1
|
Lockett J, Inder WJ, Clifton VL. The Glucocorticoid Receptor: Isoforms, Functions, and Contribution to Glucocorticoid Sensitivity. Endocr Rev 2024; 45:593-624. [PMID: 38551091 PMCID: PMC11244253 DOI: 10.1210/endrev/bnae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Indexed: 07/13/2024]
Abstract
Glucocorticoids exert pleiotropic effects on all tissues to regulate cellular and metabolic homeostasis. Synthetic forms are used therapeutically in a wide range of conditions for their anti-inflammatory benefits, at the cost of dose and duration-dependent side effects. Significant variability occurs between tissues, disease states, and individuals with regard to both the beneficial and deleterious effects. The glucocorticoid receptor (GR) is the site of action for these hormones and a vast body of work has been conducted understanding its function. Traditionally, it was thought that the anti-inflammatory benefits of glucocorticoids were mediated by transrepression of pro-inflammatory transcription factors, while the adverse metabolic effects resulted from direct transactivation. This canonical understanding of the GR function has been brought into question over the past 2 decades with advances in the resolution of scientific techniques, and the discovery of multiple isoforms of the receptor present in most tissues. Here we review the structure and function of the GR, the nature of the receptor isoforms, and the contribution of the receptor to glucocorticoid sensitivity, or resistance in health and disease.
Collapse
Affiliation(s)
- Jack Lockett
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4101, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Woolloongabba, QLD 4102, Australia
| | - Warrick J Inder
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Woolloongabba, QLD 4102, Australia
| | - Vicki L Clifton
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4101, Australia
| |
Collapse
|
2
|
Bakhireva LN, Solomon E, Roberts MH, Ma X, Rai R, Wiesel A, Jacobson SW, Weinberg J, Milligan ED. Independent and Combined Effects of Prenatal Alcohol Exposure and Prenatal Stress on Fetal HPA Axis Development. Int J Mol Sci 2024; 25:2690. [PMID: 38473937 PMCID: PMC10932119 DOI: 10.3390/ijms25052690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Prenatal alcohol exposure (PAE) and prenatal stress (PS) are highly prevalent conditions known to affect fetal programming of the hypothalamic-pituitary-adrenal (HPA) axis. The objectives of this study were to assess the effect of light PAE, PS, and PAE-PS interaction on fetal HPA axis activity assessed via placental and umbilical cord blood biomarkers. Participants of the ENRICH-2 cohort were recruited during the second trimester and classified into the PAE and unexposed control groups. PS was assessed by the Perceived Stress Scale. Placental tissue was collected promptly after delivery; gene and protein analysis for 11β-HSD1, 11β-HSD2, and pCRH were conducted by qPCR and ELISA, respectively. Umbilical cord blood was analyzed for cortisone and cortisol. Pearson correlation and multivariable linear regression examined the association of PAE and PS with HPA axis biomarkers. Mean alcohol consumption in the PAE group was ~2 drinks/week. Higher PS was observed in the PAE group (p < 0.01). In multivariable modeling, PS was associated with pCRH gene expression (β = 0.006, p < 0.01), while PAE was associated with 11β-HSD2 protein expression (β = 0.56, p < 0.01). A significant alcohol-by-stress interaction was observed with respect to 11β-HSD2 protein expression (p < 0.01). Results indicate that PAE and PS may independently and in combination affect fetal programming of the HPA axis.
Collapse
Affiliation(s)
- Ludmila N. Bakhireva
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Elizabeth Solomon
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA; (E.S.); (E.D.M.)
| | - Melissa H. Roberts
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Xingya Ma
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Rajani Rai
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Alexandria Wiesel
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Sandra W. Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| | - Erin D. Milligan
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA; (E.S.); (E.D.M.)
| |
Collapse
|
3
|
Harders EP, Agustin M, Paitz RT. Avian extraembryonic membranes respond to yolk corticosterone early in development. Biol Open 2024; 13:bio060131. [PMID: 38156650 PMCID: PMC10836647 DOI: 10.1242/bio.060131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
During times of maternal stress, developing embryos can be exposed to elevated levels of glucocorticoids, which can affect development and permanently alter offspring phenotype. In placental species, the placenta mediates fetal exposure to maternal glucocorticoids via metabolism, yet the placenta itself responds to glucocorticoids to regulate offspring growth and development. In oviparous species, maternal glucocorticoids can be deposited into the egg yolk and are metabolized early in development. This metabolism is mediated by the extraembryonic membranes, but it is unknown if the extraembryonic membranes also respond to maternal glucocorticoids in a way comparable to the placenta. In this study, we quantified the expression of acyl-CoA thioesterase 13 (Acot13) as an initial marker of the membrane's response to corticosterone in chicken (Gallus gallus) eggs. Acot13 regulates fatty acid processing in the embryo, to potentially regulate resource availability during development. We addressed the following questions using Acot13 expression: 1) Do the extraembryonic membranes respond to yolk corticosterone early in development? 2) Is the response to corticosterone dependent on the dose of corticosterone? 3) What is the duration of the response to corticosterone? 4) Does a metabolite of corticosterone (5β-corticosterone) elicit the same response as corticosterone? We found that corticosterone significantly induces the expression of Acot13 on day four of development and that expression of Acot13 increases with the dose of corticosterone. Further, we found expression of Acot13 is significantly elevated by corticosterone on days four and six of development compared to oil treated eggs, but not on days eight and ten. Although this response is transient, it occurs during a critical period of development and could initiate a cascade of events that ultimately alter offspring phenotype. Finally, we found that 5β-corticosterone does not increase the expression of Acot13, indicating that metabolism inactivates corticosterone. Ultimately, this study provides insight into the mechanisms underlying how maternally deposited glucocorticoids can affect embryonic development.
