1
|
Tang J, Chen Y, Xue P, Chen Y, Kong H, Lin C, Wang X, Liu S. Exposure to synthetic steroid hormones and precocious puberty in girls: A case-control study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116814. [PMID: 39083867 DOI: 10.1016/j.ecoenv.2024.116814] [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: 05/14/2024] [Revised: 07/15/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Synthetic steroid hormones are an emerging class of environmental pollutants, but their influence on pubertal timing remains unclear. This case-control study explored the association between synthetic steroid hormone exposure and precocious puberty. Using ultrahigh performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), synthetic steroid hormones were detected in urine samples from 229 Chinese girls, aged 6-9 years. Puberty status was assessed using Tanner staging by professional pediatricians. We conducted the least absolute shrinkage and selection operator (LASSO) regression combined with logistic regression. Besides, we evaluated the joint effects of steroid hormone mixture and identified the main contributor using the Weighted quantile sum (WQS) model and Bayesian kernel machine regression (BKMR) model. The logistic regression model reflected an inverse individual association between precocious puberty and halcinonide [OR (95 %CI): 0.20 (0.07, 0.46)], and budesonide [OR (95 %CI): 0.77 (0.62, 0.95)]. In the joint effects utilizing the WQS model, precocious puberty showed a marginal association with steroid hormone mixture, but was not significant [OR (95 %CI): 0.88 (0.75, 1.04)]. Prednisolone (0.31), fluorometholone acetate (0.24), and dexamethasone acetate (0.12) had the highest weight. Consistently, mixture exposure was not associated with precocious puberty in the BKMR model. In conclusion, precocious puberty was associated with halcinonide and budesonide exposure, but not steroid hormone mixture among girls. It highlighted the management of the residual synthetic steroid hormones in the environment and provided a direction for the prevention of precocious puberty.
Collapse
Affiliation(s)
- Jingyi Tang
- School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Hainan 572022, China
| | - Yao Chen
- Department of Endocrinology and Genetic Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Peng Xue
- School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Hainan 572022, China
| | - Yang Chen
- School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Hainan 572022, China
| | - Huijun Kong
- Department of Pediatrics, Qufu People's Hospital, Qufu, Shandong 273160, China
| | - Cuilan Lin
- Boai Hospital of Zhongshan, Southern Medical University, Zhongshan, Guangdong 528405, China
| | - Xiumin Wang
- Department of Endocrinology and Genetic Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shijian Liu
- School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Hainan 572022, China.
| |
Collapse
|
2
|
Akbarinejad V, Cushman RA. Developmental programming of reproduction in the female animal. Anim Reprod Sci 2024; 263:107456. [PMID: 38503204 DOI: 10.1016/j.anireprosci.2024.107456] [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: 12/24/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
Successful reproduction is a cornerstone in food animal industry in order to sustain food production for human. Therefore, various methods focusing on genetics and postnatal environment have been identified and applied to improve fertility in livestock. Yet there is evidence indicating that environmental factors during prenatal and/or neonatal life can also impact the function of reproductive system and fertility in the animals during adulthood, which is called the developmental programming of reproduction. The current review summarizes data associated with the developmental origins of reproduction in the female animals. In this regard, this review focuses on the effect of plane of nutrition, maternal body condition, hypoxia, litter size, maternal age, parity, level of milk production and milk components, lactocrine signaling, stress, thermal stress, exposure to androgens, endocrine disrupting chemicals, mycotoxins and pollutants, affliction with infection and inflammation, and maternal gut microbiota during prenatal and neonatal periods on the neuroendocrine system, puberty, health of reproductive organs and fertility in the female offspring. It is noteworthy that these prenatal and neonatal factors do not always exert their effects on the reproductive performance of the female by compromising the development of organs directly related to reproductive function such as hypothalamus, pituitary, ovary, oviduct and uterus. Since they can impair the development of non-reproductive organs and systems modulating reproductive function as well (e.g., metabolic system and level of milk yield in dairy animals). Furthermore, when these factors affect the epigenetics of the offspring, their adverse effects will not be limited to one generation and can transfer transgenerationally. Hence, pinpointing the factors influencing developmental programming of reproduction and considering them in management of livestock operations could be a potential strategy to help improve fertility in food animals.
Collapse
Affiliation(s)
- Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Robert A Cushman
- USDA, Agricultural Research Service, US. Meat Animal Research Center, Clay Center, NE 68933-0166, United States
| |
Collapse
|
3
|
Giri T, Maloney SE, Giri S, Goo YA, Song JH, Son M, Tycksen E, Conyers SB, Bice A, Ge X, Garbow JR, Quirk JD, Bauer AQ, Palanisamy A. Oxytocin-induced birth causes sex-specific behavioral and brain connectivity changes in developing rat offspring. iScience 2024; 27:108960. [PMID: 38327784 PMCID: PMC10847747 DOI: 10.1016/j.isci.2024.108960] [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: 08/21/2023] [Revised: 11/23/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024] Open
Abstract
Despite six decades of the use of exogenous oxytocin for management of labor, little is known about its effects on the developing brain. Motivated by controversial reports suggesting a link between oxytocin use during labor and autism spectrum disorders (ASDs), we employed our recently validated rat model for labor induction with oxytocin to address this important concern. Using a combination of molecular biological, behavioral, and neuroimaging assays, we show that induced birth with oxytocin leads to sex-specific disruption of oxytocinergic signaling in the developing brain, decreased communicative ability of pups, reduced empathy-like behaviors especially in male offspring, and widespread sex-dependent changes in functional cortical connectivity. Contrary to our hypothesis, social behavior, typically impaired in ASDs, was largely preserved. Collectively, our foundational studies provide nuanced insights into the neurodevelopmental impact of birth induction with oxytocin and set the stage for mechanistic investigations in animal models and prospective longitudinal clinical studies.
Collapse
Affiliation(s)
- Tusar Giri
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan E. Maloney
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Saswat Giri
- Graduate Student, School of Public Health and Social Justice, St. Louis University, St. Louis, MO, USA
| | - Young Ah Goo
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Jong Hee Song
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Minsoo Son
- Mass Spectrometry Technology Access Center (MTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric Tycksen
- Genome Technology Access Center (GTAC), McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Sara B. Conyers
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Annie Bice
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xia Ge
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel R. Garbow
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - James D. Quirk
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Adam Q. Bauer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Arvind Palanisamy
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
4
|
Bhaumik S, Lockett J, Cuffe J, Clifton VL. Glucocorticoids and Their Receptor Isoforms: Roles in Female Reproduction, Pregnancy, and Foetal Development. BIOLOGY 2023; 12:1104. [PMID: 37626990 PMCID: PMC10452123 DOI: 10.3390/biology12081104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023]
Abstract
Alterations in the hypothalamic-pituitary-adrenal (HPA) axis and associated changes in circulating levels of glucocorticoids are integral to an organism's response to stressful stimuli. Glucocorticoids acting via glucocorticoid receptors (GRs) play a role in fertility, reproduction, placental function, and foetal development. GRs are ubiquitously expressed throughout the female reproductive system and regulate normal reproductive function. Stress-induced glucocorticoids have been shown to inhibit reproduction and affect female gonadal function by suppressing the hypothalamic-pituitary-gonadal (HPG) axis at each level. Furthermore, during pregnancy, a mother's exposure to prenatal stress or external glucocorticoids can result in long-lasting alterations to the foetal HPA and neuroendocrine function. Several GR isoforms generated via alternative splicing or translation initiation from the GR gene have been identified in the mammalian ovary and uterus. The GR isoforms identified include the splice variants, GRα and GRβ, and GRγ and GR-P. Glucocorticoids can exert both stimulatory and inhibitory effects and both pro- and anti-inflammatory functions in the ovary, in vitro. In the placenta, thirteen GR isoforms have been identified in humans, guinea pigs, sheep, rats, and mice, indicating they are conserved across species and may be important in mediating a differential response to stress. Distinctive responses to glucocorticoids, differential birth outcomes in pregnancy complications, and sex-based variations in the response to stress could all potentially be dependent on a particular GR expression pattern. This comprehensive review provides an overview of the structure and function of the GR in relation to female fertility and reproduction and discusses the changes in the GR and glucocorticoid signalling during pregnancy. To generate this overview, an extensive non-systematic literature search was conducted across multiple databases, including PubMed, Web of Science, and Google Scholar, with a focus on original research articles, meta-analyses, and previous review papers addressing the subject. This review integrates the current understanding of GR variants and their roles in glucocorticoid signalling, reproduction, placental function, and foetal growth.
Collapse
Affiliation(s)
- Sreeparna Bhaumik
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
| | - Jack Lockett
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Brisbane 4102, Australia
| | - James Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane 4067, Australia;
| | - Vicki L. Clifton
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
| |
Collapse
|
5
|
Edwards PD, Palme R, Boonstra R. Is chronic stress a causal mechanism for small mammal population cycles? Reconciling the evidence. Oecologia 2023; 201:609-623. [PMID: 36864247 DOI: 10.1007/s00442-023-05338-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 02/17/2023] [Indexed: 03/04/2023]
Abstract
Chronic stress has long been hypothesized to play a role in driving population cycles. Christian (1950) hypothesized that high population density results in chronic stress and mass "die-offs" in small mammal populations. Updated variations of this hypothesis propose that chronic stress at high population density may reduce fitness, reproduction, or program aspects of phenotype, driving population declines. We tested the effect of density on the stress axis in meadow voles (Microtus pennsylvanicus) by manipulating population density in field enclosures over three years. Using fecal corticosterone metabolites as a non-invasive measure of glucocorticoid (GC) concentrations, we found that density alone was not associated with GC differences. However, we found that the seasonal relationship of GC levels differed by density treatment, with high-density populations having elevated GC levels early in the breeding season and decreasing towards late summer. We additionally tested hippocampal glucocorticoid receptor and mineralocorticoid receptor gene expression in juvenile voles born at different densities, with the hypothesis that high density may reduce receptor expression, altering negative feedback of the stress axis. We found that females had marginally higher glucocorticoid receptor expression at high density, no effect in males, and no detectable effect of density on mineralocorticoid receptor expression in either sex. Hence, we found no evidence that high density directly impairs negative feedback in the hippocampus, but rather female offspring may be better equipped for negative feedback. We compare our findings with prior studies to attempt to disentangle the complicated relationship between density, seasonality, sex, reproduction and the stress axis.
Collapse
Affiliation(s)
- Phoebe D Edwards
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, M5S 1A1, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, M1C 1A4, Canada.
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, L5L 1C6, Canada.
| | - Rupert Palme
- Department of Biomedical Sciences, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Rudy Boonstra
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, M1C 1A4, Canada
| |
Collapse
|
6
|
Medeiros LDS, Rodrigues PDS, Santos DNL, Silva-Sampaio AC, Kirsten TB, Suffredini IB, Coque ADC, da Silva RA, Bernardi MM. Prenatal restraint stress downregulates the hypothalamic kisspeptidergic system transcripts genes, reduces the estrogen plasma levels, delayed the onset of puberty, and reduced the sexual behavior intensity in female rats. Physiol Behav 2023; 260:114055. [PMID: 36563733 DOI: 10.1016/j.physbeh.2022.114055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 10/28/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
AIMS This study investigated the possible relationships between the expression of the Kiss1 and Gpr54 gene expressions and the pituitary-gonadal hormones with the female onset of puberty and sexual behavior. The Kiss1 and Gpr54 gene expressions were examined because they are critical to controlling the hypothalamic activation of GnRH neurons and, in turn, the pituitary-gonadal hormones related to the early onset of puberty and sexual behavior. Further, it was evaluated that the pituitary and gonadal hormones involved in the vaginal opening and the expression of sexual behavior. METHODS Pregnant rats exposed to PRS from gestation days 17 to 20 were evaluated for maternal and open-field behaviors. The maternal behavior was analyzed because it may alter brain sexual organization affecting the pups development. It was observed in female pups the physical and development and, in adult age, the open-field behavior, the anxiety-like behavior, the estrous cycle, the sexual behavior, the serum FSH, LH, estrogen, progesterone, and testosterone levels, and the gene expression of kisspeptin protein (Kiss1) and Gpr54 in the hypothalamus. RESULTS the maternal and open-field behaviors were unaffected. In the F1 generation, PRS reduced weight at weaning, delayed the day of the vaginal opening and reduced the intensity of lordosis, the estrogen levels, and the Kiss1 and Gpr54 gene expression. These effects were attributed to hypothalamic kisspeptidergic system downregulation of transcripts genes and the reduced estrogen levels affected by the PRS.
