1
|
Fioretto MN, Barata LA, de Andrade Felipe VA, Dos Santos SAA, Maciel FA, Ribeiro IT, Mattos R, Baptista HS, Bueno G, Fagundes FL, Portela LMF, Scarano WR, Seiva FRF, Lima CAH, Justulin LA. Long-term effects of maternal protein restriction on adrenal proteomic profile and steroidogenesis in male offspring rats. Cell Signal 2025; 130:111707. [PMID: 40032160 DOI: 10.1016/j.cellsig.2025.111707] [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: 01/12/2025] [Revised: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Maternal protein restriction (MPR) can significantly affect offspring's early development and aging, impacting several organs, including the adrenal glands. This study evaluated the adrenal proteomic profile in male rat offspring exposed to MPR during pregnancy and lactation. Male offspring were divided into two groups: Control (CTR), born to dams fed a normoprotein diet (17 % protein), and Gestational and Lactational Low-Protein (GLLP), born to dams fed a low-protein diet (6 % protein) throughout gestation and lactation, and after received control diet. Offspring were euthanized at postnatal day (PND) 21 or PND 540. Blood samples and adrenal glands were processed for histological, metabolic, molecular, and proteomic assessments. At PND21, the GLLP group exhibited reduced adrenal gland mass and cortical thickness. At PND21, the proteomic landscape showed that the most impacted biological pathways were associated with decreased steroid hormone synthesis, increased glucose metabolism, and stress response. At PND540, the main impacts were increased apoptotic pathway, stress response, and steroid hormone synthesis, with decreased glucose metabolism. At PND 540, the GLLP group showed higher adrenal collagen content and elevated apoptosis. Age-related changes included decreased peroxiredoxin 3 and increased expression of aldosterone synthase (Cyp11b2). Furthermore, steroid 11-Beta-Hydroxylase (Cyp11b1) expression decreased at PND540, alongside reduced serum aldosterone and elevated serum corticosterone levels. These results suggest that MPR modulates the adrenal glands' proteomic profile, serving as a pivotal mechanism underpinning diverse systemic diseases. It influences adrenal morphophysiology early in life, with long-lasting consequences for cellular stress, immune response, and catabolic pathways in male offspring with aging.
Collapse
Affiliation(s)
- Matheus Naia Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luísa Annibal Barata
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | | | - Sérgio Alexandre Alcantara Dos Santos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil.; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Flávia Alessandra Maciel
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Isabelle Tenori Ribeiro
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Renato Mattos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Hecttor Sebástian Baptista
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Gabriela Bueno
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Felipe Leonardo Fagundes
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luiz Marcos Frediane Portela
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Wellerson Rodrigo Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Clélia Akiko Hiruma Lima
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil..
| |
Collapse
|
2
|
Li B, Gao H, Xiao H, He H, Ni Q, Li Q, Wang H, Chen L. Abnormal chenodexycholic acid metabolism programming promotes cartilage matrix degradation in male adult offspring rats induced by prenatal caffeine exposure. Toxicol Res (Camb) 2025; 14:tfaf063. [PMID: 40331087 PMCID: PMC12051868 DOI: 10.1093/toxres/tfaf063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/01/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
Epidemiological evidence links osteoarthritis to fetal origins. Our study shows prenatal caffeine exposure (PCE) in rats predisposes adult offspring to osteoarthritis, associated with elevated intrauterine glucocorticoid levels. Previous research indicates that chenodeoxycholic acid (CDCA), a bile acid, can slow osteoarthritis progression when administered intra-articularly. This study explored if disrupted bile acid metabolism in cartilage affects osteoarthritis risk in adult offspring with PCE. Our findings indicate that the expression of MMP3/MMP13 was upregulated, while endogenous CDCA levels were reduced in the cartilage of PCE-exposed offspring. Furthermore, we observed a persistent reduction in H3K27ac levels at the CYP7B1 promoter and its expression in the cartilage of PCE offspring from fetus to adulthood. Moreover, a sub-physiological level of CDCA promoted NF-κB phosphorylation and the expression of MMP3/MMP13 in chondrocytes in vitro. High levels of glucocorticoids reduced H3K27ac levels and CYP7B1 expression in the promoter region of CYP7B1 through the glucocorticoid receptor and histone deacetylase 4, consequently leading to decreased CDCA levels. In summary, our findings suggest that intrauterine low-expression programming of CYP7B1, induced by elevated glucocorticoid levels, reduces local CDCA levels in the cartilage of PCE offspring, ultimately leading to increased matrix degradation and susceptibility to osteoarthritis.
Collapse
Affiliation(s)
- Bin Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Hui Gao
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hao Xiao
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Hangyuan He
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qubo Ni
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qingxian Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
- Department of Pharmacology, Wuhan University Taikang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| |
Collapse
|
3
|
Sultana S, Bruinsma F, Fisher Z, Homer CSE, Vogel JP. Long-term outcomes of antenatal corticosteroids for preterm birth: An overview of systematic reviews. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004575. [PMID: 40333673 PMCID: PMC12057917 DOI: 10.1371/journal.pgph.0004575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 04/08/2025] [Indexed: 05/09/2025]
Abstract
Antenatal corticosteroids (ACS) is a critical preterm birth intervention that reduces neonatal mortality and short-term morbidities when administered to women at risk of preterm birth <34 weeks' gestation. While numerous systematic reviews have evaluated the effects of ACS on child health outcomes, this body of evidence has not been comprehensively assessed. This overview of systematic reviews thus aimed to comprehensively summarize the findings on the effects of ACS for preterm birth on long-term outcomes in children. We searched six electronic databases, with no date and language restrictions. Systematic reviews of randomized, non-randomized trials or observational studies that evaluated childhood health outcomes (assessed at age 12 months or older) for any ACS exposure compared to no ACS exposure, including comparisons between different types and regimens of ACS were eligible. We used the AMSTAR-2 tool to assess the quality of the included reviews. We included 19 systematic reviews. We identified a wide range of outcomes across the included reviews and categorized them as neurodevelopmental, psychological, physical growth, respiratory, cardiovascular, and survival/mortality outcomes. The reported outcomes were substantially varied in terms of operational definitions, terminology, timing of assessment, and measurement. No benefits or harms were observed for most of the reported outcomes following ACS exposure, though many outcomes had few participants. Available evidence suggested that a single course of ACS possibly has positive effects on selected neurodevelopmental outcomes. However, ACS exposure might also have adverse effects on psychological and some neurodevelopmental outcome in children born at late preterm or at term. Heterogeneity in outcome measurements and reporting, including overall confidence in results, contributes to uncertainties about long-term effect of ACS and warrants caution while interpreting the findings. Further high-quality research is required to generate robust evidence base on the effects of ACS on long-term child health outcomes.
Collapse
Affiliation(s)
- Saima Sultana
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
- Women’s, Children’s and Adolescents’ Health Program, Burnet Institute, Melbourne, Australia
| | - Fiona Bruinsma
- Women’s, Children’s and Adolescents’ Health Program, Burnet Institute, Melbourne, Australia
| | - Zeshi Fisher
- Women’s, Children’s and Adolescents’ Health Program, Burnet Institute, Melbourne, Australia
| | - Caroline S. E. Homer
- Women’s, Children’s and Adolescents’ Health Program, Burnet Institute, Melbourne, Australia
| | - Joshua P. Vogel
- Women’s, Children’s and Adolescents’ Health Program, Burnet Institute, Melbourne, Australia
| |
Collapse
|
4
|
Casciaro C, Hamada H, Bloise E, Matthews SG. The paternal contribution to shaping the health of future generations. Trends Endocrinol Metab 2025; 36:459-471. [PMID: 39562264 DOI: 10.1016/j.tem.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
Paternal health and exposure to adverse environments in the period prior to conception have a profound impact on future generations. Adversities such as stress, diet, and toxicants influence offspring health. Emerging evidence indicates that epigenetic mechanisms including noncoding RNA, DNA methylation, and chromatin remodelling mediate these effects. Preclinical studies have contributed to advancing mechanistic understanding in the field; however, human research is limited and primarily observational. Here, we discuss the evidence linking paternal to offspring health and advocate for further research in this area, which may ultimately inform policy and healthcare guidelines to improve paternal preconception health and offspring outcomes.
Collapse
Affiliation(s)
| | - Hirotaka Hamada
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Enrrico Bloise
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada.
| |
Collapse
|
5
|
Miller TM, Navara KJ. Prenatal Corticosterone Impacts Nestling Condition and Immunity in Eastern Bluebirds. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2025; 343:427-437. [PMID: 39831630 PMCID: PMC11874072 DOI: 10.1002/jez.2898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/04/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Exposure of avian mothers to stressful conditions permanently alters offspring behavior and physiology. Yet, the effects of maternal stress on the development of offspring immunity in birds remain unclear, particularly in wild species. We injected Eastern bluebird (Sialia sialis) eggs with either a corticosterone or control solution, then measured the impacts on nestling morphology and two measures of immunity, bactericidal capacity and swelling responses to phytohemagglutinin. Nestlings from corticosterone-treated eggs had lower condition indices at hatch but quickly caught up to their control counterparts by Day 5 posthatch and until fledging. Corticosterone-exposed nestlings also mounted smaller swelling responses to phytohemagglutinin, whereas there were no effects on bactericidal capacity. These results indicate that maternal stress can impact offspring immunocompetence, fitness prospects, and potentially their ability to fend off parasites and pathogens.
Collapse
Affiliation(s)
- Taylor M. Miller
- Department of Poultry ScienceThe University of GeorgiaAthensGeorgiaUSA
| | - Kristen J. Navara
- Department of Poultry ScienceThe University of GeorgiaAthensGeorgiaUSA
| |
Collapse
|
6
|
Koch K, Schlüppmann K, Hüsken S, Stark LM, Förster N, Masjosthusmann S, Klose J, Dönmez A, Fritsche E. Nuclear hormone receptors control fundamental processes of human fetal neurodevelopment: Basis for endocrine disruption assessment. ENVIRONMENT INTERNATIONAL 2025; 198:109400. [PMID: 40147140 DOI: 10.1016/j.envint.2025.109400] [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: 11/24/2024] [Revised: 02/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Despite growing awareness of endocrine disrupting chemicals (EDCs), knowledge gaps remain regarding their effects on human brain development. EDC risk assessment focuses primarily on EATS modalities (estrogens, androgens, thyroid hormones, and steroidogenesis), overlooking the broader range of hormone receptors expressed in the developing brain. This limits the evaluation of chemicals for their potential to cause endocrine disruption-mediated developmental neurotoxicity (ED-DNT). The Neurosphere Assay, an in vitro test method for developmental neurotoxicity (DNT) evaluation, is an integral component of the DNT in vitro testing battery, which has been used to screen a broad domain of environmental chemicals. Here, we define the endocrine-related applicability domain of the Neurosphere Assay by assessing the impact and specificity of 14 hormone receptors on seven key neurodevelopmental processes (KNDPs), neural progenitor cell (NPC) proliferation, migration of radial glia, neurons, and oligodendrocytes, neurite outgrowth, and differentiation of neurons and oligodendrocytes. Comparative analyses in human and rat NPCs of both sexes revealed species- and sex-specific responses. Mechanistic insights were obtained through RNA sequencing and agonist/antagonist co-exposures. Most receptor agonists modulated KNDPs at concentrations in the range of physiologically relevant hormone concentrations. Phenotypic effects induced by glucocorticoid receptor (GR), liver X receptor (LXR), peroxisome proliferator-activated receptor beta/delta (PPARβδ), retinoic acid receptor (RAR) and retinoid X receptor (RXR) activation were counteracted by receptor antagonists, confirming specificity. Transcriptomics highlighted receptor crosstalk and the involvement of conserved developmental pathways (e.g. Notch and Wnt). Species comparisons identified limited concordance in hormone receptor-regulated KNDPs between human and rat NPCs. This study presents novel findings on cellular and molecular hormone actions in human fetal NPCs, highlights major species differences, and illustrates the Neurosphere Assay's relevance for detecting endocrine MoAs, supporting its application in human-based ED-DNT risk assessment.
Collapse
Affiliation(s)
- Katharina Koch
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; DNTOX GmbH, Duesseldorf, Germany.
| | - Kevin Schlüppmann
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Saskia Hüsken
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Louisa Merit Stark
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Nils Förster
- Bioinformatics Group, Faculty for Biology and Biotechnology, Ruhr-University Bochum, Germany; Center for Protein Diagnostics (ProDi), Ruhr-University Bochum, Bochum, Germany
| | | | - Jördis Klose
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; DNTOX GmbH, Duesseldorf, Germany
| | - Arif Dönmez
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; DNTOX GmbH, Duesseldorf, Germany
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; DNTOX GmbH, Duesseldorf, Germany; Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany; SCAHT - Swiss Centre for Applied Human Toxicology, Basel, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
7
|
Kim B, Kostaki A, Matthews SG. Conserved DNA methylation signatures in the prefrontal cortex of female newborn and juvenile guinea pigs following antenatal betamethasone exposure. J Neuroendocrinol 2025; 37:e13499. [PMID: 39924870 PMCID: PMC11975801 DOI: 10.1111/jne.13499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/08/2025] [Accepted: 01/26/2025] [Indexed: 02/11/2025]
Abstract
Antenatal corticosteroids (ACS) improve perinatal survival when there is a risk of preterm birth. Although evidence suggests an increased risk of developing neurobehavioural disorders in exposed offspring, the mechanisms involved remain largely unknown. Here, we investigated the DNA methylation patterns in the prefrontal cortex (PFC) of ACS-exposed guinea pig offspring. We hypothesized that differential methylation will be evident at both newborn and juvenile ages. In two separate cohorts, pregnant guinea pigs were administered a subcutaneous injection of saline or betamethasone (1 mg/kg) on gestational days 50/51 to mimic a single course of ACS. The gDNA was isolated from the PFC of term-born female offspring on postnatal day 1 (PND1, n = 7/group) and PND14 (n = 6-7/group) to identify differentially methylated CpG sites (DMCs) using reduced representative bisulphite sequencing. In the PND1 PFC, 1521 DMCs, annotating 144 genes were identified following ACS. Identified genes are involved in pathways regulating 'developmental cellular process'. In the PND14 PFC, 776 DMCs representing 46 genes were identified and enriched in 'synaptic signalling' pathways. Though no individual DMCs were identified at both PND1 and PND14, differential methylation was consistently observed at the binding sites of transcription factors PLAGL1, TFAP2C, ZNF263 and SP1 at both ages. We have established that ACS exposure leads to altered DNA methylation in the PFC of guinea pig offspring at both newborn and juvenile ages. Notably, a unique methylation signature was consistently observed at four key transcription factor binding sites at both post-natal time points. These changes may predispose the development of altered neurobehavioural phenotypes that have been described in ACS-exposed offspring.