Collapse
Affiliation(s)
- Emily P. Harders
- Illinois State University, School of Biological Sciences, Campus Box 4120, Normal, IL 61790, USA
| | - Mitch Agustin
- Illinois State University, School of Biological Sciences, Campus Box 4120, Normal, IL 61790, USA
| | - Ryan T. Paitz
- Illinois State University, School of Biological Sciences, Campus Box 4120, Normal, IL 61790, USA
| |
Collapse
|
4
|
Paul EN, Shubitidze S, Rahim R, Rucker I, Valin L, Apostle S, Pospisilik JA, Racicot KE, Smith AL. Exogenous corticosterone administration during pregnancy in mice alters placental and fetal thyroid hormone availability in females. Placenta 2023; 142:1-11. [PMID: 37579594 PMCID: PMC10763606 DOI: 10.1016/j.placenta.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023]
Abstract
INTRODUCTION Maternal prenatal psychological stress is associated with adverse pregnancy outcomes and increased risk of adverse health outcomes in children. While the molecular mechanisms that govern these associations has not been fully teased apart, stress-induced changes in placental function can drive sex-specific phenotypes in offspring. We sought to identify and examine molecular pathways in the placenta that are altered in response to maternal prenatal stress. METHODS We previously employed a mouse model of maternal prenatal stress where pregnant dams were treated with stress hormone (CORT) beginning in mid-gestation. Using this model, we conducted RNAseq analysis of whole placenta at E18.5. We used qRT-PCR to validate gene expression changes in the placenta and in a trophoblast cell line. ELISAs were used to measure the abundance of thyroid hormones in maternal and fetal serum and in the placenta. RESULTS Dio2 was amongst the top differentially expressed genes in response to exogenous stress hormone. Dio2 expression was more downregulated in placenta of female fetuses from CORT-treated dams than both control placenta from females and placenta from male fetuses. Consistent with Dio2's role in production of bioactive thyroid hormone (T3), we found that there was a reduction of T3 in placenta and serum of female embryos from CORT-treated dams at E18.5. Both T3 and T4 were reduced in the fetal compartment of the placenta of female fetuses from CORT-treated dams at E16.5. Exogenous stress hormone induced reduction in thyroid hormone in females was independent of circulating levels of TH in the dams. DISCUSSION The placental thyroid hormone synthesis pathway may be a target of elevated maternal stress hormone and modulate fetal programming of health and disease of offspring in a sex-specific fashion.
Collapse
Affiliation(s)
- Emmanuel N Paul
- Dept of Obstetrics, Gynecology and Reproductive Sciences, Michigan State University, College of Human Medicine, Grand Rapids, MI, 49503, USA
| | | | - Rodaba Rahim
- Biology Department, Kenyon College, Gambier, OH, 43050, USA
| | - Imani Rucker
- Biology Department, Kenyon College, Gambier, OH, 43050, USA
| | - Liana Valin
- Biology Department, Kenyon College, Gambier, OH, 43050, USA
| | - Stefanos Apostle
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - J Andrew Pospisilik
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Karen E Racicot
- Dept of Obstetrics, Gynecology and Reproductive Sciences, Michigan State University, College of Human Medicine, Grand Rapids, MI, 49503, USA
| | - Arianna L Smith
- Biology Department, Kenyon College, Gambier, OH, 43050, USA.
| |
Collapse
|
5
|
Paul EN, Shubitidze S, Rahim R, Rucker I, Valin L, Apostle S, Andrew Pospisilik J, Racicot KE, Smithb AL. Exogenous corticosterone administration during pregnancy alters placental and fetal thyroid hormone availability in females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547278. [PMID: 37461599 PMCID: PMC10349991 DOI: 10.1101/2023.07.05.547278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Introduction Maternal prenatal stress is associated with adverse pregnancy outcomes and predisposition to long-term adverse health outcomes in children. While the molecular mechanisms that govern these associations has not been fully teased apart, stress-induced changes in placental function can drive sex-specific phenotypes in offspring. We sought to identify and examine molecular pathways in the placenta that are altered in response to maternal prenatal stress. Methods Using a mouse model of maternal prenatal stress, we conducted RNA-seq analysis of whole placenta at E18.5. We used qRT-PCR to validate gene expression changes in the placenta and in a trophoblast cell line. ELISAs were used to measure the abundance of thyroid hormones in maternal and fetal serum and in the placenta. Results Dio2 was amongst the top differentially expressed genes in response to elevated maternal stress hormone. Dio2 expression was more downregulated in female placenta from stressed dams than both female control and male placenta. Consistent with Dio2's role in production of bioactive thyroid hormone (T3), we found that there was a reduction of T3 in placenta and serum of female embryos from stressed dams at E18.5. Both T3 and T4 were reduced in the fetal compartment of the female placenta from stressed dams at E16.5. Stress hormone induced reduction in thyroid hormone in females was independent of circulating levels of TH in the dams. Discussion The placental thyroid hormone synthesis pathway may be a target of maternal stress and modulate fetal programming of health and disease of offspring in a sex-specific fashion.
Collapse
|
6
|
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
|
7
|
Orzeł A, Unrug-Bielawska K, Filipecka-Tyczka D, Berbeka K, Zeber-Lubecka N, Zielińska M, Kajdy A. Molecular Pathways of Altered Brain Development in Fetuses Exposed to Hypoxia. Int J Mol Sci 2023; 24:10401. [PMID: 37373548 DOI: 10.3390/ijms241210401] [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: 05/10/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Perinatal hypoxia is a major cause of neurodevelopmental impairment and subsequent motor and cognitive dysfunctions; it is associated with fetal growth restriction and uteroplacental dysfunction during pregnancy. This review aims to present the current knowledge on brain development resulting from perinatal asphyxia, including the causes, symptoms, and means of predicting the degree of brain damage. Furthermore, this review discusses the specificity of brain development in the growth-restricted fetus and how it is replicated and studied in animal models. Finally, this review aims at identifying the least understood and missing molecular pathways of abnormal brain development, especially with respect to potential treatment intervention.
Collapse
Affiliation(s)
- Anna Orzeł
- Centre of Postgraduate Medical Education, I-st Department of Obstetrics and Gynecology, 01-813 Warsaw, Poland
| | - Katarzyna Unrug-Bielawska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 00-001 Warsaw, Poland
| | - Dagmara Filipecka-Tyczka
- Centre of Postgraduate Medical Education, I-st Department of Obstetrics and Gynecology, 01-813 Warsaw, Poland
| | - Krzysztof Berbeka
- Centre of Postgraduate Medical Education, I-st Department of Obstetrics and Gynecology, 01-813 Warsaw, Poland
| | - Natalia Zeber-Lubecka
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 00-001 Warsaw, Poland
- Centre of Postgraduate Medical Education, Department of Gastroenterology, Hepatology and Clinical Oncology, 01-813 Warsaw, Poland
| | - Małgorzata Zielińska
- Centre of Postgraduate Medical Education, I-st Department of Obstetrics and Gynecology, 01-813 Warsaw, Poland
| | - Anna Kajdy
- Centre of Postgraduate Medical Education, I-st Department of Obstetrics and Gynecology, 01-813 Warsaw, Poland
| |
Collapse
|
8
|
Easton ZJW, Luo X, Li L, Regnault TRH. The impact of hyperglycemia upon BeWo trophoblast cell metabolic function: A multi-OMICS and functional metabolic analysis. PLoS One 2023; 18:e0283118. [PMID: 36930661 PMCID: PMC10022812 DOI: 10.1371/journal.pone.0283118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
Pre-existing and gestationally-developed diabetes mellitus have been linked with impairments in placental villous trophoblast cell metabolic function, that are thought to underlie the development of metabolic diseases early in the lives of the exposed offspring. Previous research using placental cell lines and ex vivo trophoblast preparations have highlighted hyperglycemia is an important independent regulator of placental function. However, it is poorly understood if hyperglycemia directly influences aspects of placental metabolic function, including nutrient storage and mitochondrial respiration, that are altered in term diabetic placentae. The current study examined metabolic and mitochondrial function as well as nutrient storage in both undifferentiated cytotrophoblast and differentiated syncytiotrophoblast BeWo cells cultured under hyperglycemia conditions (25 mM glucose) for 72 hours to further characterize the direct impacts of placental hyperglycemic exposure. Hyperglycemic-exposed BeWo trophoblasts displayed increased glycogen and triglyceride nutrient stores, but real-time functional readouts of metabolic enzyme activity and mitochondrial respiratory activity were not altered. However, specific investigation into mitochondrial dynamics highlighted increased expression of markers associated with mitochondrial fission that could indicate high glucose-exposed trophoblasts are transitioning towards mitochondrial dysfunction. To further characterize the impacts of independent hyperglycemia, the current study subsequently utilized a multi-omics approach and evaluated the transcriptomic and metabolomic signatures of BeWo cytotrophoblasts. BeWo cytotrophoblasts exposed to hyperglycemia displayed increased mRNA expression of ACSL1, HSD11B2, RPS6KA5, and LAP3 and reduced mRNA expression of CYP2F1, and HK2, concomitant with increased levels of: lactate, malonate, and riboflavin metabolites. These changes highlighted important underlying alterations to glucose, glutathione, fatty acid, and glucocorticoid metabolism in BeWo trophoblasts exposed to hyperglycemia. Overall, these results demonstrate that hyperglycemia is an important independent regulator of key areas of placental metabolism, nutrient storage, and mitochondrial function, and these data continue to expand our knowledge on mechanisms governing the development of placental dysfunction.