Collapse
Affiliation(s)
- Loren da Silva Medeiros
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Paula da Silva Rodrigues
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Daniel Nascimento Lago Santos
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Ana Claudia Silva-Sampaio
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Thiago Berti Kirsten
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Ivana Barbosa Suffredini
- Núcleo de Pesquisas em Biodiversidade, Laboratório de Extração, Universidade Paulista - UNIP, Av. Paulista, 900, São Paulo, SP 01310-100, Brazil
| | - Alex de Camargo Coque
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Rodrigo Augusto da Silva
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil; School of Dentistry, Graduate Program in Health Sciences, University of Taubaté, Rua dos Operários, 9, Taubaté, SP 12020-340, Brazil
| | - Maria Martha Bernardi
- Psychoneuroimmunology Laboratory, Graduate Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil.
| |
Collapse
|
7
|
Ashby GB, Riggan KA, Huang L, Torbenson VE, Long ME, Wick MJ, Allyse MA, Rivera-Chiauzzi EY. "I had so many life-changing decisions I had to make without support": a qualitative analysis of women's pregnant and postpartum experiences during the COVID-19 pandemic. BMC Pregnancy Childbirth 2022; 22:537. [PMID: 35787675 PMCID: PMC9251587 DOI: 10.1186/s12884-022-04816-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/31/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The COVID-19 pandemic has posed profound challenges for pregnant patients and their families. Studies conducted early in the pandemic found that pregnant individuals reported increased mental health concerns in response to pandemic-related stress. Many obstetric practices changed their healthcare delivery models, further impacting the experiences of pregnant patients. We conducted a survey study to explore the ways in which COVID-19 impacted the lives of pregnant and newly postpartum people. METHODS A mixed-methods survey was distributed to all patients ≥18 years old who were pregnant between January 1st, 2020 - April 28, 2021 in a large Midwest health system. Open-ended survey responses were analyzed for common themes using standard qualitative methodology. RESULTS Among the 1182 survey respondents, 647 women provided an open-ended response. Of these, 77% were in the postpartum period. The majority of respondents identified as white, were partnered or married, and owned their own home. Respondents reported feeling greater uncertainty, social isolation, as though they had limited social and practical support, and negative mental health effects as a result of the pandemic. Many cited sudden or arbitrary changes to their medical care as a contributing factor. Though in the minority, some respondents also reported benefits from the changes to daily life, including perceived improvements to medical care, better work-life balance, and opportunities for new perspectives. CONCLUSIONS This large qualitative dataset provides insight into how healthcare policy and lifestyle changes impacted pregnant and postpartum people. Respondents expressed similar levels of uncertainty and mental health concerns compared to other cohorts but less overall positivity. Our findings suggest greater attention be given to the impact of pandemic-related stress on pregnant and postpartum women. As the pandemic continues, these data identify areas where investment in additional support may have the greatest impact.
Collapse
Affiliation(s)
| | - Kirsten A Riggan
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN, USA
| | - Lily Huang
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | | | - Margaret E Long
- Department of Obstetrics & Gynecology, Mayo Clinic, Rochester, MN, USA
| | - Myra J Wick
- Department of Obstetrics & Gynecology, Mayo Clinic, Rochester, MN, USA
| | - Megan A Allyse
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, MN, USA
- Department of Obstetrics & Gynecology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
8
|
Sánchez-Garrido MA, García-Galiano D, Tena-Sempere M. Early programming of reproductive health and fertility: novel neuroendocrine mechanisms and implications in reproductive medicine. Hum Reprod Update 2022; 28:346-375. [PMID: 35187579 PMCID: PMC9071071 DOI: 10.1093/humupd/dmac005] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/29/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND According to the Developmental Origins of Health and Disease (DOHaD) hypothesis, environmental changes taking place during early maturational periods may alter normal development and predispose to the occurrence of diverse pathologies later in life. Indeed, adverse conditions during these critical developmental windows of high plasticity have been reported to alter the offspring developmental trajectory, causing permanent functional and structural perturbations that in the long term may enhance disease susceptibility. However, while solid evidence has documented that fluctuations in environmental factors, ranging from nutrient availability to chemicals, in early developmental stages (including the peri-conceptional period) have discernible programming effects that increase vulnerability to develop metabolic perturbations, the impact and eventual mechanisms involved, of such developmental alterations on the reproductive phenotype of offspring have received less attention. OBJECTIVE AND RATIONALE This review will summarize recent advances in basic and clinical research that support the concept of DOHaD in the context of the impact of nutritional and hormonal perturbations, occurring during the periconceptional, fetal and early postnatal stages, on different aspects of reproductive function in both sexes. Special emphasis will be given to the effects of early nutritional stress on the timing of puberty and adult gonadotropic function, and to address the underlying neuroendocrine pathways, with particular attention to involvement of the Kiss1 system in these reproductive perturbations. The implications of such phenomena in terms of reproductive medicine will also be considered. SEARCH METHODS A comprehensive MEDLINE search, using PubMed as main interface, of research articles and reviews, published mainly between 2006 and 2021, has been carried out. Search was implemented using multiple terms, focusing on clinical and preclinical data from DOHaD studies, addressing periconceptional, gestational and perinatal programming of reproduction. Selected studies addressing early programming of metabolic function have also been considered, when relevant. OUTCOMES A solid body of evidence, from clinical and preclinical studies, has documented the impact of nutritional and hormonal fluctuations during the periconceptional, prenatal and early postnatal periods on pubertal maturation, as well as adult gonadotropic function and fertility. Furthermore, exposure to environmental chemicals, such as bisphenol A, and maternal stress has been shown to negatively influence pubertal development and gonadotropic function in adulthood. The underlying neuroendocrine pathways and mechanisms involved have been also addressed, mainly by preclinical studies, which have identified an, as yet incomplete, array of molecular and neurohormonal effectors. These include, prominently, epigenetic regulatory mechanisms and the hypothalamic Kiss1 system, which likely contribute to the generation of reproductive alterations in conditions of early nutritional and/or metabolic stress. In addition to the Kiss1 system, other major hypothalamic regulators of GnRH neurosecretion, such as γ-aminobutyric acid and glutamate, may be targets of developmental programming. WIDER IMPLICATIONS This review addresses an underdeveloped area of reproductive biology and medicine that may help to improve our understanding of human reproductive disorders and stresses the importance, and eventual pathogenic impact, of early determinants of puberty, adult reproductive function and fertility.
Collapse
Affiliation(s)
- Miguel Angel Sánchez-Garrido
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
| | - David García-Galiano
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
9
|
Dooley N, Ruigrok A, Holt R, Allison C, Tsompanidis A, Waldman J, Auyeung B, Lombardo MV, Baron-Cohen S. Is there an association between prenatal testosterone and autistic traits in adolescents? Psychoneuroendocrinology 2022; 136:105623. [PMID: 34896742 PMCID: PMC8783053 DOI: 10.1016/j.psyneuen.2021.105623] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/27/2022]
Abstract
Prenatal testosterone (pT) is a crucial component in physiological masculinization in humans. In line with the Prenatal Sex Steroid Theory of autism, some studies have found a positive correlation between pT and autistic traits in childhood. However, effects in adolescence have not been explored. Hormonal and environmental changes occurring during puberty may alter the strength or the nature of prenatal effects on autistic traits. The current study examines if pT relates to autistic traits in a non-clinical sample of adolescents and young adults (N = 97, 170 observations; age 13-21 years old). It also explores pT interactions with pubertal stage and timing. PT concentrations were measured from amniotic fluid extracted in the 2nd trimester of gestation via amniocentesis conducted for clinical purposes. Autistic traits were measured by self- and parent-reports on the Autism Spectrum Quotient (AQ) which provides a total score and 5 sub-scores (social skills, communication, imagination, attention switching and attention to detail). Self-reported pubertal stage was regressed on age to provide a measure of relative timing. We found no statistical evidence for a direct association between pT and autistic traits in this adolescent sample (males, females or full sample). Exploratory analyses suggested that pT correlated positively with autistic traits in adolescents with earlier puberty-onset, but statistical robustness of this finding was limited. Further exploratory post-hoc tests suggested the pT-by-pubertal timing interaction was stronger in males relative to females, in self-reported compared to parent-reported AQ and specifically for social traits. These findings require replication in larger samples. Findings have implications for understanding the effects of pT on human behavior, specifically existence of effects in adolescence.
Collapse
Affiliation(s)
- Niamh Dooley
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland; Autism Research Centre, Department of Psychiatry, University of Cambridge, UK.
| | - Amber Ruigrok
- Autism Research Centre, Department of Psychiatry, University of Cambridge, UK
| | - Rosemary Holt
- Autism Research Centre, Department of Psychiatry, University of Cambridge, UK
| | - Carrie Allison
- Autism Research Centre, Department of Psychiatry, University of Cambridge, UK
| | | | - Jack Waldman
- Autism Research Centre, Department of Psychiatry, University of Cambridge, UK
| | - Bonnie Auyeung
- Autism Research Centre, Department of Psychiatry, University of Cambridge, UK,Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, UK
| | - Michael V. Lombardo
- Autism Research Centre, Department of Psychiatry, University of Cambridge, UK,Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn,Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, UK
| |
Collapse
|
10
|
Yan S, Lyu J, Liu Z, Zhou S, Ji Y, Wang H. Association of gestational hypertension and preeclampsia with offspring adiposity: A systematic review and meta-analysis. Front Endocrinol (Lausanne) 2022; 13:906781. [PMID: 36082079 PMCID: PMC9445980 DOI: 10.3389/fendo.2022.906781] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The association of gestational hypertension (GH) and preeclampsia (PE) with offspring adiposity outcomes had controversial results in different studies. OBJECTIVE We conducted a systematic review and meta-analysis to evaluate the relationship between maternal GH/PE and offspring adiposity outcomes. SEARCH STRATEGY Studies were identified in PubMed, Embase, and Cochrane databases, with keywords including "gestational hypertension", "preeclampsia", "offspring", "weight", "cohort study", etc., without year restriction. This study was registered with PROSPERO, CRD42022292084. SELECTION CRITERIA We set the selection criteria for six aspects: population, outcome, time frame, study design, and availability. For the studies included in the meta-analysis, we required the potential confounders in these studies have been adjusted. DATA COLLECTION AND ANALYSIS Two reviewers independently evaluated the data from the included studies. The meta-analyses included mean differences, regression coefficients, and corresponding 95% confidence intervals. Results were performed using RevMan software (version 5.4; Cochrane Collaboration). Heterogeneity among the included studies was assessed using the I2 statistic. MAIN RESULTS A total of 16 studies were included in our review, 15 of which were evaluated as high quality. In all offspring, during the early life (28 days-36 months), GH/PE exposure was found to be not or inversely associated with offspring obesity, then become positively associated at larger ages (3-19 years old). In offspring with adverse birth outcomes, the maternal GH/PE-exposed group had a lower weight in the short term (28 days to 18 months), but there was a trend of rapid weight gain as they grew older, compared with the non-exposed group. The meta-analysis showed that the BMI of the female offspring in the maternal PE-exposed group was significantly higher than that of the non-exposed offspring (MD=1.04, 95% CI: 0.67~1.42, P < 0.05). CONCLUSIONS The systematic review suggested that maternal exposure to de novo hypertension disorders of pregnancy (HDP) was associated with obesity in offspring, extending from early childhood to adolescence. The meta-analysis showed that PE was associated with higher BMI in female offspring. More studies are needed to conduct stratified analyses by PE/GH, the severity of HDP, or gender. SYSTEMATIC REVIEW REGISTRATION PROSPERO, identifier CRD42022292084.