Collapse
Affiliation(s)
- Bona Kim
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
- Lunenfeld‐Tanenbaum Research InstituteSinai Health SystemTorontoOntarioCanada
| | - Alice Kostaki
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
| | - Stephen G. Matthews
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
- Lunenfeld‐Tanenbaum Research InstituteSinai Health SystemTorontoOntarioCanada
- Department of Obstetrics & GynecologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
8
|
Carter SWD, Kemp MW. A review of the potential off-target effects of antenatal steroid exposures on fetal development. J Dev Orig Health Dis 2025; 16:e18. [PMID: 40135629 DOI: 10.1017/s2040174425000078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Antenatal steroids (ANS) are one of the most widely prescribed medications in pregnancy, being administered to women at risk of preterm delivery. In the setting of preterm delivery at or below 35 weeks' gestation, systematic review data show ANS reduce perinatal morbidity and mortality, primarily by promoting fetal lung maturation. However, with the expanding use of this intervention has come a growing appreciation for the potential off-target, adverse effects of ANS therapy on wider fetal development. We undertook a narrative literature review of the animal and clinical literature to assess current evidence for adverse effects of ANS exposure and fetal development. This review presents a summary of the evidence relating to the potential for wide-ranging, off-target, adverse effects of ANS therapy on fetal development and programming. We highlight an urgent need for further animal and clinical studies investigating the effects of ANS on the fetal immune, cardiovascular, renal and hepatic systems given a current sparsity of evidence. We also strongly suggest an emphasis on open disclosure, discussion and education of clinicians and patients with regard to the potential benefits and risks of ANS therapy, particularly in late preterm and term gestations where infants derive relatively few benefits from these drugs. We also propose further studies on the optimisation of ANS therapy through improved patient selection and improved dosing regimens based on a pharmacokinetic-pharmacodynamic informed understanding of ANS action on the fetal lung.
Collapse
Affiliation(s)
- Sean W D Carter
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore
- King Edward Memorial Hospital, Perth, Western Australia, Australia
- Women and Infants Research Foundation, Perth, Western Australia, Australia
| | - Matthew W Kemp
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore
- Women and Infants Research Foundation, Perth, Western Australia, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
9
|
Vasquez LS, Stack S, Taylor WW, Dias BG. Intergenerational Effects of Stress - A Focus on Learning and Memory. Curr Top Behav Neurosci 2025. [PMID: 40119217 DOI: 10.1007/7854_2025_578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
Stress is a ubiquitous facet of life. Ranging in form (e.g., psychosocial, physical, nutritional, economic) and longevity (e.g., acute, chronic), stressors affect the biology of those directly in their line of attack. As is becoming increasingly appreciated, the pernicious effects of stress echo across generations (Dias et al. 2015; Yehuda and Lehrner 2018; Jawaid et al. 2021; Dion et al. 2022; Zhou and Ryan 2023; Dias 2024). With a focus on learning and memory, this chapter addresses how stressors derail learning and memory in the generation directly exposed to them andin future generations. To do so, with a specific emphasis on associative fear conditioning in humans and rodents, we touch upon the relevance of extinction training in the aftermath of such conditioning and the recall of such extinction training as windows into normative and disrupted learning. Next, we briefly discuss underlying neuroanatomical substrates mediating these processes. We then draw attention to influences of postnatal, in utero, and pre-conceptional stress on learning and memory across generations. Finally, we briefly outline biological factors that underlie how learning and memory is derailed by these stressors.
Collapse
Affiliation(s)
- L S Vasquez
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - S Stack
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - W W Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - B G Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA.
- Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Fowden AL, Vaughan OR, Forhead AJ. Early-life programming of livestock metabolism by glucocorticoids. J Dev Orig Health Dis 2025; 16:e16. [PMID: 40104937 DOI: 10.1017/s2040174425000091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Adverse environmental conditions during early life are known to determine adult metabolic phenotype in laboratory species and human populations. However, less is known about developmental programming of adult metabolic phenotype in livestock, given their size and longevity compared to laboratory animals. As maternal and/or fetal glucocorticoid (GC) concentrations rise in stressful conditions during pregnancy, GCs may act as a common mechanism linking early-life environmental conditions to the subsequent metabolic phenotype. This review examines prenatal and longer-term postnatal programming of metabolism by early-life GC overexposure in livestock species with a particular emphasis on sheep. It examines the effects of both cortisol, the natural glucocorticoid and more potent synthetic GCs used clinically to treat threatened pre-term delivery and other conditions during pregnancy. It considers the effects of early- life GC overexposure on the metabolism of specific feto-placental and adult tissues in relation to changes in the growth trajectory, other metabolic hormones and in the functioning of the hypothalamic-pituitary-adrenal axis itself. It highlights the role of GCs as maturational and environmental signals in programming development of a metabolic phenotype fit for survival at birth and future homeostatic challenges. However, the ensuing metabolic phenotype induced by early GC overexposure may become inappropriate for the prevailing postnatal conditions and lead to metabolic dysfunction as functional reserves decline with age. Further studies are needed in livestock to establish whether the metabolic outcomes of early-life GC overexposure are sex-linked, more pronounced in old age and inherited transgenerationally in these species.
Collapse
Affiliation(s)
- Abigail L Fowden
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Owen R Vaughan
- Institute of Women's Health, University College London, London, UK
| | - Alison J Forhead
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| |
Collapse
|
11
|
Fatori D, Shephard E, Benette D, Naspolini NF, Guzman GC, Wang JYT, Tótolo P, Mafra AL, Isaias C, Dos Santos DP, Russo FB, Kobayashi G, Argeu A, Teixeira M, Mattiello-Sverzut AC, Fernandes MTB, Petian-Alonso DC, Brentani H, Scliar M, Schüroff PA, Zuccolo P, Lerner R, Geraldini S, Euclydes VLV, Matijasevich A, de Campos AC, de Carvalho ACP, Fujita A, Taddei CR, Passos-Bueno MR, Beltrão-Braga P, Polanczyk GV. Identifying biomarkers and trajectories of executive functions and language development in the first 3 years of life: Design, methods, and findings of the Germina cohort study. Dev Psychopathol 2025:1-11. [PMID: 40040568 DOI: 10.1017/s0954579425000069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
This paper reports the methods and preliminary findings of Germina, an ongoing cohort study to identify biomarkers and trajectories of executive functions and language development in the first 3 years of life. 557 mother-infant dyads (mean age of mothers 33.7 years, 65.2% white, 48.7% male infants) have undergone baseline and are currently collecting data for other timepoints. A linear regression was used to predict baseline Bayley-III using scores derived from data-driven sparse partial least squares utilizing a multiple holdout framework of 15 domains. Significant associations were found between socioeconomic/demographic characteristics (B = 0.29), epigenetics (B = 0.11), EEG theta (B = 0.14) and beta activity (B = 0.11), and microbiome functional pathways (B = 0.08) domains, and infant development measured by the Bayley-III at T1, suggesting potential interventions to prevent impairments.
Collapse
Affiliation(s)
- Daniel Fatori
- Departamento de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Laboratorio de Psicopatologia e Terapeutica Psiquiatrica LIM-23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Elizabeth Shephard
- Departamento de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Laboratorio de Psicopatologia e Terapeutica Psiquiatrica LIM-23, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Departamento de Psicologia da Aprendizagem do Desenvolvimento e da Personalidade, Instituto de Psicologia, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Danilo Benette
- Departamento de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Grover Castro Guzman
- Instituto de Matemática e Estatística, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jaqueline Yu Ting Wang
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Sao Paulo, Brazil
| | - Pedro Tótolo
- Instituto de Matemática e Estatística, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Anthonieta Looman Mafra
- Departamento de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Caio Isaias
- Centro de Matemática, Computação e Cognição, São Bernardo do Campo, Universidade Federal do ABC, Sao Paulo, Brazil
| | - Davi Pereira Dos Santos
- Instituto de Ciências Matemáticas e de Computação, Universidade de São Paulo, São Carlos, Brazil
| | - Fabiele Baldino Russo
- Departamento de Microbiologia, Instituto de Ciências Biomédicas ICB, Universidade de São Paulo, São Paulo, Brazil
| | - Gerson Kobayashi
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Sao Paulo, Brazil
| | - Adriana Argeu
- Departamento de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Monike Teixeira
- Departamento de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ana Claudia Mattiello-Sverzut
- Departamento de Ciências da Saúde, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Danila Cristina Petian-Alonso
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Ribeirão Preto, Brazil
| | - Helena Brentani
- Departamento de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Marilia Scliar
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Sao Paulo, Brazil
| | - Paulo Alfonso Schüroff
- Escola de Artes, Ciências e Humanidades EACH, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Pedro Zuccolo
- Departamento de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Rogério Lerner
- Departamento de Psicologia da Aprendizagem do Desenvolvimento e da Personalidade, Instituto de Psicologia, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Stephania Geraldini
- Departamento de Psicologia da Aprendizagem do Desenvolvimento e da Personalidade, Instituto de Psicologia, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Alicia Matijasevich
- Departamento de Medicina Preventiva, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Alline Cristina de Campos
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - André Fujita
- Instituto de Matemática e Estatística, Universidade de Sao Paulo, Sao Paulo, Brazil
- Division of Network AI Statistics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Carla R Taddei
- Escola de Artes, Ciências e Humanidades EACH, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Maria Rita Passos-Bueno
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Sao Paulo, Brazil
| | - Patricia Beltrão-Braga
- Departamento de Microbiologia, Instituto de Ciências Biomédicas ICB, Universidade de São Paulo, São Paulo, Brazil
- Institut Pasteur de São Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
12
|
Chawanpaiboon S, Wutthigate P, Anuwutnavin S, Sutchritpongsa S. Double-blind, non-inferiority, randomized controlled trial of dexamethasone 4, 5 and 6 mg for preventing adverse neonatal and maternal outcomes in very preterm to late preterm pregnancies between 29 0 and 36 6 weeks of gestation: study protocol. Reprod Health 2025; 22:30. [PMID: 40025486 PMCID: PMC11874782 DOI: 10.1186/s12978-025-01965-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 02/04/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Premature birth poses significant health challenges, including respiratory distress syndrome (RDS). Corticosteroids reduce the incidence of RDS, but higher dexamethasone doses may lead to adverse neonatal outcomes, such as growth restriction and neurodevelopmental issues. Determining the appropriate dose is crucial to balance efficacy and safety. Dexamethasone is inexpensive and widely available in most low- and middle-income countries. This study aims to compare the efficacy and safety of 4-mg, 5-mg and 6-mg dexamethasone in preventing RDS among preterm infants. This trial aims to determine whether lower dexamethasone doses are as effective as the standard dose in preventing RDS in preterm infants. By assessing efficacy and potential adverse outcomes, this study will provide critical insights for optimizing treatment protocols and improving neonatal care. METHODS This randomized controlled trial will include pregnant women with gestational ages between 290 and 366 weeks admitted to Siriraj Hospital and three secondary centres in Thailand. The participants will be randomly assigned to receive intramuscular dexamethasone at 4 mg, 5 mg or 6 mg, which will be administered every 12 h for a total of four doses over 48 h. The same dose will be used for rescue or repeat courses. The primary outcome will be the incidence of RDS, defined by clinical criteria and confirmed by a neonatologist. The secondary outcomes will include other adverse neonatal and maternal outcomes. RESULTS The study requires 1,560 participants, accounting for a 15% loss to follow-up. The data will be analysed via descriptive statistics, chi-squared tests for categorical data, and one-way ANOVA or Kruskal-Wallis tests for continuous data. An independent Data Safety Monitoring Board will conduct interim analyses every 3 months to ensure participant safety and study integrity. DISCUSSION This trial addresses the gap in research regarding optimal dexamethasone dosing for preventing RDS in preterm infants. The study will provide evidence on whether lower doses of dexamethasone (4 and 5 mg) are as effective as the standard 6-mg dose and will examine their potential adverse outcomes. The results will guide adjustments to medical practice guidelines, aiming to align them with clinical practices while ensuring safety and efficacy. Trial registration page https://www.thaiclinicaltrials.org/export/pdf/TCTR20220511003 10/05/2022.
Collapse
Affiliation(s)
- Saifon Chawanpaiboon
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynaecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Punnanee Wutthigate
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sanitra Anuwutnavin
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynaecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sureelak Sutchritpongsa
- Division of Child Development and Behaviour, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| |
Collapse
|
13
|
Li X, Xiong Y, Ailikaiti A, Ao Y, Kou H, Wang H. Toxic effects of prenatal azithromycin exposure on fetal adrenal gland in mice: The role of stage, dose and course of treatment. Toxicol Appl Pharmacol 2025; 496:117244. [PMID: 39890031 DOI: 10.1016/j.taap.2025.117244] [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: 10/30/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Azithromycin is widely used in treating bacterial infections during pregnancy. Previous studies suggest prenatal exposure (PAzE) induces embryonic developmental toxicity. However, the influence of PAzE on fetal adrenal gland development is unknown. Pregnant mice received azithromycin in varying ways: different stages (mid- and late-pregnancy), doses (50, 100, and 200 mg/kg d), and courses (single- and multi-course). Adrenal gland morphology, cell proliferation, apoptosis, steroid synthesis, and expression of key transcriptional factors were examined. PAzE predominantly affected fetal adrenal gland development in males, characterized by obvious pathological changes (irregular arrangement and decreased density of adrenocortical cells, aggravated cytoplasmic vacuolization), weakened cell proliferation (decreased Pcna but increased Caspase3 expression), and inhibited steroidogenesis (reduced expression of Star, 3β-hsd, P450c21, and P450c11). The most significant damage occurred with multi-course high-dose (clinical dose) azithromycin treatment in late-pregnancy, possibly linked to inhibited Cited2 expression. This study delineated the sex-specific toxic effects of PAzE on fetal adrenal gland development, influenced by various stages, doses, and courses of azithromycin treatment. These findings contribute to a better understanding of azithromycin's safe use during pregnancy and offer a crucial theoretical and experimental foundation for future research.