Collapse
Affiliation(s)
- Zachary J W Easton
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Xian Luo
- The Metabolomics Innovation Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Liang Li
- The Metabolomics Innovation Centre, University of Alberta, Edmonton, Alberta, Canada
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Timothy R H Regnault
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- Department of Obstetrics and Gynaecology, London Health Science Centre-Victoria Hospital, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
9
|
Christians JK. The Placenta's Role in Sexually Dimorphic Fetal Growth Strategies. Reprod Sci 2021; 29:1895-1907. [PMID: 34699045 DOI: 10.1007/s43032-021-00780-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/19/2021] [Indexed: 12/27/2022]
Abstract
Fetal sex affects the risk of pregnancy complications and the long-term effects of prenatal environment on health. Some have hypothesized that growth strategies differ between the sexes, whereby males prioritize growth whereas females are more responsive to their environment. This review evaluates the role of the placenta in such strategies, focusing on (1) mechanisms underlying sexual dimorphism in gene expression, (2) the nature and extent of sexual dimorphism in placental gene expression, (3) sexually dimorphic responses to nutrient supply, and (4) sexual dimorphism in morphology and histopathology. The sex chromosomes contribute to sex differences in placental gene expression, and fetal hormones may play a role later in development. Sexually dimorphic placental gene expression may contribute to differences in the prevalence of complications such as preeclampsia, although this link is not clear. Placental responses to nutrient supply frequently show sexual dimorphism, but there is no consistent pattern where one sex is more responsive. There are sex differences in the prevalence of placental histopathologies, and placental changes in pregnancy complications, but also many similarities. Overall, no clear patterns support the hypothesis that females are more responsive to the maternal environment, or that males prioritize growth. While male fetuses are at greater risk of a variety of complications, total prenatal mortality is higher in females, such that males exposed to early insults may be more likely to survive and be observed in studies of adverse outcomes. Going forward, robust statistical approaches to test for sex-dependent effects must be more widely adopted to reduce the incidence of spurious results.
Collapse
Affiliation(s)
- Julian K Christians
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada. .,Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada. .,British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada. .,Women's Health Research Institute, BC Women's Hospital and Health Centre, Vancouver, BC, Canada.
| |
Collapse
|
10
|
Yu P, Chen Y, Ge C, Wang H. Sexual dimorphism in placental development and its contribution to health and diseases. Crit Rev Toxicol 2021; 51:555-570. [PMID: 34666604 DOI: 10.1080/10408444.2021.1977237] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
According to the Developmental Origin of Health and Disease (DOHaD), intrauterine exposure to adverse environments can affect fetus and birth outcomes and lead to long-term disease susceptibility. Evidence has shown that neonatal outcomes and the timing and severity of adult diseases are sexually dimorphic. As the link between mother and fetus, the placenta is an essential regulator of fetal development programming. It is found that the physiological development trajectory of the placenta has sexual dimorphism. Furthermore, under pathological conditions, the placental function undergoes sex-specific adaptation to ensure fetal survival. Therefore, the placenta may be an important mediator of sexual dimorphism in neonatal outcomes and adult disease susceptibility. Few systematic reviews have been conducted on sexual dimorphism in placental development and its underlying mechanisms. In this review, sex chromosomes and sex hormones, as the main reasons for sexual differentiation of the placenta, will be discussed. Besides, in the etiology of fetal-originated adult diseases, overexposure to glucocorticoids is closely related to adverse neonatal outcomes and long-term disease susceptibility. Studies have found that prenatal glucocorticoid overexposure leads to sexually dimorphic expression of placental glucocorticoid receptor isoforms, resulting in different sensitivity of the placenta to glucocorticoids, and may further affect fetal development. The present review examines what is currently known about sex differences in placental development and the underlying regulatory mechanisms of this sex bias. This review highlights the importance of placental contributions to the origins of sexual dimorphism in health and diseases. It may help develop personalized diagnosis and treatment strategies for fetal development in pathological pregnancies.
Collapse
Affiliation(s)
- Pengxia Yu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, China
| | - Yawen Chen
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, China
| | - Caiyun Ge
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, China.,Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
11
|
Meakin AS, Cuffe JSM, Darby JRT, Morrison JL, Clifton VL. Let's Talk about Placental Sex, Baby: Understanding Mechanisms That Drive Female- and Male-Specific Fetal Growth and Developmental Outcomes. Int J Mol Sci 2021; 22:6386. [PMID: 34203717 PMCID: PMC8232290 DOI: 10.3390/ijms22126386] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 02/06/2023] Open
Abstract
It is well understood that sex differences exist between females and males even before they are born. These sex-dependent differences may contribute to altered growth and developmental outcomes for the fetus. Based on our initial observations in the human placenta, we hypothesised that the male prioritises growth pathways in order to maximise growth through to adulthood, thereby ensuring the greatest chance of reproductive success. However, this male-specific "evolutionary advantage" likely contributes to males being less adaptable to shifts in the in-utero environment, which then places them at a greater risk for intrauterine morbidities or mortality. Comparatively, females are more adaptable to changes in the in-utero environment at the cost of growth, which may reduce their risk of poor perinatal outcomes. The mechanisms that drive these sex-specific adaptations to a change in the in-utero environment remain unclear, but an increasing body of evidence within the field of developmental biology would suggest that alterations to placental function, as well as the feto-placental hormonal milieu, is an important contributing factor. Herein, we have addressed the current knowledge regarding sex-specific intrauterine growth differences and have examined how certain pregnancy complications may alter these female- and male-specific adaptations.