Collapse
Affiliation(s)
- Shiyu Yan
- School of Public Health, Zhejiang University Medical School, Hangzhou, China
| | - Jinlang Lyu
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Zheng Liu
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Shuang Zhou
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yuelong Ji
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
- *Correspondence: Haijun Wang, ; Yuelong Ji,
| | - Haijun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
- *Correspondence: Haijun Wang, ; Yuelong Ji,
| |
Collapse
|
11
|
Thomason ME, Hect JL, Waller R, Curtin P. Interactive relations between maternal prenatal stress, fetal brain connectivity, and gestational age at delivery. Neuropsychopharmacology 2021; 46:1839-1847. [PMID: 34188185 PMCID: PMC8357800 DOI: 10.1038/s41386-021-01066-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Studies reporting significant associations between maternal prenatal stress and child outcomes are frequently confounded by correlates of prenatal stress that influence the postnatal rearing environment. The major objective of this study is to identify whether maternal prenatal stress is associated with variation in human brain functional connectivity prior to birth. We utilized fetal fMRI in 118 fetuses [48 female; mean age 32.9 weeks (SD = 3.87)] to evaluate this association and further addressed whether fetal neural differences were related to maternal health behaviors, social support, or birth outcomes. Community detection was used to empirically define networks and enrichment was used to isolate differential within- or between-network connectivity effects. Significance for χ2 enrichment was determined by randomly permuting the subject pairing of fetal brain connectivity and maternal stress values 10,000 times. Mixtures modelling was used to test whether fetal neural differences were related to maternal health behaviors, social support, or birth outcomes. Increased maternal prenatal negative affect/stress was associated with alterations in fetal frontoparietal, striatal, and temporoparietal connectivity (β = 0.82, p < 0.001). Follow-up analysis demonstrated that these associations were stronger in women with better health behaviors, more positive interpersonal support, and lower overall stress (β = 0.16, p = 0.02). Additionally, magnitude of stress-related differences in neural connectivity was marginally correlated with younger gestational age at delivery (β = -0.18, p = 0.05). This is the first evidence that negative affect/stress during pregnancy is reflected in functional network differences in the human brain in utero, and also provides information about how positive interpersonal and health behaviors could mitigate prenatal brain programming.
Collapse
Affiliation(s)
- Moriah E Thomason
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, USA.
- Department of Population Health, New York University Medical Center, New York, NY, USA.
- Neuroscience Institute, NYU Langone Health, New York, NY, USA.
| | - Jasmine L Hect
- Medical Scientist Training Program, University of Pittsburgh & Carnegie Mellon University, Pittsburgh, PA, USA
| | - Rebecca Waller
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Curtin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
12
|
Jeje S, Akpan E, Kunle-Alabi O, Akindele O, Raji Y. Protective role of Allium cepa Linn (onion) juice on maternal dexamethasone induced alterations in reproductive functions of female offspring of Wistar rats. Curr Res Physiol 2021; 4:145-154. [PMID: 34746834 PMCID: PMC8562199 DOI: 10.1016/j.crphys.2021.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/21/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
Maternal treatment with dexamethasone induces oxidative stress in the reproductive structures of offspring. Consumption of Allium cepa Linn improves antioxidant status. This study was designed to evaluate the protective role of Allium cepa Linn juice on maternal dexamethasone induced alterations in reproductive functions of the female offspring of Wistar rats. Twenty lactating dams (180-200 g) were randomly assigned into four groups (n = 5) on the day of parturition and treated as follows during lactation for 21 days: Control (5 ml/kg BW distilled water); Dexamethasone (60 μg/kg BW); Allium cepa (5 ml/kg BW); Dexamethasone + Allium cepa (60 μg/kg BW + 5 ml/kg BW). The female offspring were separated at birth. Days of vaginal opening and first oestrus cycle, length and frequency of estrous cycle as well as serum hormonal profiles were assessed as measure of reproductive functions. Ovarian superoxide dismutase (SOD) activity, catalase activity and malondialdehyde (MDA) level were measured as indices of oxidative stress. Oestrous cycle length, frequencies of diestrus as well as the Ovarian MDA were significantly increased (p < 0.05) in dexamethasone (DEX) group relative to control group. Serum 17β-oestradiol and corticosterone level in addition to SOD and catalase activities were significantly reduced (p < 0.05) in DEX group relative to control. Co-administration of Dex with Allium cepa Linn juice reduced the oestrous length, frequency of diestrous as well as ovarian MDA. There was also a significant increase in serum 17β-oestradiol, ovarian SOD and catalase activity. The results suggest that Allium cepa could protect against alterations in reproductive functions of offspring induced by maternal treatment with dexamethasone during lactation in Wistar rats. The flavonoid constituent of onion may also help in reducing oxidative stress in the offspring.
Collapse
Affiliation(s)
- S.O. Jeje
- Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - E.E. Akpan
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - O.T. Kunle-Alabi
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - O.O. Akindele
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - Y. Raji
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
13
|
Lamadé EK, Hendlmeier F, Wudy SA, Witt SH, Rietschel M, Coenen M, Gilles M, Deuschle M. Rhythm of Fetoplacental 11β-Hydroxysteroid Dehydrogenase Type 2 - Fetal Protection From Morning Maternal Glucocorticoids. J Clin Endocrinol Metab 2021; 106:1630-1636. [PMID: 33621325 DOI: 10.1210/clinem/dgab113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Indexed: 01/03/2023]
Abstract
CONTEXT Excess glucocorticoids impact fetal health. Maternal glucocorticoids peak in early morning. Fetoplacental 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) inactivates cortisol to cortisone, protecting the fetus from high glucocorticoids. However, time-specific alterations of human fetoplacental 11β-HSD2 have not been studied. OBJECTIVE We hypothesized that fetoplacental 11β-HSD2 activity shows time-specific alteration and acute affective or anxiety disorders impact fetoplacental 11β-HSD2 activity. METHODS In this observational study we investigated 78 pregnant European women undergoing amniocentesis (15.9 ± 0.9 weeks of gestation). Amniotic fluid was collected (8:00 to 16:30 hours) for analysis of fetoplacental 11β-HSD2 activity, using cortisol (F):cortisone (E) ratio in amniotic fluid, E/(E + F). Fetoplacental 11β-HSD2 rhythm and association with "acute affective or anxiety disorder" (patients with at least one of: a major depressive episode, specific phobia, panic disorder, generalized anxiety disorder, mixed anxiety and depressive disorder) and "acute anxiety disorder" (one of: panic disorder, generalized anxiety disorder, mixed anxiety, depressive disorder), assessed using Mini International Neuropsychiatric Interview, were investigated. RESULTS Activity of 11β-HSD2 correlated with time of amniocentesis, peaking in the morning (r = -0.398; P < 0.001) and increased with acute affective or anxiety disorder (mean [M] = 0.70 vs M = 0.74; P = 0.037) and acute anxiety disorder (M = 0.70 vs M = 0.75; P = 0.016). These associations remained significant when controlling for confounders. 11β-HSD2 activity correlated negatively with pre-pregnancy body mass index (r = -0.225; P = 0.047). CONCLUSION Our study indicates a time-specific alteration of fetoplacental 11β-HSD2 activity with peaking levels in the morning, demonstrating a mechanism of fetal protection from the morning maternal glucocorticoid surge.
Collapse
Affiliation(s)
- Eva Kathrin Lamadé
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, Mannheim, Germany
| | - Ferdinand Hendlmeier
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, Mannheim, Germany
| | - Stefan A Wudy
- Laboratory for Translational Hormone Analytics, Division of Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Feulgenstrasse 10-12, Giessen, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, Mannheim, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, Mannheim, Germany
| | - Michaela Coenen
- Institute for Medical Information Processing, Biometry and Epidemiology, Chair of Public Health and Health Services Research, LMU Munich, Elisabeth-Winterhalter-Weg 6, Munich, Germany
- Pettenkofer School of Public Health, Elisabeth-Winterhalter-Weg 6, 81377 Munich, Germany
| | - Maria Gilles
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, Mannheim, Germany
| | - Michael Deuschle
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, Mannheim, Germany
| |
Collapse
|
14
|
Arias A, Schander JA, Bariani MV, Correa F, Domínguez Rubio AP, Cella M, Cymeryng CB, Wolfson ML, Franchi AM, Aisemberg J. Dexamethasone-induced intrauterine growth restriction modulates expression of placental vascular growth factors and fetal and placental growth. Mol Hum Reprod 2021; 27:gaab006. [PMID: 33528567 DOI: 10.1093/molehr/gaab006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/20/2021] [Indexed: 02/02/2023] Open
Abstract
Prenatal exposure to glucocorticoids (GC) is a central topic of interest in medicine since GCs are essential for the maturation of fetal organs and intrauterine growth. Synthetic glucocorticoids, which are used in obstetric practice, exert beneficial effects on the fetus, but have also been reported to lead to intrauterine growth retardation (IUGR). In this study, a model of growth restriction in mice was established through maternal administration of dexamethasone during late gestation. We hypothesised that GC overexposure may adversely affect placental angiogenesis and fetal and placental growth. Female BALB/c mice were randomly assigned to control or dexamethasone treatment, either left to give birth or euthanised on days 15, 16, 17 and 18 of gestation followed by collection of maternal and fetal tissue. The IUGR rate increased to 100% in the dexamethasone group (8 mg/kg body weight on gestational days 14 and 15) and pups had clinical features of symmetrical IUGR at birth. Dexamethasone administration significantly decreased maternal body weight gain and serum corticosterone levels. Moreover, prenatal dexamethasone treatment not only induced fetal growth retardation but also decreased placental weight. In IUGR placentas, VEGFA protein levels and mRNA expression of VEGF receptors were reduced and NOS activity was lower. Maternal dexamethasone administration also reduced placental expression of the GC receptor, αGR. We demonstrated that maternal dexamethasone administration causes fetal and placental growth restriction. Furthermore, we propose that the growth retardation induced by prenatal GC overexposure may be caused, at least partially, by an altered placental angiogenic profile.
Collapse
Affiliation(s)
- A Arias
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J A Schander
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M V Bariani
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - F Correa
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - A P Domínguez Rubio
- Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN-UBA-CONICET), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M Cella
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - C B Cymeryng
- Laboratorio de Endocrinología Molecular, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M L Wolfson
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - A M Franchi
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J Aisemberg
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
15
|
Yao S, Lopez-Tello J, Sferruzzi-Perri AN. Developmental programming of the female reproductive system-a review. Biol Reprod 2020; 104:745-770. [PMID: 33354727 DOI: 10.1093/biolre/ioaa232] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Exposures to adverse conditions in utero can lead to permanent changes in the structure and function of key physiological systems in the developing fetus, increasing the risk of disease and premature aging in later postnatal life. When considering the systems that could be affected by an adverse gestational environment, the reproductive system of developing female offspring may be particularly important, as changes have the potential to alter both reproductive capacity of the first generation, as well as health of the second generation through changes in the oocyte. The aim of this review is to examine the impact of different adverse intrauterine conditions on the reproductive system of the female offspring. It focuses on the effects of exposure to maternal undernutrition, overnutrition/obesity, hypoxia, smoking, steroid excess, endocrine-disrupting chemicals, and pollutants during gestation and draws on data from human and animal studies to illuminate underlying mechanisms. The available data indeed indicate that adverse gestational environments alter the reproductive physiology of female offspring with consequences for future reproductive capacity. These alterations are mediated via programmed changes in the hypothalamic-pituitary-gonadal axis and the structure and function of reproductive tissues, particularly the ovaries. Reproductive programming may be observed as a change in the timing of puberty onset and menopause/reproductive decline, altered menstrual/estrous cycles, polycystic ovaries, and elevated risk of reproductive tissue cancers. These reproductive outcomes can affect the fertility and fecundity of the female offspring; however, further work is needed to better define the possible impact of these programmed changes on subsequent generations.
Collapse
Affiliation(s)
- Sijia Yao
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, UK
| | - Jorge Lopez-Tello
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
Barzilai R, Bronshtein M, Steinberg M, Weiner Z, Gover A. Small for gestational age: the familial perspective. J Matern Fetal Neonatal Med 2020; 35:3840-3844. [PMID: 33138687 DOI: 10.1080/14767058.2020.1841160] [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: 10/23/2022]
Abstract
BACKGROUND There are multiple etiologies for being born small for gestational age (SGA). However, extended familial data in idiopathic cases have been scarcely reported. OBJECTIVE Our aim was to explore the familial history of SGA newborns and describe the proportion and distribution of SGA in their parents and parental siblings. METHODS This was a retrospective study performed at an obstetrics clinic holding a detailed reliable electronic database. Between 2008 and 2017, data of 14,003 patients and 20,617 pregnancies were recorded. Parents of SGA infants were identified and extended familial history was obtained by questionnaires, including birth weights (BWs) and gestational age at birth of the parents and parents' siblings. SGA was defined as a BW below the 10th percentile. Proportions of maternal, paternal, and parental siblings' SGA were calculated. Chi-square test was performed to assess the relationship between SGA family member's gender and SGA infants' gender, and between the relative's gender and their family relationship to the infant. RESULTS About 2100 women had a history of a previous infant born SGA, however, after exclusion the final cohort comprised 926 women with a previous SGA infant. In 473 cases there was at least one other family member of the infant born SGA: father, mother, aunt, or uncle of the infant, representing a prevalence of 51% (473/926) of familial SGA. Out of familial SGA cases, maternal SGA was found in 55% (260/473), and paternal SGA was found in 28.1% (133/473). 27.6% had more than one SGA relative. Eighteen infants had both an SGA father and an SGA mother (3.8%). A history of an SGA aunt or uncle was found in 44% (209/473) of familial SGA cases, which was 22.5% (209/926) of the entire cohort. Parental sibling SGA occurred almost twice in mother's siblings as compared to father's siblings. Chi-square test revealed no association between the SGA relative's gender and their family relationship to the infant. There was no association between the SGA infant's gender and the SGA relative's gender. CONCLUSIONS A family history of SGA is common in SGA infants, and occurs most often in mothers. This study found 22% SGA in parental siblings, in maternal siblings more than paternal siblings, supporting the possibility of a genetic component in SGA trait transmission. In clinical practice, when counseling parents with a growth-restricted fetus from an unknown etiology, extended familial birthweight history should be obtained and taken into account, which may be helpful in reducing parental anxiety.