Collapse
Affiliation(s)
- Xiaomin Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Ying Xiong
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Aihemaitijiang Ailikaiti
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Ying Ao
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hao Kou
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, 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
|
14
|
Rychlik KA, Illingworth EJ, Sillé FCM. Arsenic and the placenta: A review with emphasis on the immune system. Placenta 2025; 160:73-81. [PMID: 39778256 PMCID: PMC11927389 DOI: 10.1016/j.placenta.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Chronic arsenic exposure affects over 140 million people globally. While arsenic easily crosses the placenta, the specific mechanisms impacting placental immune cell populations and fetal health are unclear. Maternal arsenic exposure is epidemiologically linked to increased infection risk, mortality, and cancer susceptibility in offspring, emphasizing the importance of understanding placentally-mediated immune effects. This review explores the potential role of the placenta, a key organ for immune transfer to the developing fetus, in mediating chronic low-dose arsenic exposure effects. Examining three potential pathways-direct contaminant transfer, altered immune transfer from the mother, and indirect impact on fetal immune programming via maternal and placental signaling-the review highlights studies associating maternal arsenic levels with immune-related outcomes, including changes in cord blood T cell populations and increased placental inflammation. Placental gene expression analysis reveals alterations in pathways related to oxidative stress, proteasome activity, and aquaglyceroporin transporter expression. Impact on placental DNA methylation and microRNA regulation as well as on trophoblast dysfunction is discussed, with evidence suggesting inhibited trophoblast migration and placental growth factor expression. The complexity of mixtures, nutrition, and environmental interactions add challenges to investigating the placenta's role in immune programming. Despite inconsistent findings on placental morphology alterations, evidence suggests a potential link between arsenic exposure, placental anomalies, and adverse birth outcomes. Further research is crucial to comprehend the effects of prenatal arsenic exposure on trophoblasts, placental immune cells, and subsequent long-term consequences for fetal immune development and birth outcomes.
Collapse
Affiliation(s)
- Kristal A Rychlik
- Public Health Program, School of Health Professions, University of Mary Hardin-Baylor, USA; Johns Hopkins University Bloomberg School of Public Health, USA
| | - Emily J Illingworth
- Johns Hopkins University Bloomberg School of Public Health, USA; iQ Biosciences, Alameda, CA, USA
| | - Fenna C M Sillé
- Johns Hopkins University Bloomberg School of Public Health, USA.
| |
Collapse
|
15
|
Romaniuk AC, Barnard S, Shreyer T, Croney C. Effects of dam fear and stress on metrics of puppy welfare in commercial breeding kennels. Sci Rep 2025; 15:2820. [PMID: 39843634 PMCID: PMC11754604 DOI: 10.1038/s41598-025-85936-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
It is well established that maternal factors can affect the abilities of offspring to cope with stressors and can influence their overall welfare states. However, maternal effects have not been extensively explored in US commercial breeding kennels (CBKs). Therefore, the objective of this study was to identify if fear and stress in dams affected puppy welfare metrics in CBKs. Bitches (n = 90) were tested at 6 weeks prepartum (6 W Pre), and again with their puppies (n = 390) at 4 (4 W Post) and 8 weeks (8 W Post) postpartum. Dams and puppies underwent stranger approach and isolation tests, and their feces were collected to measure fecal glucocorticoid metabolite (FGM) and secretory immunoglobulin A concentrations. Further, dams' hair cortisol concentrations (HCC) were analyzed at the previously mentioned time points and at 1 week prepartum. Finally, birth and weekly weights were collected from puppies, and litter health metrics were recorded. Data were analyzed using mixed-effects and simple linear regression models. There were significant positive associations between dams' exploration and stationary durations and puppies' durations of the same respective behaviors during the isolation tests (exploration: [Formula: see text]2(1) = 9.472, p = 0.002; stationary: [Formula: see text]2(1) = 5.226, p = 0.022), 8 W Post dam FGMs and 8 W Post puppy FGMs (estimate: 0.0003, SE = 0.0001, p = 0.002), and 4 W Post dam HCCs and 4 W Post litter FGMs (estimate: 0.052, SE = 0.025, p = 0.053). Significant negative associations between 6 W Pre dam HCCs and 8 W Post puppy FGMs (estimate: -0.021, SE = 0.007, p = 0.007), puppies' birth weights ([Formula: see text]2(1) = 3.908, p = 0.048), and puppies' average weekly weight gains ([Formula: see text]2(1) = 0.111, p = 0.739) were also found. These findings suggest that indicators of dam fear and stress may be associated with potential indicators of puppies' welfare states in CBKs. Findings provide new knowledge on fear and stress-related factors that may be used to support the welfare of dams and puppies in CBKs and other populations.
Collapse
Affiliation(s)
- Aynsley C Romaniuk
- Department of Comparative Pathobiology, Purdue University, 725 Harrison Street, West Lafayette, IN, 47906, USA
| | - Shanis Barnard
- Department of Comparative Pathobiology, Purdue University, 725 Harrison Street, West Lafayette, IN, 47906, USA
| | - Traci Shreyer
- Department of Comparative Pathobiology, Purdue University, 725 Harrison Street, West Lafayette, IN, 47906, USA
| | - Candace Croney
- Center for Animal Welfare Science, Departments of Comparative Pathobiology and Animal Science, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
16
|
Xiong Y, Chen Z, Xiang H, Liu Y, Wang Y. Polystyrene microplastics disrupt adrenal steroid synthesis in male mice via mitochondrial dysfunction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117528. [PMID: 39674024 DOI: 10.1016/j.ecoenv.2024.117528] [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: 09/13/2024] [Revised: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Microplastics have gained significant social attention, as they can enter our bodies through food and drinking water. The adrenal gland is essential for the maintenance of metabolic homeostasis and stress responses. Nevertheless, the effects of microplastics on the steroid synthesis in the adrenal cortex was still unclear. In this study, through both in vivo and in vitro models, we found that polystyrene microplastics (PS-MPs) impaired adrenal steroid synthesis, leading to a reduction in corticosterone levels. In vivo, we further observed that chronic exposure to PS-MPs (0.25, 0.5 and 1 mg/d for 4 weeks) could induce abnormal mitochondrial morphology and functional disruptions of adrenal glands in male mice, along with an imbalance in cellular oxidative stress, manifested as increased level of reactive oxygen species, diminished antioxidant activity (glutathione peroxidase and superoxide dismutase). In vitro, these occurrences coincided with an elevated rate of cell apoptosis observed in adrenocortical cells following exposure to PS-MPs. We proposed that mitochondrial dysfunction not only directly influenced the biosynthetic processes of steroid hormones but also induced cell apoptosis through the initiation of cellular oxidative stress. The latter may represent a common mechanism underlying the multi-organ toxicity induced by PS-MPs in the body. Our findings would provide new insights for the development of more effective environmental protection measures and the reduction of plastic pollution.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Zhe Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Hanmin Xiang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yi Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yanlin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
17
|
Laugesen K, Skajaa N, Petersen I, Andersen MS, Feldt-Rasmussen U, Kejlberg Al-Mashhadi S, Stewart P, Jørgensen JOL, Sørensen HT. Mental Disorders Among Offspring Prenatally Exposed to Systemic Glucocorticoids. JAMA Netw Open 2025; 8:e2453245. [PMID: 39752154 PMCID: PMC11699534 DOI: 10.1001/jamanetworkopen.2024.53245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/01/2024] [Indexed: 01/04/2025] Open
Abstract
Importance Current evidence of the association between prenatal exposure to glucocorticoids and long-term mental disorders is scarce and has limitations. Objective To investigate the association between prenatal exposure to systemic glucocorticoids and mental disorders in offspring at the age of 15 years, comparing exposed vs unexposed offspring born to mothers with the same underlying disease (risk of preterm delivery and autoimmune or inflammatory disorders). Design, Setting, and Participants This nationwide population-based cohort study used data from registries in Denmark with follow-up until December 31, 2018. Participants included all Danish infants born alive from 1996 to 2016. Analyses were performed from January to December 2023. Exposures Prenatal exposure to systemic glucocorticoids. Main Outcomes and Measures Fifteen-year crude and adjusted risks, risk differences, and risk ratios (RR) for mental disorders using Kaplan-Meier estimator comparing exposed vs unexposed offspring born to mothers with the same underlying disease. Results A total of 1 061 548 infants (52% male) were included in the final study cohort, including 31 518 born to mothers at risk of preterm delivery and 288 747 born to mothers with autoimmune or inflammatory disorders. Among offspring born to mothers at risk of preterm delivery, the adjusted risks for exposed vs unexposed were 6.6% vs 4.3% (RR, 1.5 [95% CI, 1.2-1.9]) for autism spectrum disorders; 1.6% vs 1.3% (RR, 1.3 [95% CI, 0.8-1.8]) for intellectual disabilities; 5.8% vs 4.3% (RR, 1.3 [95% CI, 1.0-1.7]) for attention-deficit hyperactivity disorder (ADHD); and 7.2% vs 4.6% (RR, 1.5 [95% CI, 1.1-2.0]) for mood, anxiety, and stress-related disorders. Among offspring born to mothers with autoimmune or inflammatory disorders, the adjusted risks for exposed vs unexposed were 4.8% vs 3.8% (RR, 1.3 [95% CI, 1.1-1.5]) for autism spectrum disorders; 1.1% vs 0.8% (RR 1.4, [95% CI, 0.9-2.0]) for intellectual disabilities; 5.5% vs 4.4% (RR, 1.3 [95% CI, 1.0-1.5]) for ADHD; and 6.6% vs 4.6% (RR, 1.4 [95% CI, 1.2-1.8]) for mood, anxiety, and stress-related disorders. Findings were confirmed through an active comparator and sibling design. However, confounding by disease severity could not be ruled out. Conclusions and Relevance In this cohort study, prenatal exposure to glucocorticoids was associated with higher risk of some mental disorders. These data support continued caution in the use of glucocorticoids in pregnant people.
Collapse
Affiliation(s)
- Kristina Laugesen
- Department of Clinical Epidemiology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Nils Skajaa
- Department of Clinical Epidemiology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
| | - Irene Petersen
- Department of Clinical Epidemiology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
- Department of Primary Care and Population Health, University College London, London, United Kingdom
| | - Marianne Skovsager Andersen
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ulla Feldt-Rasmussen
- Department of Medical Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofie Kejlberg Al-Mashhadi
- Department of Clinical Epidemiology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
| | - Paul Stewart
- Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | | | - Henrik Toft Sørensen
- Department of Clinical Epidemiology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
| |
Collapse
|
18
|
Zoubovsky SP, Muglia LJ. Transplacental signals involved in the programming effects of prenatal psychosocial stress on neurodevelopment. Neurotoxicol Teratol 2025; 107:107424. [PMID: 39755178 DOI: 10.1016/j.ntt.2025.107424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/03/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Exposure to psychosocial stress during pregnancy has been associated with the emergence of neurodevelopmental and neuropsychiatric disorders in offspring. The placenta is known to orchestrate various functions that are essential for normal fetal development, including the brain. It has therefore been postulated that alterations in such functions, and downstream signaling, have the potential to dramatically affect brain developmental trajectories and contribute to adverse neurodevelopmental outcomes. This review will focus on discussing various placental functions that have been proposed to be affected by exposure to prenatal psychosocial stress and the implications of such disruptions on long-term neurodevelopmental programming.
Collapse
Affiliation(s)
- Sandra P Zoubovsky
- Department of Pediatrics, University of Colorado, Denver, CO, United States
| | - Louis J Muglia
- Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Office of the President, Burroughs Wellcome Fund, Research Triangle Park, Durham, NC, United States.
| |
Collapse
|
19
|
Ryu MO, Jung JY, Suh HN, Lee CY, Kim MC, Oh JY, Song WJ, Ahn C, Yang Y, Choi GE. Glucocorticoid-Dependent Retinal Degeneration and Vision Impairment in Mice Susceptible to Prenatal Stress-Induced Behavioral Abnormalities. Cell Mol Neurobiol 2024; 45:7. [PMID: 39714626 DOI: 10.1007/s10571-024-01525-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024]
Abstract
Chronic exposure to prenatal stress can impair neurogenesis and lead to irreversible cognitive and neuropsychiatric abnormalities in offspring. The retina is part of the nervous system; however, the impacts of prenatal stress on retinal neurogenesis and visual function remain unclear. This study examined how elevated prenatal glucocorticoid levels differentially affect retinal development in the offspring of pregnant mice exposed to chronic unpredictable mild stress (CUMS). Offspring were classified into control, stress-resilient, and stress-susceptible groups based on behavioral tests assessing spatial memory and depression-like behaviors. The stress-susceptible group exhibited significantly altered synaptogenesis, reduced ganglion cell development, decreased retinal thickness, and visiual impairment. These mice also showed a pervasive transformation of retinal astrocytes into a proinflammatory A1-like reactive state, evidenced by increased GFAP and decreased STAT3 expression levels. This astrocyte phenotype shift coincided with disruptions in neurogenesis and synaptic formation. Furthermore, prenatal exposure to exogenous corticosterone confirmed that the effects of prenatal stress are mediated by glucocorticoid-induced retinal neurodegeneration. Our findings suggest that elevated prenatal glucocorticoid levels trigger a series of neurodevelopmental disturbances leading to retinal neurodegeneration and vision impairment. This research highlights the impact of prenatal stress on retinal development and visual health, suggesting new avenues for understanding and potentially mitigating the negative effects of early-life stress on neurodevelopment.
Collapse
Affiliation(s)
- Min-Ok Ryu
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Ji-Youl Jung
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Korea
| | - Han Na Suh
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongup, Jeonbukdo, 56212, Republic of Korea
| | - Chung-Young Lee
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Untreatable Infectious Disease Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Myung-Chul Kim
- Veterinary Laboratory Medicine, Clinical Pathology, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
- Research Institute of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Ji Young Oh
- Division of Intractable Disease Research, Korea Institute of Health, Osong, Cheongju, 28106, Korea
| | - Woo-Jin Song
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Korea
| | - Changhwan Ahn
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yeseul Yang
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Gee Euhn Choi
- Laboratory of Veterinary Biochemistry, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, South Korea.