Collapse
Affiliation(s)
- Ashley S. Meakin
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - James S. M. Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.S.M.); (J.R.T.D.); (J.L.M.)
| | - Vicki L. Clifton
- Mater Medical Research Institute, The University of Queensland, Brisbane, QLD 4000, Australia
| |
Collapse
|
12
|
Gomes JDA, Olstad EW, Kowalski TW, Gervin K, Vianna FSL, Schüler-Faccini L, Nordeng HME. Genetic Susceptibility to Drug Teratogenicity: A Systematic Literature Review. Front Genet 2021; 12:645555. [PMID: 33981330 PMCID: PMC8107476 DOI: 10.3389/fgene.2021.645555] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
Since the 1960s, drugs have been known to cause teratogenic effects in humans. Such teratogenicity has been postulated to be influenced by genetics. The aim of this review was to provide an overview of the current knowledge on genetic susceptibility to drug teratogenicity in humans and reflect on future directions within the field of genetic teratology. We focused on 12 drugs and drug classes with evidence of teratogenic action, as well as 29 drugs and drug classes with conflicting evidence of fetal safety in humans. An extensive literature search was performed in the PubMed and EMBASE databases using terms related to the drugs of interest, congenital anomalies and fetal development abnormalities, and genetic variation and susceptibility. A total of 29 studies were included in the final data extraction. The eligible studies were published between 1999 and 2020 in 10 different countries, and comprised 28 candidate gene and 1 whole-exome sequencing studies. The sample sizes ranged from 20 to 9,774 individuals. Several drugs were investigated, including antidepressants (nine studies), thalidomide (seven studies), antiepileptic drugs (five studies), glucocorticoids (four studies), acetaminophen (two studies), and sex hormones (estrogens, one study; 17-alpha hydroxyprogesterone caproate, one study). The main neonatal phenotypic outcomes included perinatal complications, cardiovascular congenital anomalies, and neurodevelopmental outcomes. The review demonstrated that studies on genetic teratology are generally small, heterogeneous, and exhibit inconsistent results. The most convincing findings were genetic variants in SLC6A4, MTHFR, and NR3C1, which were associated with drug teratogenicity by antidepressants, antiepileptics, and glucocorticoids, respectively. Notably, this review demonstrated the large knowledge gap regarding genetic susceptibility to drug teratogenicity, emphasizing the need for further efforts in the field. Future studies may be improved by increasing the sample size and applying genome-wide approaches to promote the interpretation of results. Such studies could support the clinical implementation of genetic screening to provide safer drug use in pregnant women in need of drugs.
Collapse
Affiliation(s)
- Julia do Amaral Gomes
- Programa de Pós-Graduação em Genética e Biologia Molecular (PPGBM), Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Sistema Nacional de Informação sobre Agentes Teratogênicos (SIAT), Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Instituto Nacional de Genética Médica Populacional (INAGEMP), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Emilie Willoch Olstad
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Thayne Woycinck Kowalski
- Programa de Pós-Graduação em Genética e Biologia Molecular (PPGBM), Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Instituto Nacional de Genética Médica Populacional (INAGEMP), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Complexo de Ensino Superior de Cachoeirinha (CESUCA), Cachoeirinha, Brazil
| | - Kristina Gervin
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Division of Clinical Neuroscience, Department of Research and Innovation, Oslo University Hospital, Oslo, Norway
| | - Fernanda Sales Luiz Vianna
- Programa de Pós-Graduação em Genética e Biologia Molecular (PPGBM), Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Sistema Nacional de Informação sobre Agentes Teratogênicos (SIAT), Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Instituto Nacional de Genética Médica Populacional (INAGEMP), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Lavínia Schüler-Faccini
- Programa de Pós-Graduação em Genética e Biologia Molecular (PPGBM), Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Sistema Nacional de Informação sobre Agentes Teratogênicos (SIAT), Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Instituto Nacional de Genética Médica Populacional (INAGEMP), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Hedvig Marie Egeland Nordeng
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
13
|
Moisan MP. Sexual Dimorphism in Glucocorticoid Stress Response. Int J Mol Sci 2021; 22:ijms22063139. [PMID: 33808655 PMCID: PMC8003420 DOI: 10.3390/ijms22063139] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic stress is encountered in our everyday life and is thought to contribute to a number of diseases. Many of these stress-related disorders display a sex bias. Because glucocorticoid hormones are the main biological mediator of chronic stress, researchers have been interested in understanding the sexual dimorphism in glucocorticoid stress response to better explain the sex bias in stress-related diseases. Although not yet demonstrated for glucocorticoid regulation, sex chromosomes do influence sex-specific biology as soon as conception. Then a transient rise in testosterone start to shape the male brain during the prenatal period differently to the female brain. These organizational effects are completed just before puberty. The cerebral regions implicated in glucocorticoid regulation at rest and after stress are thereby impacted in a sex-specific manner. After puberty, the high levels of all gonadal hormones will interact with glucocorticoid hormones in specific crosstalk through their respective nuclear receptors. In addition, stress occurring early in life, in particular during the prenatal period and in adolescence will prime in the long-term glucocorticoid stress response through epigenetic mechanisms, again in a sex-specific manner. Altogether, various molecular mechanisms explain sex-specific glucocorticoid stress responses that do not exclude important gender effects in humans.
Collapse
|
14
|
Zeng Z, Ma W, Zhao R, Dong X. Airway exposure to perfluorooctanoate exacerbates airway hyperresponsiveness and downregulates glucocorticoid receptor expression in asthmatic mice. Transl Pediatr 2021; 10:323-332. [PMID: 33708518 PMCID: PMC7944165 DOI: 10.21037/tp-20-246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Multiple environmental risk factors play a vital role in the pathogenesis of asthma, which contribute to the phenotypic expression of asthma. Perfluorooctanoate (PFOA) is the most common and abundant perfluorocarbon (PFC) in humans, and it has been detected in water and the atmosphere worldwide. Glucocorticoid receptor (GR) is considered to exert a protective effect on asthma and is associated with the sensitivity to glucocorticoids. Dermal or oral exposure to PFOA has been shown to contribute various effects on airway inflammation in individuals with ovalbumin (OVA)-induced asthma. Notably, airway exposure has a critical contribution to the pathogenesis of asthma. However, the effect of airway exposure to PFOA on airway hyperresponsiveness (AHR) in patients with asthma is not currently understood. METHODS BALB/c mice were administered OVA to induce asthma. PFOA was then administered intratracheally to OVA-induced mice for seven days. Then we assessed the effect of airway exposure to PFOA on AHR and the regulation of the GR expression in asthmatic mice. RESULTS The results showed aggravated AHR and T helper type 2 (Th2) airway inflammation in asthmatic mice. Furthermore, these mice show a substantial decrease in the expression of the GR mRNA and protein. CONCLUSIONS These data strongly suggest that acute airway exposure to PFOA leads to Th2-related AHR and decreases GR expression, which may increase the difficulty in the treatment of asthma.