Collapse
Affiliation(s)
- Roni Barzilai
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Moshe Bronshtein
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Maya Steinberg
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Zeev Weiner
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Ayala Gover
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Neonatal Intensive Care Unit, Lady Davis Carmel Medical Center, Haifa, Israel
| |
Collapse
|
17
|
Ruiz D, Padmanabhan V, Sargis RM. Stress, Sex, and Sugar: Glucocorticoids and Sex-Steroid Crosstalk in the Sex-Specific Misprogramming of Metabolism. J Endocr Soc 2020; 4:bvaa087. [PMID: 32734132 PMCID: PMC7382384 DOI: 10.1210/jendso/bvaa087] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
Early-life exposures to environmental insults can misprogram development and increase metabolic disease risk in a sex-dependent manner by mechanisms that remain poorly characterized. Modifiable factors of increasing public health relevance, such as diet, psychological stress, and endocrine-disrupting chemicals, can affect glucocorticoid receptor signaling during gestation and lead to sex-specific postnatal metabolic derangements. Evidence from humans and animal studies indicate that glucocorticoids crosstalk with sex steroids by several mechanisms in multiple tissues and can affect sex-steroid-dependent developmental processes. Nonetheless, glucocorticoid sex-steroid crosstalk has not been considered in the glucocorticoid-induced misprogramming of metabolism. Herein we review what is known about the mechanisms by which glucocorticoids crosstalk with estrogen, androgen, and progestogen action. We propose that glucocorticoid sex-steroid crosstalk is an understudied mechanism of action that requires consideration when examining the developmental misprogramming of metabolism, especially when assessing sex-specific outcomes.
Collapse
Affiliation(s)
- Daniel Ruiz
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois.,Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| | | | - Robert M Sargis
- Division of Endocrinology, Diabetes, and Metabolism; Department of Medicine; University of Illinois at Chicago, Chicago, Illinois.,Chicago Center for Health and Environment, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
18
|
Castelli V, Lavanco G, Brancato A, Plescia F. Targeting the Stress System During Gestation: Is Early Handling a Protective Strategy for the Offspring? Front Behav Neurosci 2020; 14:9. [PMID: 32082129 PMCID: PMC7006220 DOI: 10.3389/fnbeh.2020.00009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/15/2020] [Indexed: 12/28/2022] Open
Abstract
The perinatal window is a critical developmental time when abnormal gestational stimuli may alter the development of the stress system that, in turn, influences behavioral and physiological responses in the newborns. Individual differences in stress reactivity are also determined by variations in maternal care, resulting from environmental manipulations. Despite glucocorticoids are the primary programming factor for the offspring's stress response, therapeutic corticosteroids are commonly used during late gestation to prevent preterm negative outcomes, exposing the offspring to potentially aberrant stress reactivity later in life. Thus, in this study, we investigated the consequences of one daily s.c. injection of corticosterone (25 mg/kg), from gestational day (GD) 14-16, and its interaction with offspring early handling, consisting in a brief 15-min maternal separation until weaning, on: (i) maternal behavior; and (ii) behavioral reactivity, emotional state and depressive-like behavior in the adolescent offspring. Corticosterone plasma levels, under non-shock- and shock-induced conditions, were also assessed. Our results show that gestational exposure to corticosterone was associated with diminished maternal care, impaired behavioral reactivity, increased emotional state and depressive-like behavior in the offspring, associated with an aberrant corticosterone response. The early handling procedure, which resulted in increased maternal care, was able to counteract the detrimental effects induced by gestational corticosterone exposure both in the behavioral- and neurochemical parameters examined. These findings highlight the potentially detrimental consequences of targeting the stress system during pregnancy as a vulnerability factor for the occurrence of emotional and affective distress in the adolescent offspring. Maternal extra-care proves to be a protective strategy that confers resiliency and restores homeostasis.
Collapse
Affiliation(s)
- Valentina Castelli
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Gianluca Lavanco
- INSERM U1215, Neuro Centre Magendie, Bordeaux, France.,University of Bordeaux, Bordeaux, France.,Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Anna Brancato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "Giuseppe D'Alessandro", University of Palermo, Palermo, Italy
| | - Fulvio Plescia
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "Giuseppe D'Alessandro", University of Palermo, Palermo, Italy
| |
Collapse
|
19
|
Sand SA, Ernst A, Lunddorf LLH, Brix N, Gaml-Sørensen A, Ramlau-Hansen CH. In Utero Exposure to Glucocorticoids and Pubertal Timing in Sons and Daughters. Sci Rep 2019; 9:20374. [PMID: 31889153 PMCID: PMC6937234 DOI: 10.1038/s41598-019-56917-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/17/2019] [Indexed: 11/29/2022] Open
Abstract
Early pubertal timing has been associated with adult diseases, and identifying preventable causes is of importance. In utero exposure to exogenous glucocorticoids, has been associated with changes in the reproductive hormonal axes in the children, which may influence pubertal timing. Exogenous glucocorticoids can be indicated for diseases such as asthma, allergy, skin diseases, as well as muscle and joint diseases. The aim was to explore the association between in utero exposure to glucocorticoids and pubertal timing in the children. This population-based study was conducted in the Puberty Cohort including 15,819 children, which is a sub-cohort of the Danish National Birth Cohort. Information on maternal glucocorticoid treatment was collected through interviews during pregnancy. Information on pubertal timing was obtained by questionnaires every 6 months throughout puberty, including Tanner Stages, axillary hair, acne, voice break, first ejaculation and menarche. The potential impact of confounding by indication was explored by stratifying on indication and treatment status. Overall, 6.8% of the children were exposed to glucocorticoids in utero. Exposure to glucocorticoids in utero was not associated with earlier puberty for neither boys nor girls with combined estimates of 0.4 months (95% CI: -1.5; 2.2) and -0.7 months (95% CI: -2.5; 1.2).
Collapse
Affiliation(s)
- Sofie Aagaard Sand
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark.
| | - Andreas Ernst
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Nis Brix
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark
| | - Anne Gaml-Sørensen
- Department of Public Health, Research Unit for Epidemiology, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
20
|
Bridge-Comer PE, Vickers MH, Reynolds CM. Preclinical Models of Altered Early Life Nutrition and Development of Reproductive Disorders in Female Offspring. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:59-87. [PMID: 30919332 DOI: 10.1007/978-3-030-12668-1_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Early epidemiology studies in humans have and continue to offer valuable insight into the Developmental Origins of Health and Disease (DOHaD) hypothesis, which emphasises the importance of early-life nutritional and environmental changes on the increased risk of metabolic and reproductive disease in later life. Human studies are limited and constrained by a range of factors which do not apply to preclinical research. Animal models therefore offer a unique opportunity to fully investigate the mechanisms associated with developmental programming, helping to elucidate the developmental processes which influence reproductive diseases, and highlight potential biomarkers which can be translated back to the human condition. This review covers the use and limitations of a number of animal models frequently utilised in developmental programming investigations, with an emphasis on dietary manipulations which can lead to reproductive dysfunction in offspring.
Collapse
Affiliation(s)
| | - Mark H Vickers
- The Liggins Institute, University of Auckland, Auckland, New Zealand.
| | - Clare M Reynolds
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
21
|
Ristić N, Nestorović N, Manojlović-Stojanoski M, Trifunović S, Ajdžanović V, Filipović B, Pendovski L, Milošević V. Adverse effect of dexamethasone on development of the fetal rat ovary. Fundam Clin Pharmacol 2018; 33:199-207. [PMID: 30216532 DOI: 10.1111/fcp.12415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 08/06/2018] [Accepted: 09/10/2018] [Indexed: 11/30/2022]
Abstract
Dexamethasone (Dx) is often used in obstetric practice to promote fetal lung maturation and to prevent respiratory distress syndrome when the risk of preterm delivery persists. This therapy enables survival of the newborn, but also is associated with deleterious effects on the offspring, such as reproductive disorders. The aim of this study was to determine specifically whether prenatal exposure to Dx disturbs the physiological balance between proliferation and apoptosis of germinative cells (GC) in the ovary of 19- and 21-day-old fetuses and thus induces developmental programming of the female reproductive system. Pregnant Wistar rats (n = 10/group), separated into control (vehicle) and Dx-treated (0.5 mg/kg body mass) groups, received injections on gestational days 16, 17, and 18. Exposure to Dx lowered the volume of the fetal ovary by 30% (P < 0.05) in 21-day-old fetuses, as well as the total number of GC in the ovary by 21% (P < 0.05). When compared to the controls, in Dx-exposed fetuses, the total number of PCNA-positive GC was 27% lower at 19 days and 71% lower at 21 days old (P < 0.05), while total numbers of caspase-3-positive GC were 2.3-fold and 34% higher, respectively (P < 0.05). Our results demonstrate that prenatal exposure to Dx diminished proliferation but increased the rate of germinative cell apoptosis, with consequently reduced total germinative cell number and ovary volume. Impairment of fetal oogenesis and fewer GC in the fetal ovary compromise the oogonial stock and thus may constitute a risk of female fertility.
Collapse
Affiliation(s)
- Nataša Ristić
- Department of Cytology, Institute for Biological Research"Siniša Stanković", University of Belgrade, 11060, Belgrade, Serbia
| | - Nataša Nestorović
- Department of Cytology, Institute for Biological Research"Siniša Stanković", University of Belgrade, 11060, Belgrade, Serbia
| | - Milica Manojlović-Stojanoski
- Department of Cytology, Institute for Biological Research"Siniša Stanković", University of Belgrade, 11060, Belgrade, Serbia
| | - Svetlana Trifunović
- Department of Cytology, Institute for Biological Research"Siniša Stanković", University of Belgrade, 11060, Belgrade, Serbia
| | - Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research"Siniša Stanković", University of Belgrade, 11060, Belgrade, Serbia
| | - Branko Filipović
- Department of Cytology, Institute for Biological Research"Siniša Stanković", University of Belgrade, 11060, Belgrade, Serbia
| | | | - Verica Milošević
- Department of Cytology, Institute for Biological Research"Siniša Stanković", University of Belgrade, 11060, Belgrade, Serbia
| |
Collapse
|
22
|
Abolins-Abols M, Hanauer RE, Rosvall KA, Peterson MP, Ketterson ED. The effect of chronic and acute stressors, and their interaction, on testes function: an experimental test during testicular recrudescence. J Exp Biol 2018; 221:jeb180869. [PMID: 29997161 PMCID: PMC6919650 DOI: 10.1242/jeb.180869] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/29/2018] [Indexed: 01/21/2023]
Abstract
Organisms are expected to invest less in reproduction in response to a stressor, but theory predicts that this effect should depend on the frequency and duration of stressors in the environment. Here, we investigated how an acute stressor affected testes function in a songbird, and how chronic stressors influenced the acute stress response. We exposed male dark-eyed juncos (Junco hyemalis) either to chronic or minimal (control) disturbance during testicular recrudescence, after which we measured baseline testosterone, testosterone after an acute handling stressor, and capacity to produce testosterone after hormonal stimulation. In a 2×2 design, we then killed males from the two chronic treatment groups either immediately or after an acute stressor to investigate the effect of long- and short-term stressors on the testicular transcriptome. We found that chronically disturbed birds had marginally lower baseline testosterone. The acute stressor suppressed testosterone in control birds, but not in the chronic disturbance group. The ability to elevate testosterone did not differ between the chronic treatments. Surprisingly, chronic disturbance had a weak effect on the testicular transcriptome, and did not affect the transcriptomic response to the acute stressor. The acute stressor, on the other hand, upregulated the cellular stress response and affected expression of genes associated with hormonal stress response. Overall, we show that testicular function is sensitive to acute stressors but surprisingly robust to long-term stressors, and that chronic disturbance attenuates the decrease in testosterone in response to an acute stressor.