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, South Korea.
| |
Collapse
|
20
|
Castillo-Ramírez LA, Herget U, Ryu S, De Marco RJ. Early-life challenge enhances cortisol regulation in zebrafish larvae. Biol Open 2024; 13:bio061684. [PMID: 39607018 PMCID: PMC11625891 DOI: 10.1242/bio.061684] [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: 08/12/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis in mammals and the hypothalamic-pituitary-interrenal (HPI) axis in fish are open systems that adapt to the environment during development. Little is known about how this adaptation begins and regulates early stress responses. We used larval zebrafish to examine the impact of prolonged forced swimming at 5 days post-fertilization (dpf), termed early-life challenge (ELC), on cortisol responses, neuropeptide expression in the nucleus preopticus (NPO), and gene transcript levels. At 6 dpf, ELC-exposed larvae showed normal baseline cortisol but reduced reactivity to an initial stressor. Conversely, they showed increased reactivity to a second stressor within the 30-min refractory period, when cortisol responses are typically suppressed. ELC larvae had fewer corticotropin-releasing hormone (crh), arginine vasopressin (avp), and oxytocin (oxt)-positive cells in the NPO, with reduced crh and avp co-expression. Gene expression analysis revealed upregulation of genes related to cortisol metabolism (hsd11b2, cyp11c1), steroidogenesis (star), and stress modulation (crh, avp, oxt). These results suggest that early environmental challenge initiates adaptive plasticity in the HPI axis, tuning cortisol regulation to balance responsiveness and protection during repeated stress. Future studies should explore the broader physiological effects of prolonged forced swimming and its long-term impact on cortisol regulation and stress-related circuits.
Collapse
Affiliation(s)
- Luis A. Castillo-Ramírez
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
| | - Ulrich Herget
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, California 91125, USA
| | - Soojin Ryu
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road EX4 4QD, Exeter, UK
| | - Rodrigo J. De Marco
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- School of Biological and Environmental Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, L3 3AF Liverpool, UK
| |
Collapse
|
21
|
Zha X, Fang M, Zhong W, Chen L, Feng H, Zhang M, Wang H, Zhang Y. Dose-, stage- and sex- difference of prenatal prednisone exposure on placental morphological and functional development. Toxicol Lett 2024; 402:68-80. [PMID: 39580039 DOI: 10.1016/j.toxlet.2024.11.005] [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: 03/24/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/25/2024]
Abstract
Prednisone, a synthetic glucocorticoid, is commonly used to treat autoimmune diseases in pregnant women. However, some studies suggest that the use of prednisone during pregnancy may lead to adverse pregnancy outcomes. In this study, we established PPE mouse models at different doses (0.25, 0.5, 1.0 mg/kg·d) and different stages (whole pregnancy, early pregnancy and middle-late pregnancy) and determined outcomes on the placenta and fetus. The results of our study indicated that at the highest dose of 1 mg/kg PPE using a GD 0-18 dosing regime, PPE caused placental morphological changes measured as a decrease in placental weight relative to controls and a decrease in the placenta junctional zone (JZ)/labyrinth zone (LZ) ratio. No changes were observed on the fetuses for number of live, stillborn, and absorbed fetuses between the experimental groups and the control group. In the placentas at some doses, there were decreases in cell proliferation markers measured at the RNA and protein level by Western blot and increased apoptosis. Measures of gene expression at the mRNA level showed altered nutrients (including glucose, amino acid, and cholesterol) transport gene expressions with the most significant change associated with the male placentas at high-dose and whole pregnancy PPE group. It was further found that PPE led to the inhibition of the insulin-like growth factor 2 (IGF2)/insulin-like growth factor 1 receptor (IGF1R) signaling pathway, which was well correlated with the indicators of cell proliferation, syncytialization and nutrient (glucose and amino acid) transport indices. In conclusion, PPE can alter placental morphology and nutrient transport function, with differences in effect related to dose, stage and gender. Differential gene expressions measured for genes of the IGF2/IGF1R signaling pathway suggested this pathway may be involved in the effects seen with PPE. This study provides a theoretical and experimental basis for enhancing the understanding of the effects of prednisone use on placenta during human pregnancy but does not currently raise concerns for human use as effects were not seen on the fetuses and while the effects on cell proliferation are informative they were inconsistent and the differential effects on female and male placentas unexplained suggesting that further work is required to elucidate if these findings have relevance for human use of PPE during pregnancy.
Collapse
Affiliation(s)
- Xiaomeng Zha
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Man Fang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wen Zhong
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liang Chen
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui Feng
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Min Zhang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China; Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan 430071, China
| | - Hui Wang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China.
| | - Yuanzhen Zhang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China; Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan 430071, China.
| |
Collapse
|
22
|
Griffith T, White-Traut R, Tell D, Green SJ, Janusek L. Epigenetics Embedding of Oral Feeding Skill Development in Preterm Infants: A Study Protocol. Adv Neonatal Care 2024; 24:E88-E95. [PMID: 39602096 PMCID: PMC11708242 DOI: 10.1097/anc.0000000000001216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
BACKGROUND Preterm infants face challenges to feed orally, which may lead to failure to thrive. Oral feeding skill development requires intact neurobehaviors. Early life stress results in DNA methylation of NR3C1 and HSD11B2, which may disrupt neurobehaviors. Yet, the extent to which early life stress impairs oral feeding skill development and the biomechanism whereby this occurs remains unknown. Our team is conducting an NIH funded study (K23NR019847, 2022-2024) to address this knowledge gap. PURPOSE To describe an ongoing study protocol to determine the extent to which early life stress, reflected by DNA methylation of NR3C1 and HSD11B2 promoter regions, compromises oral feeding skill development. METHODS This protocol employs a longitudinal prospective cohort study. Preterm infants born between 26 and 34 weeks gestational age have been enrolled. We evaluate early life stress, DNA methylation, cortisol reactivity, neurobehaviors, and oral feeding skill development during neonatal intensive care unit hospitalization and at 2-week post-discharge. RESULTS To date, we have enrolled 70 infants. We have completed the data collection. Currently, we are in the data analysis phase of the study, and expect to disseminate the findings in 2025. IMPLICATIONS FOR PRACTICE AND RESEARCH The findings from this study will serve as a foundation for future clinical and scientific inquiries that support oral feeding and nutrition, reduce post-discharge feeding difficulties and lifelong risk of maladaptive feeding behaviors and poor health outcomes. Findings from this study will also provide further support for the implementation of interventions to minimize stress in the vulnerable preterm infant population.
Collapse
Affiliation(s)
- Thao Griffith
- Author Affiliations: Department of Family and Community Health Nursing, Marcella Niehoff School of Nursing, Loyola University Chicago, Maywood, Illinois (Drs Griffith, Tell, and Janusek); Nursing Research and Evidence-Based Practice, Children's Wisconsin, Milwaukee, Wisconsin (Dr White-Traut); Women, Children and Family Health Science, College of Nursing, University of Illinois at Chicago, Chicago, Illinois (Dr White-Traut); and Department of Internal Medicine, Division of Infectious Disease, Rush University, Chicago, Illinois (Dr Green)
| | | | | | | | | |
Collapse
|
23
|
Oh M, Yoon NH, Kim SA, Yoo HJ. Epigenetic Insights into Autism Spectrum Disorder: DNA Methylation Levels of NR3C1, ASCL1, and FOXO3 in Korean Autism Spectrum Disorder Sibling Pairs. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:635-645. [PMID: 39420610 PMCID: PMC11494426 DOI: 10.9758/cpn.24.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 10/19/2024]
Abstract
Objective Previous research on autism spectrum disorder (ASD) in Koreans has primarily focused on genetic diversity because of its high heritability. However, the emerging recognition of transgenerational epigenetic changes has recently shifted research attention towards epigenetic perspectives. Methods This study investigated the DNA methylation patterns of the promoter regions of candidate genes such as NR3C1, ASCL1, and FOXO3 in blood samples from ASD probands and their unaffected siblings. The analysis included 54 families (ASD proband group: 54; unaffected biological sibling group: 63). The diagnostic process involved screening the probands and their siblings for ASD based on the Diagnostic and Statistical Manual of Mental Disorders 5th edition. Intelligence, social ability, and medical history were thoroughly assessed using various scales and questionnaires. Genomic DNA from blood samples was analyzed using a methylation-sensitive quantitative polymerase chain reaction to examine the DNA methylation status of candidate genes. Results Methylation levels in candidate gene promoter regions differed significantly between the proband and sibling groups for all candidate genes. Correlation analysis between the proband and sibling groups revealed strong and significant correlations in NR3C1 and ASCL1 methylation. Additionally, in the analysis of the relationship between DNA and ASD phenotypes, FOXO3 methylation correlated with social quotient in probands, and ASCL1 methylation was associated with nonverbal communication, and daily living skills as measured by the Korean Vineland Adaptive Behavior Scale. Notably, ASCL1 methylation was significantly associated with parental age at pregnancy. Conclusion This study proposes DNA methylation of NR3C1, ASCL1, and FOXO3 in peripheral blood samples is a potential epigenetic biomarker of ASD.
Collapse
Affiliation(s)
- Miae Oh
- Department of Psychiatry, Kyung Hee University Hospital, Seoul, Korea
| | - Nan-He Yoon
- Division of Social Welfare and Health Administration, Wonkwang University, Iksan, Korea
| | - Soon Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejon, Korea
| | - Hee Jeong Yoo
- Department of Psychiatry, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
24
|
Chen X, Liu H, Zhou A, Jin F, Jing C, Li Y, Xia W, Kahn LG, Xie Y, Xiang X, Cao S, Zhang W, Mahai G, Cao Z, Xiao H, Xiong C, Li W, Li H, Xu S. Fetal weight growth trajectories and childhood development: A population-based cohort study. Sci Bull (Beijing) 2024; 69:3404-3414. [PMID: 39261129 DOI: 10.1016/j.scib.2024.04.077] [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: 10/07/2023] [Revised: 04/07/2024] [Accepted: 04/07/2024] [Indexed: 09/13/2024]
Abstract
This study aimed to investigate whether fetal growth trajectories (FGTs) could predict early childhood development, indicate intrauterine metabolic changes, and explore potential optimal and suboptimal FGTs. FGTs were developed by using an unsupervised machine-learning approach. Children's neurodevelopment, anthropometry, and respiratory outcomes in the first 6 years of life were assessed at different ages. In a subgroup of participants, we conducted a metabolomics analysis of cord blood to reveal the metabolic features of FGTs. We identified 6 FGTs: early decelerating, early decelerating with late catch-up growth, early accelerating, early accelerating with late medium growth, late decelerating, and late accelerating. The early accelerating with late medium growth pattern might be the optimal FGT due to its associations with better psychomotor development, mental development, intelligence quotient, and lung function and a lower risk of behaviour and respiratory problems. Compared with the optimal FGT, early decelerating and late decelerating FGTs were associated with poor neurodevelopment and lung function, while early accelerating FGT was associated with more severe autistic symptoms, poor lung function, and increased risks of overweight/obesity. Metabolic alterations were enriched in amino acid metabolism for early decelerating and late decelerating FGTs, whereas altered metabolites were enriched in lipid metabolism for early accelerating FGT. These findings suggest that FGTs are predictors of early life development and may indicate intrauterine adaptive metabolism. The discovery of optimal and suboptimal FGTs provides potential clues for the early identification and intervention of fetal origin dysplasia or disease, but further research on related mechanisms is still needed.
Collapse
Affiliation(s)
- Xinmei Chen
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongxiu Liu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Aifen Zhou
- Wuhan Medical & Healthcare Center for Women and Children, Wuhan 430015, China
| | - Feng Jin
- Shunyi Women's and Children's Hospital of Beijing Children's Hospital, Beijing 101320, China
| | - Chufeng Jing
- Wuxi Maternal and Child Health Hospital, Wuxi 214001, China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Linda G Kahn
- Department of Pediatrics, New York University Grossman School of Medicine, New York, 10016, USA
| | - Ya Xie
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xingliang Xiang
- School of Environmental Science and Engineering, Hainan University, Haikou 570208, China
| | - Shuting Cao
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenxin Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gaga Mahai
- School of Environmental Science and Engineering, Hainan University, Haikou 570208, China
| | - Zhongqiang Cao
- Wuhan Medical & Healthcare Center for Women and Children, Wuhan 430015, China
| | - Han Xiao
- Wuhan Medical & Healthcare Center for Women and Children, Wuhan 430015, China
| | - Chao Xiong
- Wuhan Medical & Healthcare Center for Women and Children, Wuhan 430015, China
| | - Wei Li
- Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | - Hanzeng Li
- School of Environmental Science and Engineering, Hainan University, Haikou 570208, China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Environmental Science and Engineering, Hainan University, Haikou 570208, China.
| |
Collapse
|
25
|
Viola MF, Franco Taveras E, Mass E. Developmental programming of tissue-resident macrophages. Front Immunol 2024; 15:1475369. [PMID: 39575254 PMCID: PMC11578957 DOI: 10.3389/fimmu.2024.1475369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Macrophages are integral components of the innate immune system that colonize organs early in development and persist into adulthood through self-renewal. Their fate, whether they are replaced by monocytes or retain their embryonic origin, depends on tissue type and integrity. Macrophages are influenced by their environment, a phenomenon referred to as developmental programming. This influence extends beyond the local tissue microenvironment and includes soluble factors that can reach the macrophage niche. These factors include metabolites, antibodies, growth factors, and cytokines, which may originate from maternal diet, lifestyle, infections, or other developmental triggers and perturbations. These influences can alter macrophage transcriptional, epigenetic, and metabolic profiles, affecting cell-cell communication and tissue integrity. In addition to their crucial role in tissue immunity, macrophages play vital roles in tissue development and homeostasis. Consequently, developmental programming of these long-lived cells can modulate tissue physiology and pathology throughout life. In this review, we discuss the ontogeny of macrophages, the necessity of developmental programming by the niche for macrophage identity and function, and how developmental perturbations can affect the programming of macrophages and their subtissular niches, thereby influencing disease onset and progression in adulthood. Understanding these effects can inform targeted interventions or preventive strategies against diseases. Finally, understanding the consequences of developmental programming will shed light on how maternal health and disease may impact the well-being of future generations.
Collapse
Affiliation(s)
| | | | - Elvira Mass
- Developmental Biology of the Immune System, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
26
|
Ünsel-Bolat G, Yıldırım S, Kılıçaslan F, Caparros-Gonzalez RA. Natural Disasters as a Maternal Prenatal Stressor and Children's Neurodevelopment: A Systematic Review. Behav Sci (Basel) 2024; 14:1054. [PMID: 39594354 PMCID: PMC11590888 DOI: 10.3390/bs14111054] [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: 10/21/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The intrauterine period is a time of high sensitivity in the development of the embryo and the fetus. Therefore, low levels of maternal stress are closely associated with healthy brain development in the neonatal and early childhood periods. There is increasing evidence linking natural disasters as prenatal maternal stress (PNMS) to neurodevelopmental disorders (including subclinical manifestations). Natural disasters involve many factors in addition to the trauma they cause, including loss and the physical and psychosocial difficulties that result from that trauma. This review article aims to bring together research findings on the neurodevelopmental effects of natural disasters on children as PNMS. It also looks at how factors such as gestational age and gender contribute to these effects. We conducted a systematic review on PubMed, Web of Science, and Scopus, with 30 studies meting the inclusion criteria. This systematic review was conducted in accordance with the PRISMA guidelines. A total of 1,327,886 mother-child dyads participated in the included studies. The results of the studies indicate that natural disasters have a negative impact on children's outcomes in terms of cognitive development, language development, autism/autism-like features, motor skills, performance in mathematics, mental development, sleep, attention, behavioral and emotional problems, and various psychiatric comorbidities.