Collapse
Affiliation(s)
- Zeyu Zeng
- Department of Respiratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weihui Ma
- Department of Respiratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ran Zhao
- Department of Respiratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyan Dong
- Department of Respiratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Panel of suitable reference genes and its gender differences of fetal rat liver under physiological conditions and exposure to dexamethasone during pregnancy. Reprod Toxicol 2021; 100:74-82. [PMID: 33453333 DOI: 10.1016/j.reprotox.2021.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/17/2020] [Accepted: 01/11/2021] [Indexed: 01/04/2023]
Abstract
The panel of suitable reference genes in the fetal liver have not been reported. In this study, five commonly used reference genes (GAPDH, β-actin, Rn18 s, Rpl13a, and Rps29) were firstly selected as candidates. Bestkeeper, GeNorm, and NormFinder software were then used to screen out the panel of suitable reference genes of male and female fetal rat liver under physiological and prenatal dexamethasone exposure (PDE) conditions. Finally, we verified the reliability of the screened panel of reference genes by standardizing sterol regulatory element binding protein 1c (SREBP1c) expression with different reference genes. The results showed that GAPDH + Rn18 s and GAPDH + Rpl13a were respectively the panel of suitable reference genes in male and female rat fetal liver under the physiological model, while Rn18 s + Rps29 and GAPDH + Rn18 s were respectively under the PDE model. The results showed that different reference genes affected the statistical results of SREBP1c expression, and the screened panel of suitable reference genes under the PDE model had smaller intragroup differences, when compared with other reference genes under physiological and PDE models. In conclusion, we screened and determined that the panel of suitable reference genes were GAPDH + Rn18 s and Rn18 s + Rps29 in the male rat fetal liver under physiological and PDE models, while they were GAPDH + Rpl13a and GAPDH + Rn18 s in the females, and confirmed that the selection of the panel of suitable reference genes in the fetal liver had gender differences and pathological model specificity.
Collapse
|
16
|
Young SL, Saif Z, Meakin AS, McMaster ES, Hayes N, Gallo LA, Reid N, Moritz KM, Clifton VL. Alterations to Placental Glucocorticoid Receptor Expression with Alcohol Consumption. Reprod Sci 2021; 28:1390-1402. [PMID: 33409870 DOI: 10.1007/s43032-020-00413-1] [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: 07/31/2020] [Accepted: 11/23/2020] [Indexed: 11/29/2022]
Abstract
Maternal alcohol consumption during pregnancy results in elevated vulnerability to intrauterine growth restriction, preterm birth, miscarriage, and stillbirth. Many of the detrimental effects of fetal alcohol exposure may be mediated through placental dysfunction; however, the exact mechanisms remain unknown. Here, we aimed to determine the effect of maternal alcohol exposure prior to and during early pregnancy on placental glucocorticoid receptor (GR) isoforms, associated GR regulated genes, and infant outcomes. Participants carrying singleton fetuses (n = 113) were recruited during early pregnancy. Amount and type of alcohol consumed over the last 12 months were obtained at 18 weeks of gestation. The level of drinking was separated into none (0 g/day), low (< 10 g/day), moderate (10-100 g/day), and heavy (> 100 g/day). At delivery, placental weight, infant sex, birthweight, and head circumference were recorded. Placental GR isoforms and genes involved in downstream signalling pathways were quantified. The majority of women (70.8%) consumed alcohol. Of these, most consumed low (48.8%) or moderate (37.5%) amounts. Placental weight was unaffected by alcohol consumption, but infants born to heavy drinkers tended to be lighter at birth. In female, but not male, placentae, maternal alcohol consumption resulted in increased GRαC and decreased GRαD1 cytoplasmic expression. In both female and male placentae, a dampened inflammatory response was evident with maternal alcohol consumption, involving downregulated IL6R and upregulated POU2F2 gene expression, respectively. Maternal alcohol consumption in the months prior to, and/or during early, pregnancy alters placental GR isoform and expression of some inflammatory genes in a sex-specific manner.
Collapse
Affiliation(s)
- S L Young
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - Z Saif
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - A S Meakin
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - E S McMaster
- School of Chemical and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - N Hayes
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - L A Gallo
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia
| | - N Reid
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | - K M Moritz
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia.
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia.
| | - V L Clifton
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| |
Collapse
|
17
|
Wood M, Whirledge S. Mechanism of glucocorticoid action in immunology—Basic concepts. REPRODUCTIVE IMMUNOLOGY 2021:147-170. [DOI: 10.1016/b978-0-12-818508-7.00020-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
18
|
Effects of Maternal Chewing on Prenatal Stress-Induced Cognitive Impairments in the Offspring via Multiple Molecular Pathways. Int J Mol Sci 2020; 21:ijms21165627. [PMID: 32781547 PMCID: PMC7460630 DOI: 10.3390/ijms21165627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
We aimed to investigate the effects of maternal chewing on prenatal stress-induced cognitive impairments in the offspring and to explore the molecular pathways of maternal chewing in a mice model. Maternal chewing ameliorated spatial learning impairments in the offspring in a Morris water maze test. Immunohistochemistry and Western blot findings revealed that maternal chewing alleviated hippocampal neurogenesis impairment and increased the expression of hippocampal brain-derived neurotrophic factor in the offspring. In addition, maternal chewing increased the expression of glucocorticoid receptor (GR) and 11β-hydroxysteroid dehydrogenase isozyme 2 (11β-HSD2) and decreased the expression of 11β-HSD1 in the placenta, thereby attenuating the increase of glucocorticoid in the offspring. Furthermore, maternal chewing increased the expression of 11β-HSD2, FK506-binding protein 51 (FKBP51) and FKBP52 and decreased the expression of 11β-HSD1, thereby increasing hippocampal nuclear GR level. In addition, maternal chewing attenuated the increase in expression of DNMT1 and DNMT3a and the decrease in expression of histone H3 methylation at lysine 4, 9, 27 and histone H3 acetylation at lysine 9 induced by prenatal stress in the offspring. Our findings suggest that maternal chewing could ameliorate prenatal stress-induced cognitive impairments in the offspring at least in part by protecting placenta barrier function, alleviating hippocampal nuclear GR transport impairment and increasing the hippocampal brain-derived neurotrophic factor (BDNF) level.