Collapse
Affiliation(s)
- Mikus Abolins-Abols
- 505 S Goodwin Ave, Department of Animal Biology, School of Integrative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- 1001 E. 3rd St., Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Rachel E Hanauer
- 1001 E. 3rd St., Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Kimberly A Rosvall
- 1001 E. 3rd St., Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Mark P Peterson
- 1800 Technology Dr. NE, Life-Science Innovations, Willmar, MN 56201, USA
| | - Ellen D Ketterson
- 1001 E. 3rd St., Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
23
|
Faykoo-Martinez M, Monks DA, Zovkic IB, Holmes MM. Sex- and brain region-specific patterns of gene expression associated with socially-mediated puberty in a eusocial mammal. PLoS One 2018; 13:e0193417. [PMID: 29474488 PMCID: PMC5825099 DOI: 10.1371/journal.pone.0193417] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022] Open
Abstract
The social environment can alter pubertal timing through neuroendocrine mechanisms that are not fully understood; it is thought that stress hormones (e.g., glucocorticoids or corticotropin-releasing hormone) influence the hypothalamic-pituitary-gonadal axis to inhibit puberty. Here, we use the eusocial naked mole-rat, a unique species in which social interactions in a colony (i.e. dominance of a breeding female) suppress puberty in subordinate animals. Removing subordinate naked mole-rats from this social context initiates puberty, allowing for experimental control of pubertal timing. The present study quantified gene expression for reproduction- and stress-relevant genes acting upstream of gonadotropin-releasing hormone in brain regions with reproductive and social functions in pre-pubertal, post-pubertal, and opposite sex-paired animals (which are in various stages of pubertal transition). Results indicate sex differences in patterns of neural gene expression. Known functions of genes in brain suggest stress as a key contributing factor in regulating male pubertal delay. Network analysis implicates neurokinin B (Tac3) in the arcuate nucleus of the hypothalamus as a key node in this pathway. Results also suggest an unappreciated role for the nucleus accumbens in regulating puberty.
Collapse
Affiliation(s)
| | - D. Ashley Monks
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Iva B. Zovkic
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Melissa M. Holmes
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Prenatal stress accelerates offspring growth to compensate for reduced maternal investment across mammals. Proc Natl Acad Sci U S A 2017; 114:E10658-E10666. [PMID: 29180423 DOI: 10.1073/pnas.1707152114] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Across mammals, prenatal maternal stress (PREMS) affects many aspects of offspring development, including offspring growth. However, how PREMS translates to offspring growth is inconsistent, even within species. To explain the full range of reported effects of prenatal adversity on offspring growth, we propose an integrative hypothesis: developmental constraints and a counteracting adaptive growth plasticity work in opposition to drive PREMS effects on growth. Mothers experiencing adversity reduce maternal investment leading to stunted growth (developmental constraints). Concomitantly, the pace of offspring life history is recalibrated to partly compensate for these developmental constraints (adaptive growth plasticity). Moreover, the relative importance of each process changes across ontogeny with increasing offspring independence. Thus, offspring exposed to PREMS may grow at the same rate as controls during gestation and lactation, but faster after weaning when direct maternal investment has ceased. We tested these predictions with a comparative analysis on the outcomes of 719 studies across 21 mammal species. First, the observed growth changes in response to PREMS varied across offspring developmental periods as predicted. We argue that the observed growth acceleration after weaning is not "catch-up growth," because offspring that were small for age grew slower. Second, only PREMS exposure early during gestation produced adaptive growth plasticity. Our results suggest that PREMS effects benefit the mother's future reproduction and at the same time accelerate offspring growth and possibly maturation and reproductive rate. In this sense, PREMS effects on offspring growth allow mother and offspring to make the best of a bad start.
Collapse
|
25
|
Abstract
Prenatal insults, such as maternal stress, are associated with an increased neurodevelopmental disease risk and impact males significantly more than females, including increased rates of autism, mental retardation, stuttering, dyslexia, and attention deficit/hyperactivity disorder (ADHD). Sex differences in the placenta, which begin with sex chromosomes, are likely to produce sex-specific transplacental signals to the developing brain. Our studies and others have identified X-linked genes that are expressed at higher levels in the female placenta. Through a genome-wide screen after maternal stress in mice, we identified the X-linked gene O-linked N-acetylglucosamine transferase (OGT) and demonstrated its causality in neurodevelopmental programming producing a male-specific stress phenotype. Elucidating the sex-specific molecular mechanisms involved in transplacental signals that impact brain development is key to understanding the sex bias in neurodevelopmental disorders and is expected to yield novel insight into disease risk and resilience.
Collapse
Affiliation(s)
- Tracy L Bale
- Department of Biomedical Sciences, School of Veterinary Medicine and Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Borges CS, Pacheco TL, Guerra MT, Barros AL, Silva PV, Missassi G, da Silva KP, Anselmo-Franci JA, Pupo AS, Kempinas WDG. Reproductive disorders in female rats after prenatal exposure to betamethasone. J Appl Toxicol 2017; 37:1065-1072. [PMID: 28326570 DOI: 10.1002/jat.3457] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 12/13/2022]
Abstract
Betamethasone is the drug of choice for antenatal treatment, promoting fetal lung maturation and decreasing mortality. Previous studies in rats reported male programming and alteration in sperm parameters and sexual behavior following intrauterine betamethasone exposure. The impact on the female reproductive development is not known. In this study, rat female offspring was assessed for sexual development, morphophysiology of the reproductive tract and fertility after maternal exposure to 0.1 mg kg-1 of betamethasone or vehicle on gestational days 12, 13, 18 and 19. The treatment promoted reduction of litter weight on postnatal day 1, morphological masculinization in females, delay in the age of puberty onset, reduction in estrus number, increase in estrous cycle length and increase in luteinizing hormone serum levels and uterus weight. The females from the betamethasone group showed an increase of myometrial uterine area and decrease in endometrial uterine area. These animals also performed less lordosis during the sexual behavior test and showed impaired reproductive performance. The uterus showed higher contraction in the treated group as shown by a pharmacological assay. In conclusion, prenatal betamethasone exposure in rats promoted female masculinization, altered sexual development and reproductive parameters. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Cibele S Borges
- Departments of Morphology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| | - Tainá L Pacheco
- Departments of Morphology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| | - Marina T Guerra
- Departments of Morphology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| | - Aline L Barros
- Departments of Morphology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| | - Patricia V Silva
- Departments of Morphology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| | - Gabriela Missassi
- Departments of Morphology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| | - Katiussia Pinho da Silva
- Departments of Pharmacology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| | - Janete A Anselmo-Franci
- Department of Morphology, Stomatology and Physiology, Dental School of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil
| | - André S Pupo
- Departments of Pharmacology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| | - Wilma De G Kempinas
- Departments of Morphology, Institute of Biosciences, Univ Estadual Paulista-UNESP, Distrito de Rubião Junior s/n°, 18618-970, Botucatu, SP, Brazil
| |
Collapse
|
27
|
Childhood body mass index at 5.5 years mediates the effect of prenatal maternal stress on daughters’ age at menarche: Project Ice Storm. J Dev Orig Health Dis 2016; 8:168-177. [DOI: 10.1017/s2040174416000726] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Early pubertal timing is known to put women at greater risk for adverse physiological and psychological health outcomes. Of the factors that influence girls’ pubertal timing, stress experienced during childhood has been found to advance age at menarche (AAM). However, it is not known if stress experienced by mothers during or in the months before conception can be similarly associated with earlier pubertal timing. Prenatal maternal stress (PNMS) is associated with metabolic changes, such as increased childhood adiposity and risk of obesity, that have been associated with earlier menarchal age. Using a prospective longitudinal design, the present study tested whether PNMS induced by a natural disaster is either directly associated with earlier AAM, or whether there is an indirect association mediated through increased girls’ body mass index (BMI) during childhood. A total of 31 girls, whose mothers were exposed to the Quebec’s January 1998 ice storm during pregnancy were followed from 6 months to 5 1/2 to 5.5 years of age. Mother’s stress was measured within 6 months of the storm. BMI was measured at 5.5 years, and AAM was assessed through teen’s self-report at 13.5 and 15.5 years of age. Results revealed that greater BMI at 5.5 years mediated the effect of PNMS on decreasing AAM [B=−0.059, 95% confidence intervals (−0.18, −0.0035)]. The present study is the first to demonstrate that maternal experience of stressful conditions during pregnancy reduces AAM in the offspring through its effects on childhood BMI. Future research should consider the impact of AAM on other measures of reproductive ability.
Collapse
|
28
|
A review of fundamental principles for animal models of DOHaD research: an Australian perspective. J Dev Orig Health Dis 2016; 7:449-472. [DOI: 10.1017/s2040174416000477] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Epidemiology formed the basis of ‘the Barker hypothesis’, the concept of ‘developmental programming’ and today’s discipline of the Developmental Origins of Health and Disease (DOHaD). Animal experimentation provided proof of the underlying concepts, and continues to generate knowledge of underlying mechanisms. Interventions in humans, based on DOHaD principles, will be informed by experiments in animals. As knowledge in this discipline has accumulated, from studies of humans and other animals, the complexity of interactions between genome, environment and epigenetics, has been revealed. The vast nature of programming stimuli and breadth of effects is becoming known. As a result of our accumulating knowledge we now appreciate the impact of many variables that contribute to programmed outcomes. To guide further animal research in this field, the Australia and New Zealand DOHaD society (ANZ DOHaD) Animals Models of DOHaD Research Working Group convened at the 2nd Annual ANZ DOHaD Congress in Melbourne, Australia in April 2015. This review summarizes the contributions of animal research to the understanding of DOHaD, and makes recommendations for the design and conduct of animal experiments to maximize relevance, reproducibility and translation of knowledge into improving health and well-being.
Collapse
|
29
|
Carbenoxolone exposure during late gestation in rats alters placental expressions of p53 and estrogen receptors. Eur J Pharmacol 2016; 791:675-685. [PMID: 27693517 DOI: 10.1016/j.ejphar.2016.09.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/20/2016] [Accepted: 09/27/2016] [Indexed: 12/31/2022]
Abstract
Gestational carbenoxolone exposure inhibits placental 11β-hydroxysteroid dehydrogenase (11β-HSD), the physiological barrier for glucocorticoids, which increases fetal exposure to glucocorticoids and induces intrauterine growth restriction (IUGR). We hypothesized that carbenoxolone exposure influences the expression of placental estrogen receptors-α and β (ERα & ERβ) and p53 leading to inhibited fetal and placental growth. Pregnant Sprague-Dawley rats were injected twice daily with either carbenoxolone (10mg/kg; s.c.) or vehicle (control group) from gestational days (dg) 12 onwards. Maternal blood and placentas were collected on 16 dg, 19 dg and 21 dg. The expression of ERα, ERβ and p53 were studied in placental basal and labyrinth zones by RT-PCR, Western blotting and immunohistochemistry. Carbenoxolone did not affect placental and fetal body weights, but ELISA showed decreased estradiol levels on 19 dg and 21 dg, and increased maternal luteinizing hormone levels on all dg. The follicle stimulating hormone levels decreased on 16 dg and 19 dg, and increased on 21 dg. Carbenoxolone decreased ERα mRNA levels on 16 dg in both zones and its protein level on 19 dg in the labyrinth zone. However, carbenoxolone increased ERβ mRNA levels on 19 dg and 21 dg and protein levels on 16 dg and 19 dg in the labyrinth zone. The p53 mRNA levels increased on all dg, but its protein levels increased on 21 dg in both zones. In conclusion, carbenoxolone exposure changes placental p53, ERα, ERβ expression in favor of cell death but these changes do not induce IUGR in rats.