Collapse
Affiliation(s)
- Gül Ünsel-Bolat
- Department of Child and Adolescent Psychiatry, Balıkesir University, 10145 Balıkesir, Turkey;
| | - Sema Yıldırım
- Department of Child and Adolescent Psychiatry, Balıkesir University, 10145 Balıkesir, Turkey;
| | - Fethiye Kılıçaslan
- Department of Child and Adolescent Psychiatry, Harran University, 63300 Şanlıurfa, Turkey;
| | - Rafael A. Caparros-Gonzalez
- Faculty of Health Sciences, University of Granada, 18071 Granada, Spain;
- Instituto de Investigacion Biosanitaria ibs.GRANADA, 18071 Granada, Spain
| |
Collapse
|
27
|
Fuma K, Ushida T, Kawaguchi M, Nosaka R, Kidokoro H, Tano S, Imai K, Sato Y, Hayakawa M, Kajiyama H, Kotani T. Impact of antenatal corticosteroids on subcortical volumes in preterm infants at term-equivalent age: A retrospective observational study. Eur J Obstet Gynecol Reprod Biol 2024; 302:7-14. [PMID: 39208714 DOI: 10.1016/j.ejogrb.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Antenatal corticosteroids (ACS) is a well-established treatment for women at risk of preterm birth that improves neonatal outcomes. However, several concerns have been raised regarding the potential long-term adverse effects of ACS on the offspring's developing brain. Here we investigated the association between ACS and subcortical segmental volumes in preterm infants at term-equivalent age. STUDY DESIGN This retrospective observational study was conducted using the clinical data of preterm singleton infants born between 220/7 and 336/7 gestational weeks at Nagoya University Hospital in 2014-2020. Subcortical volumes of the bilateral thalami, caudate nuclei, putamens, pallidums, hippocampi, amygdalae, and nuclei accumbens were evaluated using an automated segmentation tool, Infant FreeSurfer, and compared between neonates exposed to a single course of ACS (n = 46) and those who were not (n = 13) by multiple linear regression analysis (covariates: postmenstrual age at magnetic resonance imaging, infant sex, and gestational age at birth). We compared each subcortical volume stratified by gestational age at birth (<28 vs. ≥28 gestational weeks). RESULTS Multivariate analyses revealed significantly smaller volumes in the bilateral amygdalae (left, p < 0.03; right, p < 0.03) and caudate nuclei (left, p < 0.03; right, p = 0.04) in neonates with ACS. Significantly smaller volumes in these regions were observed only in neonates born at 28 weeks of gestation or later. CONCLUSIONS ACS was associated with smaller volumes of the bilateral amygdalae and caudate nuclei at term-equivalent age. This association was observed exclusively in infants born at 28 weeks of gestation or later.
Collapse
Affiliation(s)
- Kazuya Fuma
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Division of Reproduction and Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.
| | - Masahiro Kawaguchi
- Department of Neurology, Aichi Children's Health and Medical Center, Obu, Japan; Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rena Nosaka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Division of Reproduction and Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
28
|
Baud O, Sentilhes L, Ursino M, Doret-Dion M, Alberti C, Aupiais C, Schmitz T. Survival without severe neonatal morbidity after antenatal betamethasone dose reduction: a post hoc analysis of a randomized non-inferiority trial. Am J Obstet Gynecol 2024; 231:458.e1-458.e16. [PMID: 38341166 DOI: 10.1016/j.ajog.2024.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/29/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Antenatal betamethasone is recommended before preterm delivery to accelerate fetal lung maturation. However, its optimal dose remains unknown. A 50% dose reduction was proposed to decrease the potential dose-related long-term neurodevelopmental side effects, including psychological development, sleep, and emotional disorders. Because noninferiority of the half dose in terms of the need for exogenous surfactant was not shown in the primary analysis, its impact on survival without major neonatal morbidity needs to be investigated. OBJECTIVE This study aimed to investigate the impact of antenatal betamethasone dose reduction on survival of very preterm infants without severe neonatal morbidity, a factor known to have a strong correlation with long-term outcomes. STUDY DESIGN We performed a post hoc secondary analysis of a randomized, multicenter, double-blind, placebo-controlled, noninferiority trial, testing half (11.4 mg once; n=1620) vs full (11.4 mg twice, 24 hours apart; n=1624) antenatal betamethasone doses in women at risk of preterm delivery. To measure survival without severe neonatal morbidity at hospital discharge among neonates born before 32 weeks of gestation, we used the definition of the French national prospective study on preterm children, EPIPAGE 2, comprising 1 of the following morbidities: grade 3 to 4 intraventricular hemorrhage, cystic periventricular leukomalacia, necrotizing enterocolitis stage ≥2, retinopathy of prematurity requiring anti-vascular endothelial growth factor therapy or laser, and moderate-to-severe bronchopulmonary dysplasia. RESULTS After exclusion of women who withdrew consent or had pregnancy termination and of participants lost to follow-up (8 in the half-dose and 10 in the full-dose group), the rate of survival without severe neonatal morbidity among neonates born before 32 weeks of gestation was 300 of 451 (66.5%) and 304 of 462 (65.8%) in the half-dose and full-dose group, respectively (risk difference, +0.7%; 95% confidence interval, -5.6 to +7.1). There were no significant between-group differences in the cumulative number of neonatal morbidities. Results were similar when using 2 other internationally recognized definitions of severe neonatal morbidity and when considering the overall population recruited in the trial. CONCLUSION In the BETADOSE trial, severe morbidity at discharge of newborns delivered before 32 weeks of gestation was found to be similar among those exposed to 11.4-mg and 22.8-mg antenatal betamethasone. Additional studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Olivier Baud
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva, University of Geneva, Geneva, Switzerland; Université Paris Cité, Inserm U1141, Paris, France.
| | - Loic Sentilhes
- Department of Obstetrics and Gynecology, Bordeaux University Hospital, Bordeaux, France
| | - Moreno Ursino
- Clinical Epidemiology Unit, CIC-EC 1426, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Centre de Recherche des Cordeliers, Inserm U1138, Université Paris Cité, Sorbonne Université, Paris, France; HeKA, Inria Paris, Paris, France
| | - Muriel Doret-Dion
- Department of Obstetrics and Gynecology, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Corinne Alberti
- Clinical Epidemiology Unit, CIC-EC 1426, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, Inserm U1123, ECEVE, Paris, France
| | - Camille Aupiais
- Université Paris Cité, Inserm U1123, ECEVE, Paris, France; Pediatric Emergency Department, Jean Verdier Hospital, Assistance Publique-Hôpitaux de Paris, Sorbonne Paris Nord University, Paris, France
| | - Thomas Schmitz
- Department of Obstetrics and Gynecology, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, Centre for Research in Epidemiology and Statistics (CRESS), Inserm, INRAE, Paris, France
| |
Collapse
|
29
|
Baptista FI, Ambrósio AF. Tracing the influence of prenatal risk factors on the offspring retina: Focus on development and putative long-term consequences. Eur J Clin Invest 2024; 54:e14266. [PMID: 38864773 DOI: 10.1111/eci.14266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Pregnancy represents a window of vulnerability to fetal development. Disruptions in the prenatal environment during this crucial period can increase the risk of the offspring developing diseases over the course of their lifetime. The central nervous system (CNS) has been shown to be particularly susceptible to changes during crucial developmental windows. To date, research focused on disruptions in the development of the CNS has predominantly centred on the brain, revealing a correlation between exposure to prenatal risk factors and the onset of neuropsychiatric disorders. Nevertheless, some studies indicate that the retina, which is part of the CNS, is also vulnerable to in utero alterations during pregnancy. Such changes may affect neuronal, glial and vascular components of the retina, compromising retinal structure and function and possibly impairing visual function. METHODS A search in the PubMed database was performed, and any literature concerning prenatal risk factors (drugs, diabetes, unbalanced diet, infection, glucocorticoids) affecting the offspring retina were included. RESULTS This review collects evidence on the cellular, structural and functional changes occurring in the retina triggered by maternal risk factors during pregnancy. We highlight the adverse impact on retinal development and its long-lasting effects, providing a critical analysis of the current knowledge while underlining areas for future research. CONCLUSIONS Appropriate recognition of the prenatal risk factors that negatively impact the developing retina may provide critical clues for the design of preventive strategies and for early therapeutic intervention that could change retinal pathology in the progeny.
Collapse
Affiliation(s)
- Filipa I Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - António F Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
| |
Collapse
|
30
|
Specht L, Freiberg A, Mojahed A, Garthus-Niegel S, Schellong J. Adrenocortical deviations and adverse clinical outcomes in children and adolescents exposed to interparental intimate partner violence: A systematic review. Neurosci Biobehav Rev 2024; 165:105866. [PMID: 39233285 DOI: 10.1016/j.neubiorev.2024.105866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Childhood exposure to interparental intimate partner violence (i-IPV) is a pervasive form of child maltreatment, posing major public health concerns and elevating risks for enduring adverse clinical and developmental consequences. However, assessing the full spectrum of clinical effects is challenging, potentially leading to inconsistent identification of children in need of early intervention. This systematic review aimed to identify hypothalamic-pituitary-adrenocortical axis dysfunction following i-IPV exposure, elucidating the underlying biopsychobehavioural mechanisms and predicting adverse outcomes. We searched Embase, MEDLINE, and PsycINFO for peer-reviewed studies from infancy through adolescence, screened reference lists and conducted forward searches. Analysis of 23 publications (N = 1848) revealed associations between i-IPV and altered adrenocortical function from early childhood, influenced by FKBP5 haplotype, parental caregiving and offspring emotional insecurity. Results showed that the adrenocortical stress response may predict internalising and externalising problems, childhood asthma, impaired executive function and poor academic performance. Nonetheless, inconsistencies in findings between studies suggest methodological heterogeneity and potential bias. Identifying biomarkers such as cortisol can enhance prediction and mechanism-based intervention efforts but long-term studies with a common theoretical and methodological framework are needed for comprehensive understanding. Integrating biological, emotional, and behavioural assessments could potentiate trauma services and research, ultimately improving outcomes for affected children.
Collapse
Affiliation(s)
- Lina Specht
- Department of Psychotherapy and Psychosomatic Medicine, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany; Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany.
| | - Alice Freiberg
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany
| | - Amera Mojahed
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany
| | - Susan Garthus-Niegel
- Department of Psychotherapy and Psychosomatic Medicine, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany; Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany; Institute for Systems Medicine (ISM) and Faculty of Medicine, Medical School Hamburg, Am Kaiserkai 1, Hamburg 20457, Germany; Department of Childhood and Families, Norwegian Institute of Public Health, Postboks 222 Skøyen, Oslo 0213, Norway
| | - Julia Schellong
- Department of Psychotherapy and Psychosomatic Medicine, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany
| |
Collapse
|
31
|
Heigwer J, Steenbergen PJ, Gehrig J, Westhoff JH. Corticosteroids alter kidney development and increase glomerular filtration rate in larval zebrafish (Danio rerio). Toxicol Sci 2024; 201:216-225. [PMID: 38964340 DOI: 10.1093/toxsci/kfae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024] Open
Abstract
Pharmaceutical drugs and other chemicals can impact organogenesis, either during pregnancy or by postnatal exposure of very preterm infants. Corticosteroids are administered to pregnant women at risk of preterm delivery in order to reduce neonatal morbidity and mortality. In addition, high-dose corticosteroid exposure of very preterm infants regularly serves to maintain blood pressure and to prevent and treat bronchopulmonary dysplasia, a form of chronic lung disease in prematurely born infants. Despite clinical benefits, there is increasing evidence of corticosteroid-mediated short- and long-term detrimental developmental effects, especially in the kidney. Here, we performed a detailed morphological and functional analysis of corticosteroid-mediated effects on pronephros development in larval zebrafish. About 24-h postfertilization (hpf) transgenic Tg(wt1b: EGFP) zebrafish larvae were exposed to a set of natural and synthetic corticosteroids (hydrocortisone, dexamethasone, 6α-methylprednisolone, betamethasone, prednisolone, fludrocortisone, 11-deoxycorticosterone) with varying glucocorticoid and mineralocorticoid potency for 24 h at different concentrations. A semiautomated, multiparametric in vivo workflow enabled simultaneous assessment of kidney morphology, renal FITC-inulin clearance, and heart rate within the same larva. All corticosteroids exerted significant morphological and functional effects on pronephros development, including a significant hypertrophy of the pronephric glomeruli as well as dose-dependent increases in FITC-inulin clearance as a marker of glomerular filtration rate. In conclusion, the present study demonstrates a significant impact of corticosteroid exposure on kidney development and function in larval zebrafish. Hence, these studies underline that corticosteroid exposure of the fetus and the preterm neonate should be carefully considered due to potential short- and long-term harm to the kidney.
Collapse
Affiliation(s)
- Jana Heigwer
- Department I, Center for Pediatric and Adolescent Medicine, Medical Faculty Heidelberg, Heidelberg University, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Petrus J Steenbergen
- Department I, Center for Pediatric and Adolescent Medicine, Medical Faculty Heidelberg, Heidelberg University, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Jochen Gehrig
- ACQUIFER Imaging GmbH, Heidelberg 69126, Germany
- Luxendo GmbH, Fluorescence Microscopy Business Unit, Heidelberg 69126, Germany
| | - Jens H Westhoff
- Department I, Center for Pediatric and Adolescent Medicine, Medical Faculty Heidelberg, Heidelberg University, University Hospital Heidelberg, Heidelberg 69120, Germany
| |
Collapse
|
32
|
Ishikawa A, Takanuma T, Hashimoto N, Tsudzuki M. Association between Temperament and Stress-related Gene Expression in Day-old Chickens. J Poult Sci 2024; 61:2024022. [PMID: 39130209 PMCID: PMC11310665 DOI: 10.2141/jpsa.2024022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024] Open
Abstract
Stress in day-old chickens from commercial hatcheries is associated with problematic behavior in adult animals. Recently, we developed a new behavioral handling test for day-old chickens and demonstrated that it assessed temperament differences between seven breeds of native Japanese and Western chickens. In this study, we used 2-day-old male chicks from five of the above breeds to investigate the relationship between temperament and mRNA levels of three stress-related genes (nuclear receptor subfamily 3 group C member 1 (NR3C1), cytochrome P450 family 11 subfamily A member 1, and hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1) involved in the hypothalamic-pituitary-adrenal axis. Principal component analysis of 10 behavioral traits for the handling test revealed that the Fayoumi breed and Hiroshima line of the Chabo breed, both of which exhibited boisterous temperament, clustered separately from the other breeds. Only NR3C1 expression showed a significant positive correlation with two behavioral traits (general vocalization and approaching the wall), and a negative correlation with movement. These results suggest that the complex temperament of day-old chickens is regulated, in part, by stress-related genes along the hypothalamic-pituitary-adrenal axis.