Collapse
|
19
|
Silvestro S, Calcaterra V, Pelizzo G, Bramanti P, Mazzon E. Prenatal Hypoxia and Placental Oxidative Stress: Insights from Animal Models to Clinical Evidences. Antioxidants (Basel) 2020; 9:E414. [PMID: 32408702 PMCID: PMC7278841 DOI: 10.3390/antiox9050414] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023] Open
Abstract
Hypoxia is a common form of intrauterine stress characterized by exposure to low oxygen concentrations. Gestational hypoxia is associated with the generation of reactive oxygen species. Increase in oxidative stress is responsible for damage to proteins, lipids and DNA with consequent impairment of normal cellular functions. The purpose of this review is to propose a summary of preclinical and clinical evidences designed to outline the correlation between fetal hypoxia and oxidative stress. The results of the studies described show that increases of oxidative stress in the placenta is responsible for changes in fetal development. Specifically, oxidative stress plays a key role in vascular, cardiac and neurological disease and reproductive function dysfunctions. Moreover, the different finding suggests that the prenatal hypoxia-induced oxidative stress is associated with pregnancy complications, responsible for changes in fetal programming. In this way, fetal hypoxia predisposes the offspring to congenital anomalies and chronic diseases in future life. Several antioxidant agents, such as melatonin, erythropoietin, vitamin C, resveratrol and hydrogen, shown potential protective effects in prenatal hypoxia. However, future investigations will be needed to allow the implementation of these antioxidants in clinical practice for the promotion of health in early intrauterine life, in fetuses and children.
Collapse
Affiliation(s)
- Serena Silvestro
- Departmnent of Experimental Neurology, IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (P.B.)
| | - Valeria Calcaterra
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Gloria Pelizzo
- Department of Biomedical and Clinical Science “L. Sacco”, and Pediatric Surgery Department “V. Buzzi” Children’s Hospital, University of Milano, 20100 Milano, Italy;
| | - Placido Bramanti
- Departmnent of Experimental Neurology, IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (P.B.)
| | - Emanuela Mazzon
- Departmnent of Experimental Neurology, IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (P.B.)
| |
Collapse
|
20
|
Edvinsson Å, Hoyer A, Hansson M, Kallak TK, Sundström-Poromaa I, Skalkidou A, Lager S. Placental glucocorticoid receptors are not affected by maternal depression or SSRI treatment. Ups J Med Sci 2020; 125:30-36. [PMID: 31960733 PMCID: PMC7054983 DOI: 10.1080/03009734.2019.1702126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background: Prenatal depression is common, with an estimate that up to one in five pregnant women suffers from depressive symptoms. Maternal depression is associated with poor pregnancy outcomes such as preterm birth and low birth-weight. Such outcomes possibly affect offspring development. Previous studies suggest placental RNA levels of the glucocorticoid receptor are altered by maternal depression or anxiety; this stress may affect the placenta of male and female foetuses differently. However, it is unknown if the protein levels and activity of this receptor are additionally affected in women with depressive symptoms or being pharmacologically treated for depression.Methods: In this study, we investigated whether the glucocorticoid receptor (NR3C1) in the placenta is affected by maternal depression and/or selective serotonin reuptake inhibitor (SSRIs) treatment. Placentas from 45 women with singleton, term pregnancies were analysed by Western blot to determine glucocorticoid receptor levels, and by DNA-binding capacity to measure glucocorticoid receptor activation.Results: There were no differences in levels of the glucocorticoid receptor or activity between groups (control, depressive symptoms, and SSRI treatment; n = 45). Similarly, there was no difference in placental glucocorticoid receptor levels or activity dependent upon foetal sex.Conclusion: Maternal depression and SSRI treatment do not affect the glucocorticoid receptors in the placenta.
Collapse
Affiliation(s)
- Åsa Edvinsson
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Angela Hoyer
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
| | - Malin Hansson
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
| | | | | | - Alkistis Skalkidou
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
| | - Susanne Lager
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
- CONTACT Susanne Lager Department of Women’s and Children’s Health, Uppsala University, Uppsala, 751 85, Sweden
| |
Collapse
|
21
|
Clifton VL, McDonald M, Morrison JL, Holman SL, Lock MC, Saif Z, Meakin A, Wooldridge AL, Gatford KL, Wallace MJ, Muhlhausler BS, Bischof RJ, Moss TJM. Placental glucocorticoid receptor isoforms in a sheep model of maternal allergic asthma. Placenta 2019; 83:33-36. [PMID: 31477204 DOI: 10.1016/j.placenta.2019.06.380] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/20/2019] [Accepted: 06/24/2019] [Indexed: 11/30/2022]
Abstract
Maternal asthma increases the risk of adverse pregnancy outcomes and may affect fetal growth and placental function by differential effects on the expression of glucocorticoid receptor (GR) isoforms, leading to altered glucocorticoid signalling. Our aim was to examine the effect of maternal asthma on placental GR profiles using a pregnant sheep model of asthma. Nine known GR isoforms were detected. There was a significant increase in the expression of placental GR isoforms that are known to have low trans-activational activity in other species including GR A, GR P and GRγ which may result in a pro-inflammatory environment in the presence of allergic asthma.
Collapse
Affiliation(s)
- Vicki L Clifton
- Pregnancy and Development, Mater Research Institute-University of Queensland, Translational Research Institute, South Brisbane, Australia.
| | - Megan McDonald
- Pregnancy and Development, Mater Research Institute-University of Queensland, Translational Research Institute, South Brisbane, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia
| | - Zarqa Saif
- Pregnancy and Development, Mater Research Institute-University of Queensland, Translational Research Institute, South Brisbane, Australia
| | - Ashley Meakin
- Pregnancy and Development, Mater Research Institute-University of Queensland, Translational Research Institute, South Brisbane, Australia
| | - Amy L Wooldridge
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Kathryn L Gatford
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Megan J Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, 3168, Australia
| | - Beverly S Muhlhausler
- Food and Nutrition Research Group, Dept of Food and Wine Science, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Robert J Bischof
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
| | - Timothy J M Moss
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, 3168, Australia
| |
Collapse
|
22
|
Hutter S, Hepp P, Hofmann S, Kuhn C, Messner J, Andergassen U, Mayr D, Emilia Solano M, Obermeier V, Mahner S, Arck P, Jeschke U. Glucocorticoid receptors α and β are modulated sex specifically in human placentas of intrauterine growth restriction (IUGR). Arch Gynecol Obstet 2019; 300:323-335. [PMID: 31089804 DOI: 10.1007/s00404-019-05189-7] [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: 09/18/2018] [Accepted: 05/06/2019] [Indexed: 01/07/2023]
Abstract
PURPOSE The objective of this study was to analyze the expression of the glucocorticoid receptor (GR) subtypes GRα and GRβ in placentas affected by intrauterine growth restriction (IUGR). METHODS We analyzed the sex-specific placental expression of GRα and GRβ in 23 IUGR and 40 control placentas using immunohistochemistry and immunofluorescence. The GR gene, also known as nuclear receptor subfamily 3 group C member 1 (NR3C1), mRNA production in trophoblast-like cell line BeWo after stimulation with prednisolone was analyzed using quantitative polymerase chain reaction (qPCR) and on the protein level using western blot analysis. RESULTS GR subtypes showed a sex-specific upregulation in placentas from IUGR compared to control placentas. An increased expression of GRα was detectable in female placental tissue, whereas GRβ was increased in males. CONCLUSION Our data support previous findings suggesting that the glucocorticoid metabolism plays a role in the pathophysiology of IUGR. Furthermore, the data suggest that the underlying molecular mechanisms differ between male and female cases.
