiation- and maturation-related transcripts. Uniquely in neurons, differentially expressed transcripts were significantly enriched for genes associated with behavior-related phenotypes and disorders. This human neuronal glucocorticoid response profile was validated across organoids from three independent hiPSC lines reprogrammed from different source tissues from both male and female donors. CONCLUSIONS These findings suggest that excessive glucocorticoid exposure could interfere with neuronal maturation in utero, leading to increased disease susceptibility through neurodevelopmental processes at the interface of genetic susceptibility and environmental exposure. Cerebral organoids are a valuable translational resource for exploring the effects of glucocorticoids on early human brain development.
Collapse
Affiliation(s)
- Cristiana Cruceanu
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Leander Dony
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Anthi C Krontira
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - David S Fischer
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Simone Roeh
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Rossella Di Giaimo
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Christina Kyrousi
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Lea Kaspar
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Janine Arloth
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Darina Czamara
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Nathalie Gerstner
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Silvia Martinelli
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Stefanie Wehner
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Michael S Breen
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Maik Koedel
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Susann Sauer
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Vincenza Sportelli
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Monika Rex-Haffner
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Silvia Cappello
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Fabian J Theis
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| | - Elisabeth B Binder
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany (Cruceanu, Dony, Krontira, Roeh, Kaspar, Arloth, Czamara, Gerstner, Martinelli, Wehner, Koedel, Sauer, Sportelli, Rex-Haffner, Binder);International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich (Dony, Krontira, Kaspar, Gerstner);Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany (Dony, Fischer, Arloth, Theis);TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany (Fischer);Max Planck Institute of Psychiatry, Munich (Di Giaimo, Kyrousi, Cappello);Department of Biology, University of Naples Federico II, Naples, Italy (Di Giaimo);First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, and University Mental Health, Neurosciences, and Precision Medicine Research Institute "Costas Stefanis," Athens, Greece (Kyrousi);Department of Psychiatry, Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, and Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York (Breen);School of Life Sciences Weihenstephan and Department of Mathematics, Technical University of Munich, Munich (Theis);Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta (Binder)
| |
Collapse
|
40
|
Musillo C, Berry A, Cirulli F. Prenatal psychological or metabolic stress increases the risk for psychiatric disorders: the "funnel effect" model. Neurosci Biobehav Rev 2022; 136:104624. [PMID: 35304226 DOI: 10.1016/j.neubiorev.2022.104624] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 12/16/2022]
Abstract
Adverse stressful experiences in utero can redirect fetal brain development, ultimately leading to increased risk for psychiatric disorders. Obesity during pregnancy can have similar effects as maternal stress, affecting mental health in the offspring. In order to explain how similar outcomes may originate from different prenatal conditions, we propose a "funnel effect" model whereby maternal psychological or metabolic stress triggers the same evolutionarily conserved response pathways, increasing vulnerability for psychopathology. In this context, the placenta, which is the main mother-fetus interface, appears to facilitate such convergence, re-directing "stress" signals to the fetus. Characterizing converging pathways activated by different adverse environmental conditions is fundamental to assess the emergence of risk signatures of major psychiatric disorders, which might enable preventive measures in risk populations, and open up new diagnostics, and potentially therapeutic approaches for disease prevention and health promotion already during pregnancy.