Collapse
|
30
|
Huffman MC, Santo JB, French JA. Prenatal androgen exposure and parental care interact to influence timing of reproductive maturation in marmosets. Am J Primatol 2016; 79:1-12. [DOI: 10.1002/ajp.22588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/29/2016] [Accepted: 07/18/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Michelle C. Huffman
- Callitrichid Research Center; University of Nebraska; Omaha Nebraska
- Department of Psychology; University of Nebraska; Omaha Nebraska
| | | | - Jeffrey A. French
- Callitrichid Research Center; University of Nebraska; Omaha Nebraska
- Department of Psychology; University of Nebraska; Omaha Nebraska
- Department of Biology; University of Nebraska; Omaha Nebraska
| |
Collapse
|
31
|
Ramalhosa F, Soares-Cunha C, Seixal RM, Sousa N, Carvalho AF. The Impact of Prenatal Exposure to Dexamethasone on Gastrointestinal Function in Rats. PLoS One 2016; 11:e0161750. [PMID: 27584049 PMCID: PMC5008745 DOI: 10.1371/journal.pone.0161750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 08/11/2016] [Indexed: 01/23/2023] Open
Abstract
Antenatal treatment with synthetic glucocorticoids is commonly used in pregnant women at risk of preterm delivery to accelerate tissue maturation. Exposure to glucocorticoids during development has been hypothesized to underlie different functional gastrointestinal (GI) and motility disorders. Herein, we investigated the impact of in utero exposure to synthetic glucocorticoids (iuGC) on GI function of adult rats. Wistar male rats, born from pregnant dams treated with dexamethasone (DEX), were studied at different ages. Length, histologic analysis, proliferation and apoptosis assays, GI transit, permeability and serotonin (5-HT) content of GI tract were measured. iuGC treatment decreased small intestine size and decreased gut transit. However, iuGC had no impact on intestinal permeability. iuGC differentially impacts the structure and function of the GI tract, which leads to long-lasting alterations in the small intestine that may predispose subjects prone to disorders of the GI tract.
Collapse
Affiliation(s)
- Fátima Ramalhosa
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui Miguel Seixal
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Franky Carvalho
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
- General Surgery Department, Hospital of Braga, Braga, Portugal
| |
Collapse
|
32
|
Maternal treatment with dexamethasone during gestation alters sexual development markers in the F1 and F2 male offspring of Wistar rats. J Dev Orig Health Dis 2016; 8:101-112. [DOI: 10.1017/s2040174416000453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Maternal treatment with dexamethasone (Dex) in threatening preterm delivery alters activities at the hypothalamic–pituitary–adrenal axis in the offspring. This alteration may interfere with reproductive function. The impact of gestational Dex exposure on male reproductive function of the offspring was investigated. A total of 25 pregnant rats randomly assigned to five groups (n=5) were treated with normal saline (control), Dex (100 μg/kg/day sc) during gestation days (GD) 1–7, 8–14, 15–21 and 1–21, respectively. Birth weight, anogenital distance (AGD), pubertal age, sperm parameters, hormonal profile and histopathology of testis and epididymis were determined in the F1 and F2 offspring. Results showed a significant increase (P<0.05) in pubertal age, serum corticosterone and gonadotropin-releasing hormone (GnRH) levels in the male offspring of DexGD 15–21 and 1–21 groups and a significant decrease (P<0.05) in serum testosterone, luteinizing hormone, birth weight and AGD at birth in the male F1 offspring. In the F2 offspring, there was a significant reduction (P<0.05) in serum corticosterone, testosterone, follicle-stimulating hormone and GnRH when compared with the control. Dex treatment at GD 15–21 and 1–21 significantly reduced (P<0.05) sperm motility and normal morphology in the F1 and F2 offspring. Maternal Dex treatment in rats during late gestation may disrupt sexual development markers in the F1 and F2 male offspring.
Collapse
|
33
|
Camille Melón L, Maguire J. GABAergic regulation of the HPA and HPG axes and the impact of stress on reproductive function. J Steroid Biochem Mol Biol 2016; 160:196-203. [PMID: 26690789 PMCID: PMC4861672 DOI: 10.1016/j.jsbmb.2015.11.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 11/16/2015] [Accepted: 11/26/2015] [Indexed: 11/25/2022]
Abstract
The hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-gonadal (HPG) axes are regulated by GABAergic signaling at the level of corticotropin-releasing hormone (CRH) and gonadotropin-releasing hormone (GnRH) neurons, respectively. Under basal conditions, activity of CRH and GnRH neurons are controlled in part by both phasic and tonic GABAergic inhibition, mediated by synaptic and extrasynaptic GABAA receptors (GABAARs), respectively. For CRH neurons, this tonic GABAergic inhibition is mediated by extrasynaptic, δ subunit-containing GABAARs. Similarly, a THIP-sensitive tonic GABAergic current has been shown to regulate GnRH neurons, suggesting a role for δ subunit-containing GABAARs; however, this remains to be explicitly demonstrated. GABAARs incorporating the δ subunit confer neurosteroid sensitivity, suggesting a potential role for neurosteroid modulation in the regulation of the HPA and HPG axes. Thus, stress-derived neurosteroids may contribute to the impact of stress on reproductive function. Interestingly, excitatory actions of GABA have been demonstrated in both CRH neurons at the apex of control of the HPA axis and in GnRH neurons which mediate the HPG axis, adding to the complexity for the role of GABAergic signaling in the regulation of these systems. Here we review the effects that stress has on GnRH neurons and HPG axis function alongside evidence supporting GABAARs as a major interface between the stress and reproductive axes.
Collapse
Affiliation(s)
- Laverne Camille Melón
- Tufts University School of Medicine, Department of Neuroscience, Boston, MA 02111, United States
| | - Jamie Maguire
- Tufts University School of Medicine, Department of Neuroscience, Boston, MA 02111, United States.
| |
Collapse
|
34
|
Luo Q, Li W, Li M, Zhang X, Zhang H. Leptin/leptinR-kisspeptin/kiss1r-GnRH pathway reacting to regulate puberty onset during negative energy balance. Life Sci 2016; 153:207-12. [DOI: 10.1016/j.lfs.2016.03.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 03/22/2016] [Accepted: 03/24/2016] [Indexed: 11/30/2022]
|
35
|
Evans NP, Bellingham M, Robinson JE. Prenatal programming of neuroendocrine reproductive function. Theriogenology 2016; 86:340-8. [PMID: 27142489 DOI: 10.1016/j.theriogenology.2016.04.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/24/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022]
Abstract
It is now well recognized that the gestational environment can have long-lasting effects not only on the life span and health span of an individual but also, through potential epigenetic changes, on future generations. This article reviews the "prenatal programming" of the neuroendocrine systems that regulate reproduction, with a specific focus on the lessons learned using ovine models. The review examines the critical roles played by steroids in normal reproductive development before considering the effects of prenatal exposure to exogenous steroid hormones including androgens and estrogens, the effects of maternal nutrition and stress during gestation, and the effects of exogenous chemicals such as alcohol and environment chemicals. In so doing, it becomes evident that, to maximize fitness, the regulation of reproduction has evolved to be responsive to many different internal and external cues and that the GnRH neurosecretory system expresses a degree of plasticity throughout life. During fetal life, however, the system is particularly sensitive to change and at this time, the GnRH neurosecretory system can be "shaped" both to achieve normal sexually differentiated function but also in ways that may adversely affect or even prevent "normal function". The exact mechanisms through which these programmed changes are brought about remain largely uncharacterized but are likely to differ depending on the factor, the timing of exposure to that factor, and the species. It would appear, however, that some afferent systems to the GnRH neurons such as kisspeptin, may be critical in this regard as it would appear to be sensitive to a wide variety of factors that can program reproductive function. Finally, it has been noted that the prenatal programming of neuroendocrine reproductive function can be associated with epigenetic changes, which would suggest that in addition to direct effects on the exposed offspring, prenatal programming could have transgenerational effects on reproductive potential.
Collapse
Affiliation(s)
- Neil P Evans
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Michelle Bellingham
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jane E Robinson
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
36
|
Khor YM, Soga T, Parhar IS. Early-life stress changes expression of GnRH and kisspeptin genes and DNA methylation of GnRH3 promoter in the adult zebrafish brain. Gen Comp Endocrinol 2016; 227:84-93. [PMID: 26686318 DOI: 10.1016/j.ygcen.2015.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/25/2015] [Accepted: 12/04/2015] [Indexed: 01/05/2023]
Abstract
Early-life stress can cause long-term effects in the adulthood such as alterations in behaviour, brain functions and reproduction. DNA methylation is a mechanism of epigenetic change caused by early-life stress. Dexamethasone (DEX) was administered to zebrafish larvae to study its effect on reproductive dysfunction. The level of GnRH2, GnRH3, Kiss1 and Kiss2 mRNAs were measured between different doses of DEX treatment groups in adult zebrafish. Kiss1 and GnRH2 expression were increased in the 200mg/L DEX treated while Kiss2 and GnRH3 mRNA levels were up-regulated in the 2mg/L DEX-treated zebrafish. The up-regulation may be related to programming effect of DEX in the zebrafish larvae, causing overcompensation mechanism to increase the mRNA levels. Furthermore, DEX treatment caused negative impact on the development and maturation of the testes, in particular spermatogenesis. Therefore, immature gonadal development may cause positive feedback by increasing GnRH and Kiss. This indicates that DEX can alter the regulation of GnRH2, GnRH3, Kiss1 and Kiss2 in adult zebrafish, which affects maturation of gonads. Computer analysis of 1.5 kb region upstream of the 5' UTR of Kiss1, Kiss2, GnRH2 and GnRH3 promoter showed that there are putative binding sites of glucocorticoid response element and transcription factors involved in stress response. GnRH3 promoter analysed from pre-optic area, ventral telencephalon and ventral olfactory bulb showed higher methylation at CpG residues located on -1410, -1377 and -1355 between control and 2mg/L DEX-treated groups. Hence, early-life DEX treatment can alter methylation of GnRH3 gene promoter, which subsequently affects gene regulation and reproductive functions.
Collapse
Affiliation(s)
- Yee Min Khor
- Brain Research Institute, School of Medicine and Health Sciences, Monash University, Malaysia
| | - Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University, Malaysia.
| | - Ishwar S Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University, Malaysia
| |
Collapse
|
37
|
Jeje SO, Akindele OO, Balogun ME, Raji Y. Maternal treatment with dexamethasone during lactation delays male puberty and disrupts reproductive functions via hypothalamic-pituitary-gonadal axis alterations. ACTA ACUST UNITED AC 2016; 23:43-9. [PMID: 26774541 DOI: 10.1016/j.pathophys.2015.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/25/2015] [Accepted: 12/26/2015] [Indexed: 01/09/2023]
Abstract
The effects of maternal treatment with dexamethasone during lactation on pubertal timing, serum hormonal profile and sperm indices in the male offspring were assessed. Twenty lactating dams were divided into 4 groups (n=5). Group 1 was administered subcutaneously 0.02ml/100g/day normal saline at lactation days 1-21. Groups 2-4 were administered subcutaneously 100μg/kg/day dexamethasone (Dex) at lactation days 1-7, 1-14, and 1-21 respectively. Results showed that there was significant reduction in serum testosterone in the DexLD 1-7 (p<0.05), DexLD 1-14 (p<0.01) and DexLD 1-21 (p<0.001) relative to control. In addition there was a significant reduction in serum FSH and LH in the DexLD 1-7 (p<0.01), DexLD 1-14 (p<0.001) and DexLD 1-21 (p<0.001) when compared with the control. Treatment with dexamethasone during lactation significantly increased the days of preputial separation in the DexLD 1-7 (p<0.05), DexLD 1-14 (p<0.05) and DexLD 1-21 (p<0.001) relative to control. Maternal treatment with dexamethasone throughout lactation period also significantly reduced sperm counts (p<0.001), motility (p<0.01) and increased percentage abnormal sperm (p<0.001) in the offspring when compared with the control. In conclusion, maternal treatment with dexamethasone during lactation may induce delayed puberty and disrupt reproductive functions by altering activities at hypothalamic-pituitary-gonadal axis in the male offspring.
Collapse
Affiliation(s)
- S O Jeje
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria; Department of Human Physiology, Cross River University of Technology, Okuku Campus, Cross River State, Nigeria.
| | - O O Akindele
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - M E Balogun
- Department of Physiology, Faculty of Medicine, College of Health Sciences, Ebonyi State University, Abakaliki, Nigeria
| | - Y Raji
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
38
|
Lim WL, Idris MM, Kevin FS, Soga T, Parhar IS. Maternal Dexamethasone Exposure Alters Synaptic Inputs to Gonadotropin-Releasing Hormone Neurons in the Early Postnatal Rat. Front Endocrinol (Lausanne) 2016; 7:117. [PMID: 27630615 PMCID: PMC5005956 DOI: 10.3389/fendo.2016.00117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 08/11/2016] [Indexed: 11/18/2022] Open
Abstract
Maternal dexamethasone [(DEX); a glucocorticoid receptor agonist] exposure delays pubertal onset and alters reproductive behavior in the adult offspring. However, little is known whether maternal DEX exposure affects the offspring's reproductive function by disrupting the gonadotropin-releasing hormone (GnRH) neuronal function in the brain. Therefore, this study determined the exposure of maternal DEX on the GnRH neuronal spine development and synaptic cluster inputs to GnRH neurons using transgenic rats expressing enhanced green fluorescent protein (EGFP) under the control of GnRH promoter. Pregnant females were administered with DEX (0.1 mg/kg) or vehicle (VEH, water) daily during gestation day 13-20. Confocal imaging was used to examine the spine density of EGFP-GnRH neurons by three-dimensional rendering and synaptic cluster inputs to EGFP-GnRH neurons by synapsin I immunohistochemistry on postnatal day 0 (P0) males. The spine morphology and number on GnRH neurons did not change between the P0 males following maternal DEX and VEH treatment. The number of synaptic clusters within the organum vasculosum of the lamina terminalis (OVLT) was decreased by maternal DEX exposure in P0 males. Furthermore, the number and levels of synaptic cluster inputs in close apposition with GnRH neurons was decreased following maternal DEX exposure in the OVLT region of P0 males. In addition, the postsynaptic marker molecule, postsynaptic density 95, was observed in GnRH neurons following both DEX and VEH treatment. These results suggest that maternal DEX exposure alters neural afferent inputs to GnRH neurons during early postnatal stage, which could lead to reproductive dysfunction during adulthood.