Collapse
Affiliation(s)
- Stefan Hutter
- Department of Gynecology and Obstetrics, LMU Munich, Maistraße 11, 80337, Munich, Germany
| | - Paula Hepp
- Department of Gynecology and Obstetrics, LMU Munich, Maistraße 11, 80337, Munich, Germany
| | - Simone Hofmann
- Department of Gynecology and Obstetrics, LMU Munich, Maistraße 11, 80337, Munich, Germany
| | - Christina Kuhn
- Department of Gynecology and Obstetrics, LMU Munich, Maistraße 11, 80337, Munich, Germany
| | - Julia Messner
- Department of Gynecology and Obstetrics, LMU Munich, Maistraße 11, 80337, Munich, Germany
| | - Ulrich Andergassen
- Department of Gynecology and Obstetrics, LMU Munich, Maistraße 11, 80337, Munich, Germany
| | - Doris Mayr
- Department of Pathology, LMU Munich, Thalkirchner Str. 142, 80337, Munich, Germany
| | - Maria Emilia Solano
- Department of Gynecology and Obstetrics, University of Hamburg, Martinistr. 52, 20246, Hamburg, Germany
| | - Viola Obermeier
- Institute of Social Pediatrics and Adolescent Medicine, LMU Munich, Haydnstr. 5, 80336, Munich, Germany
| | - Sven Mahner
- Department of Gynecology and Obstetrics, LMU Munich, Maistraße 11, 80337, Munich, Germany
| | - Petra Arck
- Department of Gynecology and Obstetrics, University of Hamburg, Martinistr. 52, 20246, Hamburg, Germany
| | - Udo Jeschke
- Department of Gynecology and Obstetrics, LMU Munich, Maistraße 11, 80337, Munich, Germany.
| |
Collapse
|
23
|
Seitz J, Morales-Prieto DM, Favaro RR, Schneider H, Markert UR. Molecular Principles of Intrauterine Growth Restriction in Plasmodium Falciparum Infection. Front Endocrinol (Lausanne) 2019; 10:98. [PMID: 30930847 PMCID: PMC6405475 DOI: 10.3389/fendo.2019.00098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/01/2019] [Indexed: 12/21/2022] Open
Abstract
Malaria in pregnancy still constitutes a particular medical challenge in tropical and subtropical regions. Of the five Plasmodium species that are pathogenic to humans, infection with Plasmodium falciparum leads to fulminant progression of the disease with massive impact on pregnancy. Severe anemia of the mother, miscarriage, stillbirth, preterm delivery and intrauterine growth restriction (IUGR) with reduced birth weight are frequent complications that lead to more than 10,000 maternal and 200,000 perinatal deaths annually in sub-Saharan Africa alone. P. falciparum can adhere to the placenta via the expression of the surface antigen VAR2CSA, which leads to sequestration of infected erythrocytes in the intervillous space. This process induces a placental inflammation with involvement of immune cells and humoral factors. Especially, monocytes get activated and change the release of soluble mediators, including a variety of cytokines. This proinflammatory environment contributes to disorders of angiogenesis, blood flow, autophagy, and nutrient transport in the placenta and erythropoiesis. Collectively, they impair placental functions and, consequently, fetal growth. The discovery that women in endemic regions develop a certain immunity against VAR2CSA-expressing parasites with increasing number of pregnancies has redefined the understanding of malaria in pregnancy and offers strategies for the development of vaccines. The following review gives an overview of molecular processes in P. falciparum infection in pregnancy which may be involved in the development of IUGR.
Collapse
Affiliation(s)
- Johanna Seitz
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | | | - Rodolfo R. Favaro
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Henning Schneider
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- Department of Obstetrics and Gynecology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Udo Rudolf Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| |
Collapse
|
24
|
Wilsterman K, Gotlieb N, Kriegsfeld LJ, Bentley GE. Pregnancy stage determines the effect of chronic stress on ovarian progesterone synthesis. Am J Physiol Endocrinol Metab 2018; 315:E987-E994. [PMID: 30106623 PMCID: PMC6293174 DOI: 10.1152/ajpendo.00183.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although stress-induced glucocorticoid release is thought to be a primary driver by which maternal stress negatively impacts pregnancy outcomes, the downstream neuroendocrine targets mediating these adverse outcomes are less well understood. We hypothesized that stress-induced glucocorticoid secretion inhibits pituitary hormone secretion, resulting in decreased ovarian progesterone synthesis. Using a chronic restraint model of stress in mice, we quantified steroid hormone production, pituitary hormones, and expression of ovarian genes that support progesterone production at both early ( day 5) and midpregnancy ( day 10). Females subjected to daily restraint had elevated baseline glucocorticoids during both early and midpregnancy; however, lower circulating progesterone was observed only during early pregnancy. Lower progesterone production was associated with lower expression of steroidogenic enzymes in the ovary of restrained females during early pregnancy. There were no stress-related changes to luteinizing hormone (LH) or prolactin (PRL). By midpregnancy, circulating LH decreased regardless of treatment, and this was associated with downregulation of ovarian steroidogenic gene expression. Our results are consistent with a role for LH in maintaining steroidogenic enzyme expression in the ovary, but neither circulating PRL nor LH were associated with the stress-induced inhibition of ovarian progesterone production during early pregnancy. We conclude that chronic stress impacts endocrine networks differently in pregnant and nonpregnant mammals. These findings underscore the need for further studies exploring dynamic changes in endocrine networks participating in pregnancy initiation and progression to elucidate the physiological mechanisms that connect stress exposure to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Kathryn Wilsterman
- Department of Integrative Biology, University of California Berkeley, California
| | - Neta Gotlieb
- Department of Psychology, University of California Berkeley, California
| | - Lance J Kriegsfeld
- Department of Psychology, University of California Berkeley, California
- Helen Wills Neuroscience Institute, University of California Berkeley, California
| | - George E Bentley
- Department of Integrative Biology, University of California Berkeley, California
- Helen Wills Neuroscience Institute, University of California Berkeley, California
| |
Collapse
|
25
|