Collapse
Affiliation(s)
- Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; PhD Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
41
|
Ahmad SI, Shih EW, LeWinn KZ, Rivera L, Graff JC, Mason WA, Karr CJ, Sathyanarayana S, Tylavsky FA, Bush NR. Intergenerational Transmission of Effects of Women's Stressors During Pregnancy: Child Psychopathology and the Protective Role of Parenting. Front Psychiatry 2022; 13:838535. [PMID: 35546925 PMCID: PMC9085155 DOI: 10.3389/fpsyt.2022.838535] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/23/2022] [Indexed: 12/22/2022] Open
Abstract
Objective Experiences of stress and adversity, such as intimate partner violence, confer risk for psychiatric problems across the life span. The effects of these risks are disproportionately borne by women and their offspring-particularly those from communities of color. The prenatal period is an especially vulnerable period of fetal development, during which time women's experiences of stress can have long-lasting implications for offspring mental health. Importantly, there is a lack of focus on women's capacity for resilience and potential postnatal protective factors that might mitigate these intergenerational risks and inform intervention efforts. The present study examined intergenerational associations between women's prenatal stressors and child executive functioning and externalizing problems, testing maternal parenting quality and child sex as moderators, using a large, prospective, sociodemographically diverse cohort. Methods We used data from 1,034 mother-child dyads (64% Black, 30% White) from the Conditions Affecting Neurocognitive Development and Learning in Early Childhood (CANDLE) pregnancy cohort within the ECHO PATHWAYS consortium. Women's prenatal stressors included stressful life events (pSLE) and intimate partner violence (pIPV). Measures of child psychopathology at age 4-6 included executive functioning and externalizing problems. Parenting behaviors were assessed by trained observers, averaged across two sessions of mother-child interactions. Linear regression models were used to estimate associations between women's prenatal stressors and child psychopathology, adjusting for confounders and assessing moderation effects by maternal parenting quality and child sex. Results Women's exposures to pSLE and pIPV were independently associated with child executive functioning problems and externalizing problems in fully-adjusted models. Maternal parenting quality moderated associations between pSLE and both outcomes, such that higher parenting quality was protective for the associations between women's pSLE and child executive functioning and externalizing problems. No moderation by child sex was found. Discussion Findings from this large, sociodemographically diverse cohort suggest women's exposures to interpersonal violence and major stressful events-common for women during pregnancy-may prenatally program her child's executive functioning and externalizing problems. Women's capacity to provide high quality parenting can buffer this intergenerational risk. Implications for universal and targeted prevention and early intervention efforts to support women's and children's wellbeing are discussed.
Collapse
Affiliation(s)
- Shaikh I. Ahmad
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Emily W. Shih
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Kaja Z. LeWinn
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Luisa Rivera
- Department of Anthropology, Emory University, Atlanta, GA, United States
| | - J. Carolyn Graff
- College of Nursing, The University of Tennessee Health Science Center, Memphis, TN, United States
- Center on Developmental Disabilities, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - W. Alex Mason
- Department of Preventative Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Catherine J. Karr
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, Seattle, WA, United States
| | - Frances A. Tylavsky
- Department of Preventative Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nicole R. Bush
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
42
|
Petrican R, Fornito A, Jones N. Psychological Resilience and Neurodegenerative Risk: A Connectomics-Transcriptomics Investigation in Healthy Adolescent and Middle-Aged Females. Neuroimage 2022; 255:119209. [PMID: 35429627 DOI: 10.1016/j.neuroimage.2022.119209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 11/25/2022] Open
Abstract
Adverse life events can inflict substantial long-term damage, which, paradoxically, has been posited to stem from initially adaptative responses to the challenges encountered in one's environment. Thus, identification of the mechanisms linking resilience against recent stressors to longer-term psychological vulnerability is key to understanding optimal functioning across multiple timescales. To address this issue, our study tested the relevance of neuro-reproductive maturation and senescence, respectively, to both resilience and longer-term risk for pathologies characterised by accelerated brain aging, specifically, Alzheimer's Disease (AD). Graph theoretical and partial least squares analyses were conducted on multimodal imaging, reported biological aging and recent adverse experience data from the Lifespan Human Connectome Project (HCP). Availability of reproductive maturation/senescence measures restricted our investigation to adolescent (N =178) and middle-aged (N=146) females. Psychological resilience was linked to age-specific brain senescence patterns suggestive of precocious functional development of somatomotor and control-relevant networks (adolescence) and earlier aging of default mode and salience/ventral attention systems (middle adulthood). Biological aging showed complementary associations with the neural patterns relevant to resilience in adolescence (positive relationship) versus middle-age (negative relationship). Transcriptomic and expression quantitative trait locus data analyses linked the neural aging patterns correlated with psychological resilience in middle adulthood to gene expression patterns suggestive of increased AD risk. Our results imply a partially antagonistic relationship between resilience against proximal stressors and longer-term psychological adjustment in later life. They thus underscore the importance of fine-tuning extant views on successful coping by considering the multiple timescales across which age-specific processes may unfold.