Collapse
Affiliation(s)
- Akira Ishikawa
- Graduate School of Bioagricultural Sciences, Nagoya
University, Nagoya 464-8601, Japan
| | - Tomoka Takanuma
- Graduate School of Bioagricultural Sciences, Nagoya
University, Nagoya 464-8601, Japan
| | - Norikazu Hashimoto
- Livestock Experiment Station, Wakayama Prefecture,
Hidaka-Gun 644-1111, Japan
| | - Masaoki Tsudzuki
- Graduate School of Integrated Sciences for Life, Hiroshima
University, Higashi-Hiroshima 739-8525, Japan
| |
Collapse
|
33
|
Yael A, Fishman R, Matas D, Doniger T, Vortman Y, Koren L. Fetal endocrine axes mRNA expression levels are related to sex and intrauterine position. Biol Sex Differ 2024; 15:61. [PMID: 39103957 DOI: 10.1186/s13293-024-00637-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND The hypothalamic-pituitary-adrenal (HPA) and -gonadal (HPG) axes are two major pathways that connect the neural and endocrine systems in vertebrates. Factors such as prenatal stress and maternal exposure to exogenous steroids have been shown to affect these pathways during fetal development. Another less studied factor is the transfer of hormones across fetuses in multifetal pregnancies. This form of transfer has been shown to influence the morphology, anatomy, physiology, and behavior of the offspring in litter-bearing mammals, an influence termed the intrauterine position (IUP) effect. In this study, we sought to delineate how the IUP effects HPA and HPG brain receptors, peptides, and enzymes (hereafter components) in utero and how these influences may differ between males and females. METHODS We utilized the unconventional model of culled free-ranging nutria (Myocastor coypus), with its large natural variation. We collected brain tissues from nutria fetuses and quantified the expression of key HPA and HPG components in three brain regions: prefrontal cortex, hypothalamus, and striatum. RESULTS We found an interaction between sex and IUP in the mineralocorticoid receptor (MR), gonadotropin-releasing hormone receptor (GNRHR), androgen receptor (AR), and estrogen receptor alpha (ESR1). IUP was significant in both gonadotropin-releasing hormone (GnRH) and its receptor GNRHR, but in different ways. In the hypothalamus, fetuses adjacent to same-sex neighbors had higher expression of GnRH than fetuses neighboring the opposite sex. Conversely, in the cortex, GNRHR exhibited the inverse pattern, and fetuses that were neighboring the opposite sex had higher expression levels than those neighboring the same sex. Regardless of IUP, in most components that showed significant sex differences, female fetuses had higher mRNA expression levels than male fetuses. We also found that HPA and HPG components were highly related in the early stages of gestation, and that there was an interaction between sex and developmental stage. In the early stages of pregnancy, female component expression levels were more correlated than males', but in the last trimester of pregnancy, male components were more related to each other than female's. CONCLUSIONS This study suggests that there are sexually different mechanisms to regulate the HPA and HPG axes during fetal development. Higher mRNA expression levels of endocrine axes components may be a mechanism to help females cope with prolonged androgen exposure over a long gestational period. Additionally, these findings suggest different coordination requirements of male and female endocrine axes during stages of fetal development.
Collapse
Affiliation(s)
- Ariel Yael
- The Faculty of Life Sciences, Bar-Ilan University, 5290002, Ramat Gan, Israel
| | - Ruth Fishman
- The Faculty of Life Sciences, Bar-Ilan University, 5290002, Ramat Gan, Israel
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Devorah Matas
- The Faculty of Life Sciences, Bar-Ilan University, 5290002, Ramat Gan, Israel
| | - Tirza Doniger
- The Faculty of Life Sciences, Bar-Ilan University, 5290002, Ramat Gan, Israel
| | - Yoni Vortman
- Department of Animal Sciences, Hula Research Center, Tel Hai Academic College, Upper Galilee, 1220800, Qiryat Shemona, Israel
| | - Lee Koren
- The Faculty of Life Sciences, Bar-Ilan University, 5290002, Ramat Gan, Israel.
| |
Collapse
|
34
|
Tain YL, Li LC, Kuo HC, Hsu CN. Gestational Exposure to Maternal Systemic Glucocorticoids and Childhood Risk of CKD. Am J Kidney Dis 2024; 84:215-223.e1. [PMID: 38479460 DOI: 10.1053/j.ajkd.2024.01.523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 07/23/2024]
Abstract
RATIONALE & OBJECTIVE The potential effects of antenatal glucocorticoid exposure on the health of children are unclear. We examined the association of gestational exposure to maternal systemic glucocorticoids and the risk of developing chronic kidney disease (CKD) in childhood. STUDY DESIGN Retrospective cohort study. SETTING & PARTICIPANTS Newborns cared for at the largest health care delivery system in Taiwan between 2004 and 2018. EXPOSURE Maternal prescriptions for systemic glucocorticoids between the last menstrual period and birth as a proxy for gestational exposure. OUTCOME Incidence of childhood CKD, including congenital anomalies of the kidney and urinary tract (CAKUT) and other kidney diseases (non-CAKUT), over 10 years. ANALYTICAL APPROACH Cox proportional hazards models with stabilized inverse probability of treatment weighting and robust sandwich estimator were used to estimate the average association between systemic glucocorticoids and incident CKD after adjustment for offspring characteristics (adjusted HR: AHR). RESULTS Among 23,363 singleton-born children, gestational systemic glucocorticoid exposure was significantly associated with a higher risk of childhood CKD (AHR, 1.69 [95% CI, 1.01-2.84]). Stratified analyses showed stronger associations between systemic glucocorticoids and childhood CKD within the strata of birth<37 weeks' gestational age (AHR, 2.38 [95% CI, 1.19-4.78]), male sex (AHR, 1.89 [95% CI, 1.00-3.55]), gestational exposure in the second trimester (AHR, 6.70 [95% CI, 2.17-20.64]), and total dose of>24mg hydrocortisone equivalent (AHR, 1.91 [95% CI, 1.05-3.47]). LIMITATIONS Study was limited to the Taiwan health care delivery system and childhood CKD events through the age of 10 years. CONCLUSIONS The findings of this study suggest that gestational exposure to systemic glucocorticoids is associated with the occurrence of kidney disease in childhood. If these findings are confirmed, they may inform clinicians who are considering prescribing systemic glucocorticoids during pregnancy. PLAIN-LANGUAGE SUMMARY In a singleton-born cohort of neonates, maternal exposure to antenatal systemic glucocorticoids was significantly associated with a 1.7-fold increased risk of the children developing chronic kidney disease over the first 10 years of life. Children of mothers who received>24mg of hydrocortisone equivalent, systemic glucocorticoid treatment in second trimester of gestation, and children born at<37 weeks of gestational age had a higher risk of childhood kidney disease after gestational systemic glucocorticoid exposure. If these findings are confirmed, they may inform clinicians who are considering prescribing systemic glucocorticoids during pregnancy.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - Lung-Chih Li
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine; Department of Internal Medicine, Division of Nephrology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - Hsiao-Ching Kuo
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
35
|
Rakers F, Schleussner E, Cornelius A, Kluckow S, Muth I, Hoyer D, Rupprecht S, Schultze T, Schiecke K, Ligges C, Schwab M, Hoyer H. Association between prenatal glucocorticoid exposure and adolescent neurodevelopment: An observational follow-up study. Acta Obstet Gynecol Scand 2024; 103:1530-1540. [PMID: 38877646 PMCID: PMC11266634 DOI: 10.1111/aogs.14885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/16/2024]
Abstract
INTRODUCTION Prenatal exposure to supraphysiological glucocorticoid (GC) levels may lead to long-lasting developmental changes in numerous biological systems. Our prior study identified an association between prenatal GC prophylaxis and reduced cognitive performance, electrocortical changes, and altered autonomic nervous system (ANS) activity in children aged 8-9 years. This follow-up study aimed to examine whether these findings persisted into adolescence. MATERIAL AND METHODS Prospective observational follow-up study involving twenty-one 14- to 15-year-old adolescents born to mothers who received betamethasone for induction of fetal lung maturation in threatened preterm birth, but who were born with a normal weight appropriate for their gestational age (median 37+4 gestational weeks). Thirty-five children not exposed to betamethasone served as the reference group (median 37+6 gestational weeks). The primary endpoint was cognitive performance, measured by intelligence quotient (IQ). Key secondary endpoints included symptoms of attention-deficit/hyperactivity disorder (ADHD) and metabolic markers. Additionally, we determined electrocortical (electroencephalogram), hypothalamus-pituitary-adrenal axis (HPAA), and ANS activity in response to a standardized stress paradigm. RESULTS No statistically significant group difference was observed in global IQ (adjusted mean: betamethasone 103.9 vs references 105.9, mean difference -2.0, 95% confidence interval [CI]: -7.12 to 3.12, p = 0.44). Similarly, ADHD symptoms, metabolic markers, the overall and stress-induced activity of the HPAA and the ANS did not differ significantly between groups. However, the betamethasone group exhibited reduced electrocortical activity in the frontal brain region (spectral edge frequency-adjusted means: 16.0 Hz vs 17.8 Hz, mean difference -1.83 Hz, 95% CI: -3.21 to -0.45, p = 0.01). CONCLUSIONS In 14- to 15-year-old adolescents, prenatal GC exposure was not associated with differences in IQ scores or ANS activity compared to unexposed controls. However, decelerated electrocortical activity in the frontal region potentially reflects disturbances in the maturation of cortical and/or subcortical brain structures. The clinical significance of these changes remains unknown. Given the small sample size, selective participation/loss of follow-up and potential residual confounding, these findings should be interpreted cautiously. Further research is required to replicate these results in larger cohorts before drawing firm clinical conclusions.
Collapse
Affiliation(s)
- Florian Rakers
- Hans Berger Department of NeurologyJena University HospitalJenaGermany
| | | | | | - Steffen Kluckow
- Hans Berger Department of NeurologyJena University HospitalJenaGermany
| | - Isabel Muth
- Department of ObstetricsJena University HospitalJenaGermany
| | - Dirk Hoyer
- Hans Berger Department of NeurologyJena University HospitalJenaGermany
| | - Sven Rupprecht
- Hans Berger Department of NeurologyJena University HospitalJenaGermany
| | - Torsten Schultze
- Hans Berger Department of NeurologyJena University HospitalJenaGermany
| | - Karin Schiecke
- Institute of Medical Statistics, Computer and Data SciencesJena University HospitalJenaGermany
| | - Carolin Ligges
- Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and PsychotherapyJena University HospitalJenaGermany
| | - Matthias Schwab
- Hans Berger Department of NeurologyJena University HospitalJenaGermany
| | - Heike Hoyer
- Institute of Medical Statistics, Computer and Data SciencesJena University HospitalJenaGermany
| |
Collapse
|
36
|
Deng F, Lei J, Chen J, Zhao M, Zhao C, Fu M, Sun M, Zhang M, Qiu J, Gao Q. DNA methylation-mediated 11βHSD2 downregulation drives the increases in angiotensin-converting enzyme and angiotensin II within preeclamptic placentas. FASEB J 2024; 38:e23714. [PMID: 38814727 DOI: 10.1096/fj.202400199r] [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: 01/25/2024] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024]
Abstract
Preeclampsia (PE) is a complex human-specific complication frequently associated with placental pathology. The local renin-angiotensin system (RAS) in the human placenta, which plays a crucial role in regulating placental function, has been extensively documented. Glucocorticoids (GCs) are a class of steroid hormones. PE cases often have abnormalities in GCs levels and placental GCs barrier. Despite extensive speculation, there is currently no robust evidence indicating that GCs regulate placental RAS. This study aims to investigate these potential relationships. Plasma and placental samples were collected from both normal and PE pregnancies. The levels of angiotensin-converting enzyme (ACE), angiotensin II (Ang II), cortisol, and 11β-hydroxysteroid dehydrogenases (11βHSD) were analyzed. In PE placentas, cortisol, ACE, and Ang II levels were elevated, while 11βHSD2 expression was reduced. Interestingly, a positive correlation was observed between ACE and cortisol levels in the placenta. A significant inverse correlation was found between the methylation statuses within the 11βHSD2 gene promoter and its expression, meanwhile, 11βHSD2 expression was negatively correlated with cortisol and ACE levels. In vitro experiments using placental trophoblast cells confirmed that active GCs can stimulate ACE transcription and expression through the GR pathway. Furthermore, 11βHSD2 knockdown could enhance this activating effect. An in vivo study using a rat model of intrauterine GCs overexposure during mid-to-late gestation suggested that excess GCs in utero lead to increased ACE and Ang II levels in the placenta. Collectively, this study provides the first evidence of the relationships between 11βHSD2 expression, GCs barrier, ACE, and Ang II levels in the placenta. It not only contributes to understanding the pathological features of the placental GCs barrier and RAS under PE conditions, also provides important information for revealing the pathological mechanism of PE.