Gross M, Romi H, Gilimovich Y, Drori E, Pinhasov A. Placental glucocorticoid receptor and 11β-hydroxysteroid dehydrogenase-2 recruitment indicates impact of prenatal adversity upon postnatal development in mice. Stress 2018; 21:474-483. [PMID: 29648494 DOI: 10.1080/10253890.2018.1460660] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Prenatal stress may increase concentrations of maternal glucocorticoids, which restrict fetal growth, with variable impact upon postnatal development. Among key regulators of stress hormone effects are the glucocorticoid receptor (GR) and 11β-hydroxysteroid dehydrogenase-2 (11βHSD2), the enzyme that inactivates glucocorticoid. This study utilized mice selectively bred for social dominance (Dom) or submissiveness (Sub), respectively exhibiting resilience or sensitivity to stress, to test whether stress-induced alterations in placental GR and 11βHSD2 protein expression may mediate divergent effects of prenatal adversity upon postnatal development. Pregnant Dom and Sub dams underwent prenatal restraint stress (PRS) for 45 min on gestational days (GD) 15-17. PRS induced a similar spike in serum corticosterone concentrations of dams from each strain on GD15 (p < .001, n = 8), and impaired fetal growth (p < .01, n = 5 litters), although Dom placentae were larger than Sub placentae (p < .01). Among placentae from Dom dams, PRS elevated protein contents of both GR (p < .05, n = 5 litters) and 11βHSD2 (p < .01) on GD19. In contrast, GR contents were reduced among placentae from PRS-exposed Sub mice (p < .01), without changes in 11βHSD2 content. Correspondingly, Dom PRS pup growth recovered by PND14, yet Sub PRS pups remained underweight into adolescence (p < .0001, n = 40 pups). Thus, prenatal stress more strongly increased placental GR and 11βHSD2 levels among Dom mice than in Subs. Increased GR may improve placental function and up-regulate 11βHSD2 expression, protecting fetuses from effects of prenatal stress upon postnatal development. Placental recruitment of GR and 11βHSD2 are potential markers of stress-induced developmental disorders, in accordance with maternal resilience or sensitivity to stress.
Collapse
Affiliation(s)
- Moshe Gross
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Hava Romi
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | | | - Elyashiv Drori
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel, Israel
- Agriculture and Oenology Research Department, Eastern R&D center, Ariel, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Ariel University, Ariel, Israel
| |
Collapse
|
26
|
Tosun G, İnan AH, Kanmaz AG, Biler A, İleri A, Beyan E, Ertas IE. Does fetal sex affect placental delivery times? A prospective observational study. J Matern Fetal Neonatal Med 2018; 33:217-221. [PMID: 29886800 DOI: 10.1080/14767058.2018.1488163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Objective: The aim of this study was to determine the potential effect of fetal sex on placental delivery times.Study design: This was a prospective observational study of term, singleton, and primiparous pregnant women who underwent vaginal delivery and subsequently delivered a phenotypically normal live infant. Women with labor or pregnancy complications and comorbid diseases were excluded. Women with factors who could lengthen the placental delivery time were also excluded. The cohort was divided into two groups according to fetal sex. A total of 299 vaginal deliveries were included, and placental delivery times were analyzed in both groups.Results: There were 3938 vaginal deliveries during the study period. Of these, 150 male-bearing pregnant women and 149 female-bearing pregnant women who met the inclusion criteria were included in the analysis. The mean placental delivery time was significantly longer in the male-bearing group than the female-bearing group (12.20 versus 8.21 min, p = .01). Birth weight was significantly greater in the male-bearing group than the female-bearing group (3194 versus 3059 g, p = .004). There was no significant between-group difference in maternal age, gestational age, and preconception body mass index (BMI).Conclusion: Fetal sex had a significant effect on the placental delivery time in the present study. Fetal sex should be considered in future clinical trials of placental delivery times.
Collapse
Affiliation(s)
- Gökhan Tosun
- Department of Obstetrics and Gynecology, University of Health Sciences, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Abdurrahman Hamdi İnan
- Department of Obstetrics and Gynecology, University of Health Sciences, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Ahkam Göksel Kanmaz
- Department of Obstetrics and Gynecology, University of Health Sciences, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Alper Biler
- Department of Obstetrics and Gynecology, University of Health Sciences, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Alper İleri
- Department of Obstetrics and Gynecology, University of Health Sciences, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Emrah Beyan
- Department of Obstetrics and Gynecology, University of Health Sciences, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Ibrahim Egemen Ertas
- Department of Obstetrics and Gynecology, University of Health Sciences, Tepecik Training and Research Hospital, Izmir, Turkey
| |
Collapse
|
27
|
Hofstee P, McKeating DR, Perkins AV, Cuffe JS. Placental adaptations to micronutrient dysregulation in the programming of chronic disease. Clin Exp Pharmacol Physiol 2018; 45:871-884. [PMID: 29679395 DOI: 10.1111/1440-1681.12954] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/27/2018] [Accepted: 04/09/2018] [Indexed: 12/19/2022]
Abstract
Poor nutrition during pregnancy is known to impair foetal development and increase the risk of chronic disease in offspring. Both macronutrients and micronutrients are required for a healthy pregnancy although significantly less is understood about the role of micronutrients in the programming of chronic disease. This is despite the fact that modern calorie rich diets are often also deficient in key micronutrients. The importance of micronutrients in gestational disorders is clearly understood but how they impact long term disease in humans requires further investigation. In contrast, animal studies have demonstrated how diets high or low in specific micronutrients influence offspring physiology. Many of these studies highlight the importance of the placenta in determining disease risk. This review will explore the effects of individual vitamins, minerals and trace elements on offspring disease outcomes and discuss several key placental adaptations that are affected by multiple micronutrients. These placental adaptations include micronutrient induced dysregulation of oxidative stress, altered methyl donor availability and its impact on epigenetic mechanisms as well as endocrine dysfunction. Critical gaps in our current knowledge and the relative importance of different micronutrients at different gestational ages will also be highlighted. Finally, this review will discuss the need for further studies to characterise the micronutrient status of Australian women of reproductive age and correlate micronutrient status to placental adaptations, pregnancy complications and offspring disease.
Collapse
Affiliation(s)
- Pierre Hofstee
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - Daniel R McKeating
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - Anthony V Perkins
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| | - James Sm Cuffe
- School of Medical Science, Menzies Health Institute Queensland, Griffith University Gold Coast Campus, Southport, QLD, Australia
| |
Collapse
|