Collapse
Affiliation(s)
- Raluca Petrican
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, United Kingdom.
| | - Alex Fornito
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia
| | - Natalie Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, United Kingdom
| |
Collapse
|
43
|
von Gall C. The Effects of Light and the Circadian System on Rhythmic Brain Function. Int J Mol Sci 2022; 23:ijms23052778. [PMID: 35269920 PMCID: PMC8911243 DOI: 10.3390/ijms23052778] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
Life on earth has evolved under the influence of regularly recurring changes in the environment, such as the 24 h light/dark cycle. Consequently, organisms have developed endogenous clocks, generating 24 h (circadian) rhythms that serve to anticipate these rhythmic changes. In addition to these circadian rhythms, which persist in constant conditions and can be entrained to environmental rhythms, light drives rhythmic behavior and brain function, especially in nocturnal laboratory rodents. In recent decades, research has made great advances in the elucidation of the molecular circadian clockwork and circadian light perception. This review summarizes the role of light and the circadian clock in rhythmic brain function, with a focus on the complex interaction between the different components of the mammalian circadian system. Furthermore, chronodisruption as a consequence of light at night, genetic manipulation, and neurodegenerative diseases is briefly discussed.
Collapse
Affiliation(s)
- Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, 40225 Dusseldorf, Germany
| |
Collapse
|
44
|
Levhar M, Schonblum A, Arnon L, Michael Y, Sheelo LS, Eisner M, Hadar E, Meizner I, Wiznitzer A, Weller A, Koren L, Agay-Shay K. Residential greenness and hair cortisol levels during the first trimester of pregnancy. ENVIRONMENTAL RESEARCH 2022; 204:112378. [PMID: 34780787 DOI: 10.1016/j.envres.2021.112378] [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: 06/02/2021] [Revised: 10/24/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AND AIM Studies have shown that increased maternal cortisol level is associated with child adverse health outcomes. Hair cortisol (HC) is suitable for assessing long-term circulating cortisol concentration. Only two previous studies reported beneficial associations between cortisol and residential greenness during pregnancy and no study focused on the first trimester. Our aim was to evaluate the association between residential greenness and first trimester HC levels among pregnant women in Israel. METHODS Women were recruited during second and third trimesters. Hair samples were collected from the scalp and retrospective HC levels during the first trimester were quantified for 217 women. HC levels were natural log transformed and outliers were excluded. Based on geocoded birth address, small area sociodemographic status (SES) and mean residential surrounding greenness were calculated using high-resolution satellite-based Normalised Difference Vegetation Index (NDVI) data at 100, 300 and 500-m buffers in a cross-sectional approach. In addition, longitudinal exposure to mean greenness during a week preconception and during the first trimester were calculated. Missing covariates were imputed and linearity of the associations were evaluated. Generalized linear models were used to estimate the crude and adjusted associations controlled for the relevant covariates. RESULTS After exclusion of outliers, for 211 women, crude and adjusted beneficial associations between exposure to higher mean NDVI and HC levels were observed for all the exposure measures. An increase in 1 interquartile range of greenness (100 m buffer) was associated with a statistically significant lower estimated natural log mean HC level (-0.27 95% CI: -0.44; -0.11). The associations were robust to adjustment for covariates. The findings were consistent for different buffers, for the longitudinal approach, when all observations were included in the analysis and slightly stronger associations were observed for women with addresses geocoded at the home or street level. For most of the exposure measures, stronger associations were observed among those of lower sociodemographic status. CONCLUSION Our findings that more greenness associated with reduced maternal cortisol levels measured in the hair during the first trimester, could have substantial implications for urban planners and public health professional. If our observations will be replicated, it may present a useful avenue for public-health intervention to promote health through the provision of greenness exposure during early pregnancy, specifically to disadvantage populations.