Collapse
Affiliation(s)
- Fengying Deng
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jiahui Lei
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jie Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Meng Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
- Department of Obstetrics and Gynecology, the Third People's Hospital of Bengbu Affiliated to Bengbu Medical College, Bengbu, Anhui, China
| | - Chenxuan Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Mengyu Fu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Junlan Qiu
- Department of Oncology and Hematology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, P.R. China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
37
|
Liu Y, Chen SJ, Ai C, Yu PX, Fang M, Wang H. Prenatal dexamethasone exposure impairs rat blood-testis barrier function and sperm quality in adult offspring via GR/KDM1B/FSTL3/TGFβ signaling. Acta Pharmacol Sin 2024; 45:1237-1251. [PMID: 38472317 PMCID: PMC11130295 DOI: 10.1038/s41401-024-01244-5] [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: 10/22/2023] [Accepted: 02/15/2024] [Indexed: 03/14/2024]
Abstract
Both epidemiological and animal studies suggest that adverse environment during pregnancy can change the offspring development programming, but it is difficult to achieve prenatal early warning. In this study we investigated the impact of prenatal dexamethasone exposure (PDE) on sperm quality and function of blood-testis barrier (BTB) in adult offspring and the underlying mechanisms. Pregnant rats were injected with dexamethasone (0.1, 0.2 and 0.4 mg·kg-1·d-1, s.c.) from GD9 to GD20. After weaning (PW4), the pups were fed with lab chow. At PW12 and PW28, the male offspring were euthanized to collect blood and testes samples. We showed that PDE significantly decreased sperm quality (including quantity and motility) in male offspring, which was associated with impaired BTB and decreased CX43/E-cadherin expression in the testis. We demonstrated that PDE induced morphological abnormalities of fetal testicle and Sertoli cell development originated from intrauterine. By tracing to fetal testicular Sertoli cells, we found that PDE dose-dependently increased expression of histone lysine demethylases (KDM1B), decreasing histone 3 lysine 9 dimethylation (H3K9me2) levels of follistatin-like-3 (FSTL3) promoter region and increased FSTL3 expression, and inhibited TGFβ signaling and CX43/E-cadherin expression in offspring before and after birth. These results were validated in TM4 Sertoli cells following dexamethasone treatment. Meanwhile, the H3K9me2 levels of FSTL3 promoter in maternal peripheral blood mononuclear cell (PBMC) and placenta were decreased and its expression increased, which was positively correlated with the changes in offspring testis. Based on analysis of human samples, we found that the H3K9me2 levels of FSTL3 promoter in maternal blood PBMC and placenta were positively correlated with fetal blood testosterone levels after prenatal dexamethasone exposure. We conclude that PDE can reduce sperm quality in adult offspring rats, which is related to the damage of testis BTB via epigenetic modification and change of FSTL3 expression in Sertoli cells. The H3K9me2 levels of the FSTL3 promoter and its expression in the maternal blood PBMC can be used as a prenatal warning marker for fetal testicular dysplasia.
Collapse
Affiliation(s)
- Yi Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Si-Jia Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Can Ai
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Peng-Xia Yu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Man Fang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, 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.
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
38
|
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
|
39
|
Ninan K, Murphy KE, Asztalos EV, Jiang Y, Huszti E, Matthews SG, Santaguida P, Mukerji A, McDonald SD. The Impact of Infant Sex on Multiple Courses versus a Single Course of Antenatal Corticosteroids: A Secondary Analysis of a Randomized Controlled Trial. Am J Perinatol 2024; 41:e2919-e2926. [PMID: 37935374 DOI: 10.1055/s-0043-1776348] [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: 11/09/2023]
Abstract
OBJECTIVE Animal literature has suggested that the impact of antenatal corticosteroids (ACS) may vary by infant sex. Our objective was to assess the impact of infant sex on the use of multiple courses versus a single course of ACS and perinatal outcomes. STUDY DESIGN We conducted a secondary analysis of the Multiple Courses of Antenatal Corticosteroids for Preterm Birth trial, which randomly allocated pregnant people to multiple courses versus a single course of ACS. Our primary outcome was a composite of perinatal mortality or clinically significant neonatal morbidity (including neonatal death, stillbirth, severe respiratory distress syndrome, intraventricular hemorrhage [grade III or IV], cystic periventricular leukomalacia, and necrotizing enterocolitis [stage II or III]). Secondary outcomes included individual components of the primary outcome as well as anthropometric measures. Baseline characteristics were compared between participants who received multiple courses versus a single course of ACS. An interaction between exposure to ACS and infant sex was assessed for significance and multivariable regression analyses were conducted with adjustment for predefined covariates, when feasible. RESULTS Data on 2,300 infants were analyzed. The interaction term between treatment status (multiple courses vs. a single course of ACS) and infant sex was not significant for the primary outcome (p = 0.86), nor for any of the secondary outcomes (p > 0.05). CONCLUSION Infant sex did not modify the association between exposure to ACS and perinatal outcomes including perinatal mortality or neonatal morbidity or anthropometric outcomes. However, animal literature indicates that sex-specific differences after exposure to ACS may emerge over time and thus investigating long-term sex-specific outcomes warrants further attention. KEY POINTS · We explored the impact of infant sex on perinatal outcomes after multiple versus a single course of ACS.. · Infant sex was not a significant effect modifier of ACS exposure and perinatal outcomes.. · Animal literature indicates that sex-specific differences after ACS exposure may emerge over time.. · Further investigation of long-term sex-specific outcomes is warranted..
Collapse
Affiliation(s)
- Kiran Ninan
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Kellie E Murphy
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth V Asztalos
- Division of Neonatology, Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Yidi Jiang
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Ella Huszti
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Pasqualina Santaguida
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Amit Mukerji
- Division of Neonatology, Department of Pediatrics, McMaster University, Ontario, Canada
| | - Sarah D McDonald
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Radiology, McMaster University, Hamilton, Ontario, Canada
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
40
|
Eachus H, Choi MK, Tochwin A, Kaspareit J, Ho M, Ryu S. Elevated glucocorticoid alters the developmental dynamics of hypothalamic neurogenesis in zebrafish. Commun Biol 2024; 7:416. [PMID: 38580727 PMCID: PMC10997759 DOI: 10.1038/s42003-024-06060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 03/16/2024] [Indexed: 04/07/2024] Open
Abstract
Exposure to excess glucocorticoid (GC) during early development is implicated in adult dysfunctions. Reduced adult hippocampal neurogenesis is a well-known consequence of exposure to early life stress or elevated GC, however the effects on neurogenesis during development and effects on other brain regions are not well understood. Using an optogenetic zebrafish model, here we analyse the effects of GC exposure on neurogenesis during development in the whole brain. We identify that the hypothalamus is a highly GC-sensitive region where elevated GC causes precocious development. This is followed by failed maturation and early decline accompanied by impaired feeding, growth, and survival. In GC-exposed animals, the developmental trajectory of hypothalamic progenitor cells is strikingly altered, potentially mediated by direct regulation of transcription factors such as rx3 by GC. Our data provide cellular and molecular level insight into GC-induced alteration of the hypothalamic developmental trajectory, a process crucial for health across the life-course.
Collapse
Affiliation(s)
- Helen Eachus
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
- Institute of Health and Neurodevelopment & Aston Pharmacy School, Aston University, Birmingham, B4 7ET, UK
| | - Min-Kyeung Choi
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Anna Tochwin
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Johanna Kaspareit
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - May Ho
- Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Soojin Ryu
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
41
|
Yeramilli V, Rizek CS, Graham J, Taylor C, Cheddadi R, Patterson S, Watts S, Martin C. Parental preconception stress in zebrafish induces long-lasting anxiety in offspring. Physiol Behav 2024; 277:114477. [PMID: 38301945 DOI: 10.1016/j.physbeh.2024.114477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/03/2024]
Abstract
The growth and function of the vertebrate brain are impacted by environmental stimuli and early life stress. Adults who experience chronic stress during early life are more likely to suffer various neurodevelopmental and health issues. However, our understanding of how these specific environmental signals at different developmental stages affect brain development is poorly understood. In this study, we investigated if stress in parents prior to conception modulates neurodevelopment in offspring. We used a chronic unpredictable stress model adapted to zebrafish, which is an increasingly popular vertebrate model in neuroscience research to investigate the effects of both maternal and paternal preconception stress on offspring behavior. We evaluated the responsiveness of three anxiety-related behavioral paradigms in zebrafish: the novel tank test, thigmotaxis, and shoaling behavior. We found larvae from stressed females exhibited anxiety-like behavior in a thigmotaxis assay. As these larvae matured into adults, they continued to exhibit anxiety-like behavior in a novel tank and shoaling behavioral assay. These studies indicate preconception stress exposure in parents can induce life-long alterations in offspring neurodevelopment. Further, these results expand the hypothesis that chronically elevated glucocorticoid signaling not only in stressed mothers, but also stressed dads can affect neurodevelopment in offspring. We propose that zebrafish may be a useful model to study the transgenerational effects of chronic stress mediated via the maternal and paternal line.
Collapse
Affiliation(s)
- Venkata Yeramilli
- Dept of Surgery, Washington University School of Medicine, Saint Louis, MO, US
| | | | - Jessica Graham
- Dept of Surgery, Washington University School of Medicine, Saint Louis, MO, US
| | - Christopher Taylor
- Dept of Biology, University of Alabama at Birmingham, Birmingham, AL, US
| | - Riadh Cheddadi
- Dept of Surgery, Washington University School of Medicine, Saint Louis, MO, US
| | - Sophie Patterson
- Dept of Biology, University of Alabama at Birmingham, Birmingham, AL, US
| | - Stephen Watts
- Dept of Biology, University of Alabama at Birmingham, Birmingham, AL, US
| | - Colin Martin
- Dept of Surgery, Washington University School of Medicine, Saint Louis, MO, US.
| |
Collapse
|
42
|
Eachus H, Ryu S. Glucocorticoid effects on the brain: from adaptive developmental plasticity to allostatic overload. J Exp Biol 2024; 227:jeb246128. [PMID: 38449327 PMCID: PMC10949071 DOI: 10.1242/jeb.246128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Exposure to stress during early life may alter the developmental trajectory of an animal by a mechanism known as adaptive plasticity. For example, to enhance reproductive success in an adverse environment, it is known that animals accelerate their growth during development. However, these short-term fitness benefits are often associated with reduced longevity, a phenomenon known as the growth rate-lifespan trade-off. In humans, early life stress exposure compromises health later in life and increases disease susceptibility. Glucocorticoids (GCs) are major stress hormones implicated in these processes. This Review discusses the evidence for GC-mediated adaptive plasticity in development, leading to allostatic overload in later life. We focus on GC-induced effects on brain structure and function, including neurogenesis; highlight the need for longitudinal studies; and discuss approaches to identify molecular mechanisms mediating GC-induced alteration of the brain developmental trajectory leading to adult dysfunctions. Further understanding of how stress and GC exposure can alter developmental trajectories at the molecular and cellular level is of critical importance to reduce the burden of mental and physical ill health across the life course.
Collapse
Affiliation(s)
- Helen Eachus
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Soojin Ryu
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
43
|
Nemec-Bakk AS, Bel J, Niccoli S, Boreham DR, Tai TC, Lees SJ, Khaper N. Effects of prenatal dexamethasone exposure on adult C57BL/6J mouse metabolism and oxidative stress. Can J Physiol Pharmacol 2024; 102:180-195. [PMID: 38329060 DOI: 10.1139/cjpp-2023-0254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Prenatal glucocorticoid exposure has been shown to alter hypothalamic-pituitary-adrenal axis function resulting in altered fetal development that can persist through adulthood. Fetal exposure to excess dexamethasone, a synthetic glucocorticoid, has been shown to alter adult behaviour and metabolism. This study investigated the effects prenatal dexamethasone exposure had on adult offspring cardiac and liver metabolism and oxidative stress. Pregnant C57BL/6 mice received a dose of 0.4 mg/kg dexamethasone on gestational days 15-17. Once pups were approximately 7 months old, glucose uptake was determined using positron emission tomography and insulin resistance (IR) was determined by homeostatic model assessment (HOMA) IR calculation. Oxidative stress was assessed by measuring 4-hydroxynonenal protein adduct formation and total reactive oxygen species. Female dexamethasone group had significantly increased glucose uptake when insulin stimulated compared to vehicle-treated mice. HOMA IR revealed no evidence of IR in either male or female offspring. There was also no change in oxidative stress markers in either cardiac or liver tissues of male or female offspring. These data suggest that prenatal dexamethasone exposure in male mice does not alter oxidative stress or metabolism. However, prenatal dexamethasone exposure increased glucocorticoids, cardiac glucose uptake, and pAkt signaling in female heart tissues in adult mice, suggesting there are sex differences in prenatal dexamethasone exposure.
Collapse
Affiliation(s)
- A S Nemec-Bakk
- Department of Science and Environmental studies, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| | - J Bel
- Department of Science and Environmental studies, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| | - S Niccoli
- Medical Science Division, NOSM University, Thunder Bay, ON P7B 5E1, Canada
| | - D R Boreham
- Medical Science Division, NOSM University, Sudbury, ON P3E 2C6, Canada
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - T C Tai
- Medical Science Division, NOSM University, Sudbury, ON P3E 2C6, Canada
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - S J Lees
- Medical Science Division, NOSM University, Thunder Bay, ON P7B 5E1, Canada
- Department of Biology, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| | - N Khaper
- Medical Science Division, NOSM University, Thunder Bay, ON P7B 5E1, Canada
- Biomolecular Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Department of Biology, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| |
Collapse
|
44
|
Liu K, Chen Z, Hu W, He B, Xu D, Guo Y, Wang H. Intrauterine developmental origin, programming mechanism, and prevention strategy of fetal-originated hypercholesterolemia. Obes Rev 2024; 25:e13672. [PMID: 38069529 DOI: 10.1111/obr.13672] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/30/2023] [Accepted: 11/05/2023] [Indexed: 02/28/2024]
Abstract
There is increasing evidence that hypercholesterolemia has an intrauterine developmental origin. However, the pathogenesis of fetal-originated is still lacking in a theoretical system, which makes its clinical early prevention and treatment difficult. It has been found that an adverse environment during pregnancy (e.g., xenobiotic exposure) may lead to changes in fetal blood cholesterol levels through changing maternal cholesterol metabolic function and/or placental cholesterol transport function and may also directly affect the liver cholesterol metabolic function of the offspring in utero and continue after birth. Adverse environmental conditions during pregnancy may also raise maternal glucocorticoid levels and promote the placental glucocorticoid barrier opening, leading to fetal overexposure to maternal glucocorticoids. Intrauterine high-glucocorticoid exposure can alter the liver cholesterol metabolism of offspring, resulting in an increased susceptibility to hypercholesterolemia after birth. Abnormal epigenetic modifications are involved in the intrauterine programming mechanism of fetal-originated hypercholesterolemia. Some interventions targeted at pregnant mothers or offspring in early life have been proposed to effectively prevent and treat the development of fetal-originated hypercholesterolemia. In this paper, the recent research progress on fetal-originated hypercholesterolemia was reviewed, with emphasis on intrauterine maternal glucocorticoid programming mechanisms, in order to provide a theoretical basis for its early clinical warning, prevention, and treatment.