Collapse
Affiliation(s)
- Wei Ling Lim
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Marshita Mohd Idris
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Felix Suresh Kevin
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
- *Correspondence: Tomoko Soga,
| | - Ishwar S. Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| |
Collapse
|
39
|
Parent AS, Franssen D, Fudvoye J, Gérard A, Bourguignon JP. Developmental variations in environmental influences including endocrine disruptors on pubertal timing and neuroendocrine control: Revision of human observations and mechanistic insight from rodents. Front Neuroendocrinol 2015; 38:12-36. [PMID: 25592640 DOI: 10.1016/j.yfrne.2014.12.004] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 12/13/2014] [Accepted: 12/15/2014] [Indexed: 12/21/2022]
Abstract
Puberty presents remarkable individual differences in timing reaching over 5 years in humans. We put emphasis on the two edges of the age distribution of pubertal signs in humans and point to an extended distribution towards earliness for initial pubertal stages and towards lateness for final pubertal stages. Such distortion of distribution is a recent phenomenon. This suggests changing environmental influences including the possible role of nutrition, stress and endocrine disruptors. Our ability to assess neuroendocrine effects and mechanisms is very limited in humans. Using the rodent as a model, we examine the impact of environmental factors on the individual variations in pubertal timing and the possible underlying mechanisms. The capacity of environmental factors to shape functioning of the neuroendocrine system is thought to be maximal during fetal and early postnatal life and possibly less important when approaching the time of onset of puberty.
Collapse
Affiliation(s)
- Anne-Simone Parent
- Developmental Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Sart-Tilman, B-4000 Liège, Belgium; Department of Pediatrics, CHU de Liège, Rue de Gaillarmont 600, B-4032 Chênée, Belgium
| | - Delphine Franssen
- Developmental Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Sart-Tilman, B-4000 Liège, Belgium
| | - Julie Fudvoye
- Developmental Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Sart-Tilman, B-4000 Liège, Belgium; Department of Pediatrics, CHU de Liège, Rue de Gaillarmont 600, B-4032 Chênée, Belgium
| | - Arlette Gérard
- Developmental Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Sart-Tilman, B-4000 Liège, Belgium; Department of Pediatrics, CHU de Liège, Rue de Gaillarmont 600, B-4032 Chênée, Belgium
| | - Jean-Pierre Bourguignon
- Developmental Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Sart-Tilman, B-4000 Liège, Belgium; Department of Pediatrics, CHU de Liège, Rue de Gaillarmont 600, B-4032 Chênée, Belgium.
| |
Collapse
|
40
|
Wilsterman K, Mast AD, Luu TH, Haussmann MF. The timing of embryonic exposure to elevated temperature alters stress endocrinology in domestic chickens (Gallus domesticus). Gen Comp Endocrinol 2015; 212:10-6. [PMID: 25623149 DOI: 10.1016/j.ygcen.2015.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 12/28/2014] [Accepted: 01/14/2015] [Indexed: 10/24/2022]
Abstract
Patterns of glucocorticoid (GC) release in response to stimuli vary both among individuals and within individuals across their lifetime. While much work has focused on how the prenatal steroid environment can affect GC release, relatively little is known about how environmental parameters, such as incubation temperature affect GCs. We tested the hypothesis that variation and timing of elevated incubation temperature within the thermoneutral zone can alter the pattern of GC release. We incubated domestic chicken eggs (Gallus domesticus) at the optimal incubation temperature (37.5 °C) or at a slightly higher temperature (+1.1 °C) either early, late, or throughout incubation. At three weeks post-hatch, all birds were (i) exposed to a capture-restraint stress to measure stress-induced GC release (naïve). Three days following the naïve stressor, birds were (ii) exposed to a heat challenge, which was followed the next day by a second capture-restraint stress (post-heat challenge). Regardless of treatment, birds had similar patterns of GC release following the naïve stress series. However, during the post-heat challenge stress series, birds incubated at optimal temperatures increased their peak GC release. In contrast, birds exposed to slightly elevated temperatures for any period of development failed to increase peak GC release, and their specific response varied with timing of exposure to the elevated incubation temperature. Our results demonstrate that subtle variation in the embryonic environment, such as elevated incubation temperature within the thermoneutral zone, can impact the pattern of GC release of offspring. Further work is needed to understand the mechanisms underlying these changes and the relationship between fitness and environmentally-altered phenotypes.
Collapse
Affiliation(s)
| | - Andrew D Mast
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA
| | - Thuyvan H Luu
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA
| | - Mark F Haussmann
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA.
| |
Collapse
|
41
|
Zelena D. The janus face of stress on reproduction: from health to disease. Int J Endocrinol 2015; 2015:458129. [PMID: 25945091 PMCID: PMC4405284 DOI: 10.1155/2015/458129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/17/2015] [Accepted: 03/20/2015] [Indexed: 12/16/2022] Open
Abstract
Parenthood is a fundamental feature of all known life. However, infertility has been recognized as a public health issue worldwide. But even when the offspring are conceived, in utero problems can lead to immediate (abortion), early (birth), and late (adulthood) consequences. One of the most studied factors is stress. However, stress response is, per se, of adaptive nature allowing the organism to cope with challenges. Stressors lead to deterioration if one is faced with too long lasting, too many, and seemingly unsolvable situations. In stress adaptation the hypothalamus-pituitary-adrenocortical axis and the resulting glucocorticoid elevation are one of the most important mechanisms. At cellular level stress can be defined as an unbalance between production of free radicals and antioxidant defenses. Oxidative stress is widely accepted as an important pathogenic mechanism in different diseases including infertility. On the other hand, the goal of free radical production is to protect the cells from infectious entities. This review aims to summarize the negative and positive influence of stress on reproduction as a process leading to healthy progeny. Special emphasis was given to the balance at the level of the organism and cells.
Collapse
Affiliation(s)
- Dóra Zelena
- Hungarian Academy of Sciences, Institute of Experimental Medicine, Szigony 43, Budapest 1083, Hungary
- *Dóra Zelena:
| |
Collapse
|
42
|
Lim R, Fedulov AV, Kobzik L. Maternal stress during pregnancy increases neonatal allergy susceptibility: role of glucocorticoids. Am J Physiol Lung Cell Mol Physiol 2014; 307:L141-8. [PMID: 24838749 PMCID: PMC4101791 DOI: 10.1152/ajplung.00250.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 05/13/2014] [Indexed: 01/29/2023] Open
Abstract
We sought to test experimentally whether maternal stress can promote susceptibility to development of asthma-like allergic airways disease in offspring. Normal pregnant mice (day 15) were subjected to a single restraint stress exposure. We subsequently tested their offspring for the development of airway hyperreactivity (AHR) and allergic airway inflammation (AI), after an intentionally suboptimal sensitization protocol. The offspring of stressed mothers showed levels of AI and enhanced airway responses to methacholine comparable to those seen in fully sensitized and challenged positive control animals; in contrast, minimal effects were seen in control offspring. Restraint stress caused a rapid and large increase in plasma corticosterone levels. Maternal treatment with dexamethasone on day 15 of pregnancy mimicked the stress effect and reproduced the AI and AHR outcomes, whereas blockade of the stress-induced corticosterone surge with metyrapone pretreatment of pregnant mice abrogated the effect. We conclude that stress-triggered glucocorticoids during pregnancy can increase susceptibility to allergy in offspring. Because inflammation typically includes a stress hormone response, the results also suggest a common pathway by which various injurious exposures during pregnancy might increase offspring susceptibility to asthma.
Collapse
Affiliation(s)
- Robert Lim
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| | - Alexey V Fedulov
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| | - Lester Kobzik
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| |
Collapse
|
43
|
Mark PJ, Wyrwoll CS, Zulkafli IS, Mori TA, Waddell BJ. Rescue of glucocorticoid-programmed adipocyte inflammation by omega-3 fatty acid supplementation in the rat. Reprod Biol Endocrinol 2014; 12:39. [PMID: 24886466 PMCID: PMC4022445 DOI: 10.1186/1477-7827-12-39] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 04/26/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Adverse fetal environments predispose offspring to pathologies associated with the metabolic syndrome. Previously we demonstrated that adult offspring of dexamethasone-treated mothers had elevated plasma insulin and pro-inflammatory cytokines, effects prevented by a postnatal diet enriched with omega (n)-3 fatty acids. Here we tested whether prenatal glucocorticoid excess also programmed the adipose tissue phenotype, and whether this outcome is rescued by dietary n-3 fatty acids. METHODS Offspring of control and dexamethasone-treated mothers (0.75 μg/ml in drinking water, day 13 to term) were cross-fostered to mothers on a standard (Std) or high n-3 (Hn3) diet at birth. Offspring remained on these diets post-weaning, and serum and retroperitoneal fat were obtained at 6 months of age (n = 5-8 per group). Serum was analysed for blood lipids and fatty acid profiles, adipocyte cross sectional area was measured by unbiased stereological analysis and adipose expression of markers of inflammation, glucocorticoid sensitivity and lipid metabolism were determined by RT-qPCR analysis. RESULTS Serum total fatty acid levels were elevated (P < 0.01) in male offspring of dexamethasone-treated mothers, an effect prevented by Hn3 consumption. Prenatal dexamethasone also programmed increased adipose expression of Il6, Il1b (both P < 0.05) and Tnfa (P < 0.001) mRNAs regardless of fetal sex, but again this effect was prevented (for Il6 and Il1b) by Hn3 consumption. Offspring of dexamethasone-treated mothers had increased adipose expression of Gr (P = 0.008) and Ppara (P < 0.05) regardless of sex or postnatal diet, while 11bHsd1 was upregulated in males only. The Hn3 diet increased Ppard expression and reduced adipocyte size in all offspring (both P < 0.05) irrespective of prenatal treatment. CONCLUSIONS Prenatal glucocorticoid exposure programmed increased expression of inflammatory markers and enhanced glucocorticoid sensitivity of adipose tissue. Partial prevention of this phenotype by high n-3 consumption indicates that postnatal dietary manipulations can limit adverse fetal programming effects on adipose tissue.
Collapse
Affiliation(s)
- Peter J Mark
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Perth, Australia
| | - Caitlin S Wyrwoll
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Perth, Australia
| | - Intan S Zulkafli
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Perth, Australia
| | - Trevor A Mori
- School of Medicine and Pharmacology, The University of Western Australia, Perth, Australia
| | - Brendan J Waddell
- School of Anatomy, Physiology & Human Biology, The University of Western Australia, Perth, Australia
| |
Collapse
|
44
|
Guzmán C, García-Becerra R, Aguilar-Medina MA, Méndez I, Merchant-Larios H, Zambrano E. Maternal protein restriction during pregnancy and/or lactation negatively affects follicular ovarian development and steroidogenesis in the prepubertal rat offspring. Arch Med Res 2014; 45:294-300. [PMID: 24819035 DOI: 10.1016/j.arcmed.2014.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/14/2014] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Maternal protein restriction during rat pregnancy and lactation is associated with alterations in reproductive function of female offspring including delayed onset of puberty, decreased fertility and premature reproductive aging. These alterations may be related to ovarian prepubertal development, distribution of follicle populations and their steroidogenic capacities. We undertook this study to evaluate the ovarian function of prepubertal female offspring exposed to maternal protein restriction during pregnancy and/or lactation. METHODS Adult female Wistar rats were fed a control (C-20% casein diet) or restricted isocaloric diet (R-10% casein) during pregnancy--first letter--and lactation--second letter, to form four groups, CC, RR, CR, RC. Ovaries were collected from 21-day-old female offspring. Preantral and antral follicles were quantified and mRNA expression of key genes involved in follicular development and steroidogenesis (gonadotropin receptors, StAR, P450scc and P450 aromatase) was evaluated. Serum gonadotropin levels were measured. RESULTS Significantly decreased numbers of preantral and antral follicles were observed in CR and RC ovaries compared with CC. LH levels were lower and FSH higher in CR pups. mRNA expression of LH receptor (LH-R) was decreased in RR in comparison with the other groups. CR and RC expressed higher StAR, RC increased and RR decreased P450scc, whereas RR and CR decreased aromatase expression in comparison with CC. CONCLUSIONS Maternal protein restriction influences prepubertal ovarian follicular number and steroidogenic function in the rat offspring, although RR and CR nutritional schemes have similar outcomes, the mechanisms affecting ovarian function are at different levels of the hypothalamic-pituitary-ovarian axis.