Collapse
Affiliation(s)
- Maya Levhar
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel; Health and Environment Research (HER) Lab, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Anat Schonblum
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel; Health and Environment Research (HER) Lab, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Liat Arnon
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Yaron Michael
- Department of Geography and Environment, Bar-Ilan University, Ramat-Gan, Israel
| | - Liat Salzer Sheelo
- Helen Schneider Hospital for Women, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Eisner
- Helen Schneider Hospital for Women, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Hadar
- Helen Schneider Hospital for Women, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Israel Meizner
- Helen Schneider Hospital for Women, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arnon Wiznitzer
- Helen Schneider Hospital for Women, Rabin Medical Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Aron Weller
- Department of Psychology & Gonda Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Lee Koren
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Keren Agay-Shay
- Health and Environment Research (HER) Lab, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| |
Collapse
|
45
|
Calibration and recalibration of stress response systems across development: Implications for mental and physical health. ADVANCES IN CHILD DEVELOPMENT AND BEHAVIOR 2022; 63:35-69. [DOI: 10.1016/bs.acdb.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
46
|
Høgh S, Hegaard HK, Renault KM, Cvetanovska E, Kjærbye-Thygesen A, Juul A, Borgsted C, Bjertrup AJ, Miskowiak KW, Væver MS, Stenbæk DS, Dam VH, Binder E, Ozenne B, Mehta D, Frokjaer VG. Short-term oestrogen as a strategy to prevent postpartum depression in high-risk women: protocol for the double-blind, randomised, placebo-controlled MAMA clinical trial. BMJ Open 2021; 11:e052922. [PMID: 35763351 PMCID: PMC8719185 DOI: 10.1136/bmjopen-2021-052922] [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: 04/28/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Postpartum depression affects 10%-15% of women and has a recurrence rate of 40% in subsequent pregnancies. Women who develop postpartum depression are suspected to be more sensitive to the rapid and large fluctuations in sex steroid hormones, particularly estradiol, during pregnancy and postpartum. This trial aims to evaluate the preventive effect of 3 weeks transdermal estradiol treatment immediately postpartum on depressive episodes in women at high risk for developing postpartum depression. METHODS AND ANALYSIS The Maternal Mental Health Trial is a double-blind, randomised and placebo-controlled clinical trial. The trial involves three departments of obstetrics organised under Copenhagen University Hospital in Denmark. Women who are singleton pregnant with a history of perinatal depression are eligible to participate. Participants will be randomised to receive either transdermal estradiol patches (200 µg/day) or placebo patches for 3 weeks immediately postpartum. The primary outcome is clinical depression, according to the Diagnostic and Statistical Manual of Mental Disorders-V criteria of Major Depressive Disorder with onset at any time between 0 and 6 months postpartum. Secondary outcomes include, but are not limited to, symptoms of depression postpartum, exclusive breastfeeding, cortisol dynamics, maternal distress sensitivity and cognitive function. The primary statistical analysis will be performed based on the intention-to-treat principle. With the inclusion of 220 participants and a 20% expected dropout rate, we anticipate 80% power to detect a 50% reduction in postpartum depressive episodes while controlling the type 1 error at 5%. ETHICS AND DISSEMINATION The study protocol is approved by the Regional Committees on Health Research Ethics in the Capital Region of Denmark, the Danish Medicines Agency and the Centre for Data Protection Compliance in the Capital Region of Denmark. We will present results at scientific meetings and in peer-reviewed journals and in other formats to engage policymakers and the public. TRIAL REGISTRATION NUMBER NCT04685148.