Collapse
Affiliation(s)
- Kexin Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ze Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wen Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bo He
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Dan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
45
|
Lu X, Chen B, Xu D, Hu W, Wang X, Dai Y, Wang Q, Peng Y, Chen K, Zhao D, Wang H. Epigenetic programming mediates abnormal gut microbiota and disease susceptibility in offspring with prenatal dexamethasone exposure. Cell Rep Med 2024; 5:101398. [PMID: 38301654 PMCID: PMC10897547 DOI: 10.1016/j.xcrm.2024.101398] [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: 11/18/2022] [Revised: 05/08/2023] [Accepted: 01/08/2024] [Indexed: 02/03/2024]
Abstract
Prenatal dexamethasone exposure (PDE) can lead to increased susceptibility to various diseases in adult offspring, but its effect on gut microbiota composition and the relationship with disease susceptibility remains unclear. In this study, we find sex-differential changes in the gut microbiota of 6-month-old infants with prenatal dexamethasone therapy (PDT) that persisted in female infants up to 2.5 years of age with altered bile acid metabolism. PDE female offspring rats show abnormal colonization and composition of gut microbiota and increased susceptibility to cholestatic liver injury. The aberrant gut microbiota colonization in the PDE offspring can be attributed to the inhibited Muc2 expression caused by decreased CDX2 expression before and after birth. Integrating animal and cell experiments, we further confirm that dexamethasone could inhibit Muc2 expression by activating GR/HDAC11 signaling and regulating CDX2 epigenetic modification. This study interprets abnormal gut microbiota and disease susceptibility in PDT offspring from intrauterine intestinal dysplasia.
Collapse
Affiliation(s)
- Xiaoqian Lu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Beidi Chen
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100191, China
| | - Dan Xu
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wen Hu
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Xia Wang
- Department of Pediatrics, Children's Digital Health, and Data Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Qian Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yu Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Kaiqi Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Dongchi Zhao
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China; Department of Pediatrics, Children's Digital Health, and Data Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China.
| |
Collapse
|
46
|
Liu J, Yang T, Li Y, Li S, Li Y, Xu S, Xia W. Associations of maternal exposure to 2,4-dichlorophenoxyacetic acid during early pregnancy with steroid hormones among one-month-old infants. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169414. [PMID: 38114038 DOI: 10.1016/j.scitotenv.2023.169414] [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: 10/28/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Exposure to 2,4-dichlorophenoxyacetic acid (2,4-D), a widely used hormonal herbicide, may disrupt steroid hormone homeostasis. However, evidence from population-based studies is limited, especially for one-month-old infants whose steroid hormones are in a state of adjustment to extrauterine life and can be important indicators of endocrine development. This study aimed to explore the associations between maternal 2,4-D exposure during early pregnancy and infant steroid hormone levels. METHODS The 885 mother-infant pairs were from a birth cohort in Wuhan, China. Maternal exposure to 2,4-D was determined in urine samples from early pregnancy, and nine steroid hormones were determined in infant urine. The associations of maternal 2,4-D exposure with infant steroid hormones and their product-to-precursor ratios were estimated based on generalized linear models, and bioinformatic analysis was conducted with public databases to explore the potential mechanisms involved. RESULTS The detection frequency of 2,4-D was 99.32 %, and the detection frequency of steroid hormones ranged from 98.42 % to 100.00 %. After adjusting for covariates, an interquartile range increase in 2,4-D concentrations was associated with a 7.84 % decrease in 11-deoxycortisol (95 % confidence interval, CI: -14.12 %, -1.10 %), an 8.09 % decrease in corticosterone (95 % CI: -14.56 %, -1.14 %), an 8.67 % decrease in cortisol (95 % CI: -14.43 %, -2.52 %), a 13.00 % decrease in cortisone (95 % CI: -20.64 %, -4.62 %), and an 11.17 % decrease in aldosterone (95 % CI: -19.62 %, -1.83 %). Maternal 2,4-D was also associated with lower infant cortisol/17α-OH-progesterone, cortisol/pregnenolone, and aldosterone/pregnenolone ratios. In bioinformatic analysis, pathways/biological processes related to steroid hormone synthesis and secretion were enriched from target genes of 2,4-D exposure. CONCLUSIONS Maternal urinary 2,4-D during early pregnancy was associated with lower infant urinary 11-deoxycortisol, corticosterone, cortisol, cortisone, and aldosterone, reflecting that 2,4-D exposure may interfere with infant steroid hormone homeostasis. Further efforts are still needed to study the relevant health effects of exposure to 2,4-D, particularly for vulnerable populations.
Collapse
Affiliation(s)
- Jiangtao Liu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Yang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Shulan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
47
|
van Haren JS, Delbressine FLM, Schoberer M, te Pas AB, van Laar JOEH, Oei SG, van der Hout-van der Jagt MB. Transferring an extremely premature infant to an extra-uterine life support system: a prospective view on the obstetric procedure. Front Pediatr 2024; 12:1360111. [PMID: 38425664 PMCID: PMC10902175 DOI: 10.3389/fped.2024.1360111] [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: 12/22/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
To improve care for extremely premature infants, the development of an extrauterine environment for newborn development is being researched, known as Artificial Placenta and Artificial Womb (APAW) technology. APAW facilitates extended development in a liquid-filled incubator with oxygen and nutrient supply through an oxygenator connected to the umbilical vessels. This setup is intended to provide the optimal environment for further development, allowing further lung maturation by delaying gas exposure to oxygen. This innovative treatment necessitates interventions in obstetric procedures to transfer an infant from the native to an artificial womb, while preventing fetal-to-neonatal transition. In this narrative review we analyze relevant fetal physiology literature, provide an overview of insights from APAW studies, and identify considerations for the obstetric procedure from the native uterus to an APAW system. Lastly, this review provides suggestions to improve sterility, fetal and maternal well-being, and the prevention of neonatal transition.
Collapse
Affiliation(s)
- Juliette S. van Haren
- Department of Industrial Design, Eindhoven University of Technology, Eindhoven, Netherlands
- Obstetrics and Gynaecology, Máxima Medical Centre, Veldhoven, Netherlands
| | | | - Mark Schoberer
- Institute for Applied Medical Engineering and Clinic for Neonatology, University Hospital Aachen, Aachen, Germany
| | - Arjan B. te Pas
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Judith O. E. H. van Laar
- Obstetrics and Gynaecology, Máxima Medical Centre, Veldhoven, Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - S. Guid Oei
- Obstetrics and Gynaecology, Máxima Medical Centre, Veldhoven, Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - M. Beatrijs van der Hout-van der Jagt
- Obstetrics and Gynaecology, Máxima Medical Centre, Veldhoven, Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
48
|
Chawanpaiboon S, Chukaew R, Pooliam J. A comparison of 2 doses of antenatal dexamethasone for the prevention of respiratory distress syndrome: an open-label, noninferiority, pragmatic randomized trial. Am J Obstet Gynecol 2024; 230:260.e1-260.e19. [PMID: 37442247 DOI: 10.1016/j.ajog.2023.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Antenatal corticosteroids have been used for the prevention of respiratory complications, intraventricular hemorrhage, necrotizing enterocolitis, and other adverse neonatal outcomes for over 50 years, with limited evidence about their optimal doses. Higher steroid doses or frequencies of antenatal corticosteroids in preterm newborns pose adverse effects such as prolonged adrenal suppression, negative effects on fetal programming and metabolism, and increased risks of neurodevelopmental and neuropsychological impairments. Conversely, lower doses of antenatal corticosteroids may be an effective alternative to induce fetal lung maturation with less risk to the fetus. Late preterm births represent the largest population of all preterm neonates, with a respiratory distress syndrome risk of 8.83%. Therefore, determining the optimal antenatal corticosteroid dosage is of particular importance for this population. OBJECTIVE This study aimed to compare the efficacy of 5-mg and 6-mg dexamethasone in preventing neonatal respiratory distress syndrome in women with preterm births at 320 to 366 weeks of gestation. STUDY DESIGN This was an open-label, randomized, controlled, noninferiority trial. Singleton pregnant women (n=370) at 320 to 366 weeks of gestation with spontaneous preterm labor or preterm premature rupture of membranes were enrolled. They were randomly assigned (1:1) to a 5-mg or 6-mg dexamethasone group. Dexamethasone was administered intramuscularly every 12 hours for 4 doses or until delivery. The primary outcome was the reduction in neonatal respiratory distress syndrome cases, whereas the secondary outcomes were any adverse maternal or neonatal events. RESULTS Between December 2020 and April 2022, 370 eligible women, anticipating deliveries within the gestational range of 32 0/7 to 36 6/7 weeks, willingly participated in the study. They were evenly split, with 185 women assigned to the 5-mg group and 185 to the 6-mg group. The study revealed that the demographic profiles of the participants in the 2 groups were remarkably similar, with no statistically significant disparities (P>.05). It is noteworthy that most of these women gave birth after 34 weeks of gestation. Despite a substantial proportion not completing the full course of steroid treatment, the 5-mg dose exhibited noninferiority compared with the 6-mg dose of dexamethasone, as indicated by a modest proportional difference of 0.5% (95% confidence interval, -2.8 to 43.9). Neonatal respiratory distress syndrome occurred in a relatively low percentage of newborns in both groups, affecting 2.2% in the 5-mg group and 1.6% in the 6-mg group. Notably, the risk difference of 0.6% fell comfortably within the predefined noninferiority threshold of 10%. CONCLUSION Our study suggests that a 5-mg dexamethasone dose is noninferior to a standard 6-mg dose in preventing neonatal respiratory distress syndrome in preterm births.
Collapse
Affiliation(s)
- Saifon Chawanpaiboon
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynaecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Ronnakorn Chukaew
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynaecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Julaporn Pooliam
- Clinical Epidemiological Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
49
|
Harders EP, Agustin M, Paitz RT. Avian extraembryonic membranes respond to yolk corticosterone early in development. Biol Open 2024; 13:bio060131. [PMID: 38156650 PMCID: PMC10836647 DOI: 10.1242/bio.060131] [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: 08/21/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
During times of maternal stress, developing embryos can be exposed to elevated levels of glucocorticoids, which can affect development and permanently alter offspring phenotype. In placental species, the placenta mediates fetal exposure to maternal glucocorticoids via metabolism, yet the placenta itself responds to glucocorticoids to regulate offspring growth and development. In oviparous species, maternal glucocorticoids can be deposited into the egg yolk and are metabolized early in development. This metabolism is mediated by the extraembryonic membranes, but it is unknown if the extraembryonic membranes also respond to maternal glucocorticoids in a way comparable to the placenta. In this study, we quantified the expression of acyl-CoA thioesterase 13 (Acot13) as an initial marker of the membrane's response to corticosterone in chicken (Gallus gallus) eggs. Acot13 regulates fatty acid processing in the embryo, to potentially regulate resource availability during development. We addressed the following questions using Acot13 expression: 1) Do the extraembryonic membranes respond to yolk corticosterone early in development? 2) Is the response to corticosterone dependent on the dose of corticosterone? 3) What is the duration of the response to corticosterone? 4) Does a metabolite of corticosterone (5β-corticosterone) elicit the same response as corticosterone? We found that corticosterone significantly induces the expression of Acot13 on day four of development and that expression of Acot13 increases with the dose of corticosterone. Further, we found expression of Acot13 is significantly elevated by corticosterone on days four and six of development compared to oil treated eggs, but not on days eight and ten. Although this response is transient, it occurs during a critical period of development and could initiate a cascade of events that ultimately alter offspring phenotype. Finally, we found that 5β-corticosterone does not increase the expression of Acot13, indicating that metabolism inactivates corticosterone. Ultimately, this study provides insight into the mechanisms underlying how maternally deposited glucocorticoids can affect embryonic development.
Collapse
Affiliation(s)
- Emily P. Harders
- Illinois State University, School of Biological Sciences, Campus Box 4120, Normal, IL 61790, USA
| | - Mitch Agustin
- Illinois State University, School of Biological Sciences, Campus Box 4120, Normal, IL 61790, USA
| | - Ryan T. Paitz
- Illinois State University, School of Biological Sciences, Campus Box 4120, Normal, IL 61790, USA
| |
Collapse
|
50
|
Keeton VF, Hoffmann TJ, Goodwin KM, Powell B, Tupuola S, Weiss SJ. Prenatal exposure to social adversity and infant cortisol in the first year of life. Stress 2024; 27:2316042. [PMID: 38377153 PMCID: PMC11006384 DOI: 10.1080/10253890.2024.2316042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/03/2024] [Indexed: 02/22/2024] Open
Abstract
Exposure to social adversity has been associated with cortisol dysregulation during pregnancy and in later childhood; less is known about how prenatal exposure to social stressors affects postnatal cortisol of infants. In a secondary analysis of data from a longitudinal study, we tested whether a pregnant woman's reports of social adversity during the third trimester were associated with their infant's resting cortisol at 1, 6, and 12 months postnatal. Our hypothesis was that prenatal exposure to social adversity would be associated with elevation of infants' cortisol. Measures included prenatal survey reports of social stressors and economic hardship, and resting cortisol levels determined from infant saliva samples acquired at each postnatal timepoint. Data were analyzed using linear mixed effects models. The final sample included 189 women and their infants (46.56% assigned female sex at birth). Prenatal economic hardship was significantly associated with infant cortisol at 6 months postnatal; reports of social stressors were not significantly associated with cortisol at any time point. Factors associated with hardship, such as psychological distress or nutritional deficiencies, may alter fetal HPA axis development, resulting in elevated infant cortisol levels. Developmental changes unique to 6 months of age may explain effects at this timepoint. More work is needed to better comprehend the complex pre- and post-natal physiologic and behavioral factors that affect infant HPA axis development and function, and the modifying role of environmental exposures.
Collapse
Affiliation(s)
- Victoria F. Keeton
- Assistant Professor, Betty Irene Moore School of Nursing, University of California, Davis, 2570 48 St., Sacramento, CA, USA 95817
- University of California, San Francisco, Department of Obstetrics, Gynecology, and Reproductive Sciences, CA Preterm Birth Initiative, USA
| | - Thomas J. Hoffmann
- Department of Epidemiology and Biostatistics, and Office of Research School of Nursing, University of California, San Francisco, USA
| | - Kalisha Moneé Goodwin
- University of California, San Francisco, Department of Obstetrics, Gynecology, and Reproductive Sciences, CA Preterm Birth Initiative, USA
| | - Bree Powell
- University of California, San Francisco, Department of Obstetrics, Gynecology, and Reproductive Sciences, CA Preterm Birth Initiative, USA
| | - Sophia Tupuola
- University of California, San Francisco, Department of Obstetrics, Gynecology, and Reproductive Sciences, CA Preterm Birth Initiative, USA
| | - Sandra J. Weiss
- Department of Community Health Systems, University of California, San Francisco, USA
| |
Collapse
|