Collapse
Affiliation(s)
- Carolina Guzmán
- HIPAM, Unidad de Medicina Experimental, Hospital General de México, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), México, D.F., México
| | - Rocío García-Becerra
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SalvadorZubirán, México, D.F., México
| | - Marco Antonio Aguilar-Medina
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SalvadorZubirán, México, D.F., México
| | - Isabel Méndez
- Instituto de Neurobiología, UNAM, Campus UNAM-Juriquilla, México
| | - Horacio Merchant-Larios
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, UNAM, México, D.F., México
| | - Elena Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SalvadorZubirán, México, D.F., México.
| |
Collapse
|
45
|
Rogers SL, Hughes BA, Jones CA, Freedman L, Smart K, Taylor N, Stewart PM, Shackleton CHL, Krone NP, Blissett J, Tomlinson JW. Diminished 11β-hydroxysteroid dehydrogenase type 2 activity is associated with decreased weight and weight gain across the first year of life. J Clin Endocrinol Metab 2014; 99:E821-31. [PMID: 24517145 DOI: 10.1210/jc.2013-3254] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Low birth weight is associated with adverse metabolic outcome in adulthood. Exposure to glucocorticoid (GC) excess in utero is associated with decreased birth weight, but the prospective longitudinal relationship between GC metabolism and growth has not been examined. OBJECTIVE We have hypothesized that changes in GC metabolism leading to increased availability may impair growth. DESIGN This was a prospective, longitudinal study with clinical measurements and 24-hour urinary steroid metabolite analysis at 1, 4, 12, 26, and 52 weeks after delivery in mothers and their babies. SETTING The study was conducted with observations and samples collected in the volunteers' own homes. PARTICIPANTS Healthy mothers and newborn babies/infants participated in the study. INTERVENTIONS There were no interventions. MAIN OUTCOME MEASURES Urinary steroid metabolite excretion quantified by gas chromatography/mass spectroscopy across the first year of life in relation to change in weight was measured. RESULTS The total production of the GC metabolites quantified increased across the first year of life. Markers of 11β-hydroxysteroid dehydrogenase type 1 activity increased from the age of 3 months as did those of 5α-reductase activity. After correcting for confounding variables, low markers of 11β-hydroxysteroid dehydrogenase type 2 activity was associated with reduced absolute weight and decreased weight gain over the first year of life. In the mothers, 5α-reductase activity was low at birth and progressively increased to normal over the first 6 months postpartum. CONCLUSIONS Increased GC exposure as a consequence of reduced 11β-hydroxysteroid dehydrogenase type 2 activity is likely to be a critical determinant of growth in early life. This not only highlights the central role of GCs and their metabolism, but also emphasizes the need for detailed longitudinal analyses.
Collapse
Affiliation(s)
- Samantha L Rogers
- School of Psychology (S.L.R., C.A.J., L.F., K.S., J.B.) and Centre for Endocrinology, Diabetes, and Metabolism (B.A.H., P.M.S., C.H.L.S., N.P.K., J.W.T.), Institute of Biomedical Research, School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham B15 2TH, United Kingdom; Department of Clinical Biochemistry (N.T.), King's College Hospital National Health Service Foundation Trust, London WC2R 2LS, United Kingdom; and Children's Hospital, Oakland's Research Institute (C.H.L.S.), Oakland, California 94609
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Iwasa T, Matsuzaki T, Munkhzaya M, Tungalagsuvd A, Kawami T, Murakami M, Yamasaki M, Kato T, Kuwahara A, Yasui T, Irahara M. Prenatal exposure to glucocorticoids affects body weight, serum leptin levels, and hypothalamic neuropeptide‐Y expression in pre‐pubertal female rat offspring. Int J Dev Neurosci 2014; 36:1-4. [DOI: 10.1016/j.ijdevneu.2014.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/15/2014] [Accepted: 03/26/2014] [Indexed: 12/21/2022] Open
Affiliation(s)
- Takeshi Iwasa
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Toshiya Matsuzaki
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Munkhsaikhan Munkhzaya
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Altankhuu Tungalagsuvd
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Takako Kawami
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Masahiro Murakami
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Mikio Yamasaki
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Takeshi Kato
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Akira Kuwahara
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Toshiyuki Yasui
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| | - Minoru Irahara
- Department of Obstetrics and GynecologyThe University of Tokushima Graduate SchoolInstitute of Health Biosciences3‐18‐15 Kuramoto‐ChoTokushima770‐8503Japan
| |
Collapse
|
47
|
Durrant LM, Khorram O, Buchholz JN, Pearce WJ. Maternal food restriction modulates cerebrovascular structure and contractility in adult rat offspring: effects of metyrapone. Am J Physiol Regul Integr Comp Physiol 2014; 306:R401-10. [PMID: 24477541 DOI: 10.1152/ajpregu.00436.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Although the effects of prenatal undernutrition on adult cardiovascular health have been well studied, its effects on the cerebrovascular structure and function remain unknown. We used a pair-fed rat model of 50% caloric restriction from day 11 of gestation to term, with ad libitum feeding after birth. We validated that maternal food restriction (MFR) stress is mediated by glucocorticoids by administering metyrapone, a corticosterone synthesis inhibitor, to MFR mothers at day 11 of gestation. At age 8 mo, offspring from Control, MFR, and MFR + Metyrapone groups were killed, and middle cerebral artery (MCA) segments were studied using vessel-bath myography and confocal microscopy. Colocalization of smooth muscle α-actin (SMαA) with nonmuscle (NM), SM1 and SM2 myosin heavy-chain (MHC) isoforms was used to assess smooth muscle phenotype. Our results indicate that artery stiffness and wall thickness were increased, pressure-evoked myogenic reactivity was depressed, and myofilament Ca(2+) sensitivity was decreased in offspring of MFR compared with Control rats. MCA from MFR offspring exhibited a significantly greater SMαA/NM colocalization, suggesting that the smooth muscle cells had been altered toward a noncontractile phenotype. MET significantly reversed the effects of MFR on stiffness but not myogenic reactivity, lowered SMαA/NM colocalization, and increased SMαA/SM2 colocalization. Together, our data suggest that MFR alters cerebrovascular contractility via both glucocorticoid-dependent and glucocorticoid-independent mechanisms.
Collapse
Affiliation(s)
- Lara M Durrant
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California; and
| | | | | | | |
Collapse
|
48
|
Zambrano E, Guzmán C, Rodríguez-González GL, Durand-Carbajal M, Nathanielsz PW. Fetal programming of sexual development and reproductive function. Mol Cell Endocrinol 2014; 382:538-549. [PMID: 24045010 DOI: 10.1016/j.mce.2013.09.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 11/15/2022]
Abstract
The recent growth of interest in developmental programming of physiological systems has generally focused on the cardiovascular system (especially hypertension) and predisposition to metabolic dysfunction (mainly obesity and diabetes). However, it is now clear that the full range of altered offspring phenotypes includes impaired reproductive function. In rats, sheep and nonhuman primates, reproductive capacity is altered by challenges experienced during critical periods of development. This review will examine available experimental evidence across commonly studied experimental species for developmental programming of female and male reproductive function throughout an individual's life-course. It is necessary to consider events that occur during fetal development, early neonatal life and prior to and during puberty, during active reproductive life and aging as reproductive performance declines.
Collapse
Affiliation(s)
- Elena Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México.
| | - Carolina Guzmán
- HIPAM, Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM)/Hospital General de México, México
| | - Guadalupe L Rodríguez-González
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México
| | - Marta Durand-Carbajal
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México
| | - Peter W Nathanielsz
- Center for Pregnancy and Newborn Research, Department of Obstetrics, University of Texas Health Sciences Center San Antonio, TX, United States
| |
Collapse
|
49
|
Iozzo P, Holmes M, Schmidt MV, Cirulli F, Guzzardi MA, Berry A, Balsevich G, Andreassi MG, Wesselink JJ, Liistro T, Gómez-Puertas P, Eriksson JG, Seckl J. Developmental ORIgins of Healthy and Unhealthy AgeiNg: the role of maternal obesity--introduction to DORIAN. Obes Facts 2014; 7:130-51. [PMID: 24801105 PMCID: PMC5644840 DOI: 10.1159/000362656] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/12/2014] [Indexed: 12/31/2022] Open
Abstract
Europe has the highest proportion of elderly people in the world. Cardiovascular disease, type 2 diabetes, sarcopenia and cognitive decline frequently coexist in the same aged individual, sharing common early risk factors and being mutually reinforcing. Among conditions which may contribute to establish early risk factors, this review focuses on maternal obesity, since the epidemic of obesity involves an ever growing number of women of reproductive age and children, calling for appropriate studies to understand the consequences of maternal obesity on the offspring's health and for developing effective measures and policies to improve people's health before their conception and birth. Though the current knowledge suggests that the long-term impact of maternal obesity on the offspring's health may be substantial, the outcomes of maternal obesity over the lifespan have not been quantified, and the molecular changes induced by maternal obesity remain poorly characterized. We hypothesize that maternal insulin resistance and reduced placental glucocorticoid catabolism, leading to oxidative stress, may damage the DNA, either in its structure (telomere shortening) or in its function (via epigenetic changes), resulting in altered gene expression/repair, disease during life, and pathological ageing. This review illustrates the background to the EU-FP7-HEALTH-DORIAN project.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pis
- *Patricia Iozzo, MD, PhD, Institute of Clinical Physiology, National Research Council (CNR), Via Moruzzi 1, 56124 Pisa (Italy),
| | - Megan Holmes
- Endocrinology Unit, Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | | | | - Tiziana Liistro
- Institute of Clinical Physiology, National Research Council (CNR), Pis
| | | | - Johan G. Eriksson
- Samfundet Folkhälsan i Svenska Finland rf (Folkhälsan), Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Jonathan Seckl
- Endocrinology Unit, Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
50
|
Maternal dexamethasone exposure during pregnancy in rats disrupts gonadotropin-releasing hormone neuronal development in the offspring. Cell Tissue Res 2013; 355:409-23. [PMID: 24374911 PMCID: PMC3921457 DOI: 10.1007/s00441-013-1765-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/29/2013] [Indexed: 12/02/2022]
Abstract
The migration of gonadotropin-releasing hormone (GnRH) neurons from the olfactory placode to the preoptic area (POA) from embryonic day 13 is important for successful reproduction during adulthood. Whether maternal glucocorticoid exposure alters GnRH neuronal morphology and number in the offspring is unknown. This study determines the effect of maternal dexamethasone (DEX) exposure on enhanced green fluorescent protein (EGFP) driven by GnRH promoter neurons (TG-GnRH) in transgenic rats dual-labelled with GnRH immunofluorescence (IF-GnRH). The TG-GnRH neurons were examined in intact male and female rats at different postnatal ages, as a marker for GnRH promoter activity. Pregnant females were subcutaneously injected with DEX (0.1 mg/kg) or vehicle daily during gestation days 13–20 to examine the number of GnRH neurons in P0 male offspring. The total number of TG-GnRH neurons and TG-GnRH/IF-GnRH neuronal ratio increased from P0 and P5 stages to P47–52 stages, suggesting temporal regulation of GnRH promoter activity during postnatal development in intact rats. In DEX-treated P0 males, the number of IF-GnRH neurons decreased within the medial septum, organum vasculosom of the lamina terminalis (OVLT) and anterior hypothalamus. The percentage of TG-GnRH neurons with branched dendritic structures decreased in the OVLT of DEX-P0 males. These results suggest that maternal DEX exposure affects the number and dendritic development of early postnatal GnRH neurons in the OVLT/POA, which may lead to altered reproductive functions in adults.
Collapse
|