Collapse
Affiliation(s)
- Stinne Høgh
- Department of Obstetrics and Gynaecology, Rigshospitalet, Copenhagen, Denmark
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | - Anders Juul
- Department of Growth and Reproduction, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Camilla Borgsted
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
- Department of Psychiatry, Rigshospitalet, Copenhagen, Denmark
| | | | - Kamilla Woznica Miskowiak
- Department of Psychiatry, Rigshospitalet, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | | | - Dea Siggaard Stenbæk
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | | | - Elisabeth Binder
- Department of Translational Research in Psychiatry, Max-Planck-Institute for Psychiatry, Munchen, Bayern, Germany
| | - Brice Ozenne
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
| | - Divya Mehta
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Vibe G Frokjaer
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
- Department of Psychiatry, Rigshospitalet, Copenhagen, Denmark
- Capital Region of Denmark Mental Health Services, Copenhagen, Denmark
| |
Collapse
|
47
|
Ksinan Jiskrova G, Pikhart H, Bobák M, Klanova J, Stepanikova I. Prenatal psychosocial stress and children's sleep problems: Evidence from the ELSPAC-CZ study. J Sleep Res 2021; 31:e13531. [PMID: 34879444 DOI: 10.1111/jsr.13531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/24/2021] [Accepted: 11/23/2021] [Indexed: 11/28/2022]
Abstract
Prenatal exposure to maternal stress may increase the risk of developing sleep problems in childhood. This study examined the association between prenatal stressful life events (PSLE) and children's sleep problems, taking into consideration their trajectory over time. Data were obtained from the Czech portion of the European Longitudinal Cohort Study of Pregnancy and Childhood (ELSPAC-CZ; N = 4,371 children). Mothers reported PSLE using an inventory of 42 life events and child sleep problems at five time-points (child age of 1.5, 3, 5, 7, and 11 years). The association was tested by a Poisson latent growth model, controlling for maternal and family demographics, birth characteristics, maternal depression, and alcohol use in pregnancy. The average rate of sleep problems was 2.06 (p < 0.001) at the age of 1.5 years and the rate of sleep problems decreased in a linear fashion over time (estimate = -0.118; p < 0.001). A higher number of PSLE was associated with a higher rate of sleep problems at the age of 1.5 years (incidence rate ratio [IRR] per interquartile range = 1.08, 95% confidence interval [CI] 1.05-1.12, p < 0.001) and with a reduced rate of decrease in sleep problems between the ages of 1.5 and 11 years (p < 0.001). Thus, PSLE were associated with chronicity of sleep problems in addition to their amount during early childhood. Prenatal exposure to stress may predispose individuals to the development of sleep problems in later life.
Collapse
Affiliation(s)
| | - Hynek Pikhart
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Epidemiology & Public Health, University College London, London, UK
| | - Martin Bobák
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Epidemiology & Public Health, University College London, London, UK
| | - Jana Klanova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Irena Stepanikova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Sociology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
48
|
The role of glucocorticoid receptor gene in the association between attention deficit-hyperactivity disorder and smaller brain structures. J Neural Transm (Vienna) 2021; 128:1907-1916. [PMID: 34609638 DOI: 10.1007/s00702-021-02425-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
ADHD is associated with smaller subcortical brain volumes and cortical surface area, with greater effects observed in children than adults. It is also associated with dysregulation of the HPA axis. Considering the effects of the glucocorticoid receptor (NR3C1) in neurophysiology, we hypothesize that the blurred relationships between brain structures and ADHD in adults could be partly explained by NR3C1 gene variation. Structural T1-weighted images were acquired on a 3 T scanner (N = 166). Large-scale genotyping was performed, and it was followed by quality control and pruning procedures, which resulted in 48 independent NR3C1 gene variants analyzed. After a stringent Bonferroni correction, two SNPs (rs2398631 and rs72801070) moderated the association between ADHD and accumbens and amygdala volumes in adults. The significant SNPs that interacted with ADHD appear to have a role in gene expression regulation, and they are in linkage disequilibrium with NR3C1 variants that present well-characterized physiological functions. The literature-reported associations of ADHD with accumbens and amygdala were only observed for specific NR3C1 genotypes. Our findings reinforce the influence of the NR3C1 gene on subcortical volumes and ADHD. They suggest a genetic modulation of the effects of a pivotal HPA axis component in the neuroanatomical features of ADHD.
Collapse
|
49
|
Environmental Alterations during Embryonic Development: Studying the Impact of Stressors on Pluripotent Stem Cell-Derived Cardiomyocytes. Genes (Basel) 2021; 12:genes12101564. [PMID: 34680959 PMCID: PMC8536136 DOI: 10.3390/genes12101564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/16/2022] Open
Abstract
Non-communicable diseases (NCDs) sauch as diabetes, obesity and cardiovascular diseases are rising rapidly in all countries world-wide. Environmental maternal factors (e.g., diet, oxidative stress, drugs and many others), maternal illnesses and other stressors can predispose the newborn to develop diseases during different stages of life. The connection between environmental factors and NCDs was formulated by David Barker and colleagues as the Developmental Origins of Health and Disease (DOHaD) hypothesis. In this review, we describe the DOHaD concept and the effects of several environmental stressors on the health of the progeny, providing both animal and human evidence. We focus on cardiovascular diseases which represent the leading cause of death worldwide. The purpose of this review is to discuss how in vitro studies with pluripotent stem cells (PSCs), such as embryonic and induced pluripotent stem cells (ESC, iPSC), can underpin the research on non-genetic heart conditions. The PSCs could provide a tool to recapitulate aspects of embryonic development “in a dish”, studying the effects of environmental exposure during cardiomyocyte (CM) differentiation and maturation, establishing a link to molecular mechanism and epigenetics.
Collapse
|
50
|
Czamara D, Dieckmann L, Röh S, Kraemer S, Rancourt RC, Sammallahti S, Kajantie E, Laivuori H, Eriksson JG, Räikkönen K, Henrich W, Plagemann A, Binder EB, Braun T, Entringer S. Betamethasone administration during pregnancy is associated with placental epigenetic changes with implications for inflammation. Clin Epigenetics 2021; 13:165. [PMID: 34446099 PMCID: PMC8393766 DOI: 10.1186/s13148-021-01153-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/12/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Glucocorticoids (GCs) play a pivotal role in fetal programming. Antenatal treatment with synthetic GCs (sGCs) in individuals in danger of preterm labor is common practice. Adverse short- and long-term effects of antenatal sGCs have been reported, but their effects on placental epigenetic characteristics have never been systematically studied in humans. RESULTS We tested the association between exposure to the sGC betamethasone (BET) and placental DNA methylation (DNAm) in 52 exposed cases and 84 gestational-age-matched controls. We fine-mapped associated loci using targeted bisulfite sequencing. The association of placental DNAm with gene expression and co-expression analysis on implicated genes was performed in an independent cohort including 494 placentas. Exposure to BET was significantly associated with lower placenta DNAm at an enhancer of FKBP5. FKBP5 (FK506-binding protein 51) is a co-chaperone that modulates glucocorticoid receptor activity. Lower DNAm at this enhancer site was associated with higher expression of FKBP5 and a co-expressed gene module. This module is enriched for genes associated with preeclampsia and involved in inflammation and immune response. CONCLUSIONS Our findings suggest that BET exposure during pregnancy associates with few but lasting changes in placental DNAm and may promote a gene expression profile associated with placental dysfunction and increased inflammation. This may represent a pathway mediating GC-associated negative long-term consequences and health outcomes in offspring.
Collapse
Affiliation(s)
- Darina Czamara
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
| | - Linda Dieckmann
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry, München, Germany
| | - Simone Röh
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
| | - Sarah Kraemer
- Institute of Medical Psychology, Charité − Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Luisenstr. 57, 10117 Berlin, Germany
| | - Rebecca C. Rancourt
- Department of Experimental Obstetrics, Charité − Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sara Sammallahti
- Department of Child and Adolescent Psychiatry, Erasmus MC, Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Eero Kajantie
- Finnish Institute for Health and Welfare, Helsinki, Finland
- Children’s Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Faculty of Medicine, PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Johan G. Eriksson
- Department of General Practice and Primary Care, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wolfgang Henrich
- Department of Obstetrics, Charité − Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andreas Plagemann
- Department of Experimental Obstetrics, Charité − Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
- Department of Obstetrics, Charité − Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Elisabeth B. Binder
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329 USA
| | - Thorsten Braun
- Department of Experimental Obstetrics, Charité − Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
- Department of Obstetrics, Charité − Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sonja Entringer
- Institute of Medical Psychology, Charité − Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Luisenstr. 57, 10117 Berlin, Germany
- Development, Health, and Disease Research Program, University of California, Irvine, Orange, CA USA
| |
Collapse
|