1
|
Zhang T, Wang H, Ouyang F, Yang H, Zhang J, Zhang N. Does brain-derived neurotrophic factor play a role in the association between maternal prenatal mental health and neurodevelopment in 2-year-old children? J Affect Disord 2024; 359:171-179. [PMID: 38777264 DOI: 10.1016/j.jad.2024.05.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE To evaluate the role of brain-derived neurotrophic factor (BDNF)-a crucial modulator of neural development and plasticity-in the association between prenatal maternal anxiety, depression, and perceived stress and child neurodevelopment in a prospective cohort study. METHODS We included 526 eligible mother-child pairs from the Shanghai Birth Cohort in the study. Maternal mental health was assessed at mid-pregnancy using Zung's Self-Rating Anxiety Scale, Center for Epidemiologic Studies Depression Scale, and Perceived Stress Scale. The concentration of BDNF in cord blood was measured by ELISA. The offspring neurodevelopment at 24 months of age was assessed using the Bayley Scales. Linear and non-linear regression models were used. RESULTS The average cord blood BDNF levels were higher in female newborns and those born via vaginal delivery, full term, and normal birth weight. Prenatal maternal anxiety (β = -0.32; 95 % CI: -0.55, -0.09), depression (β = -0.30; 95 % CI: -0.52, -0.08), and perceived stress (β = -0.41; 95 % CI: -0.71, -0.12) scores were negatively associated with social-emotional performance at 24 months of age. However, no significant associations were found between prenatal maternal anxiety, depression, or perceived stress at mid-pregnancy and cord blood BDNF levels, as well as between cord blood BDNF levels and child neurodevelopment. LIMITATIONS Maternal mental health at different timepoints during pregnancy and generalizability of the results warrant further assessment. CONCLUSIONS Prenatal mental health was not associated with cord blood BDNF level and that BDNF may not be a mediator in the association between prenatal mental health and child neurodevelopment.
Collapse
Affiliation(s)
- Tian Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huizi Wang
- Hainan Women and Children's Medical Center, Haikou, China
| | - Fengxiu Ouyang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Yang
- Hainan Women and Children's Medical Center, Haikou, China
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Na Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Hoffman MC, Hunter SJ, D'Alessandro A, Christians U, Law AJ, Freedman R. Maternal Plasma Choline during Gestation and Small for Gestational Age Infants. Am J Perinatol 2024; 41:e939-e948. [PMID: 36584689 PMCID: PMC11407527 DOI: 10.1055/s-0042-1759775] [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] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Small for gestational age (SGA) infants are at increased risk for neonatal morbidity and developmental problems in childhood. No current interventions during human pregnancy address this problem. This study investigated the possible relationship between maternal choline concentration during pregnancy and SGA infants. STUDY DESIGN Maternal plasma choline concentrations were sampled at 16 and 28 weeks' gestation from women in a public prenatal clinic. Additional factors assessed were maternal age, body mass index, infection, C-reactive protein, hair cortisol, and compliance with prenatal vitamins and folate. Infants below the 10th percentile for gestational age were classified as SGA. Binary logistic regression was used to identify significant associated factors in pregnancies resulting in SGA infants compared with pregnancies resulting in non-SGA infants. RESULTS Thirteen (8%) of 159 women had SGA infants. Maternal plasma choline concentrations were low for pregnant participants whose infants were SGA, with the 28-week concentration significantly lower compared with other participants. Plasma choline concentrations ≥7 μM at 28 weeks, consistent with a minimally adequate dietary intake of choline-containing foods, were achieved by only 2 (15%) of mothers with SGA infants, compared with 51% of mothers whose infants were not SGA. Choline concentrations <7 μM at 28 weeks' gestation were associated with an odds ratio for SGA of 16.6 (95% confidence interval: 1.5-189.2, p = 0.023). Other significant factors were female sex and maternal C-reactive protein plasma concentration during gestation. CONCLUSION This observational study suggests that higher maternal choline levels may influence the risk for SGA. Maternal plasma choline concentrations are not routinely available in clinical laboratories. However, plasma choline levels can be increased by the mothers' intake of choline or phosphatidylcholine supplements. No nutritional intervention is currently recommended to prevent SGA, but the evidence from this study suggests that further consideration of the role of maternal choline may be warranted. KEY POINTS · More females are small for gestational age.. · Low maternal choline is related to small infants.. · Maternal choline ≥7 μM at 28 weeks appears optimal..
Collapse
Affiliation(s)
- Maria C Hoffman
- Division of Maternal and Fetal Medicine, Departments of Obstetrics and Gynecology and Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| | - Sharon J Hunter
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Uwe Christians
- Department of Anesthesiology, iC42 Clinical Research and Development, University of Colorado School of Medicine, Aurora, Colorado
| | - Amanda J Law
- Department of Psychiatry, Cell and Developmental Biology, and Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Robert Freedman
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
3
|
Nazzari S, Grumi S, Mambretti F, Villa M, Giorda R, Bordoni M, Pansarasa O, Borgatti R, Provenzi L. Sex-dimorphic pathways in the associations between maternal trait anxiety, infant BDNF methylation, and negative emotionality. Dev Psychopathol 2024; 36:908-918. [PMID: 36855816 DOI: 10.1017/s0954579423000172] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Maternal antenatal anxiety is an emerging risk factor for child emotional development. Both sex and epigenetic mechanisms, such as DNA methylation, may contribute to the embedding of maternal distress into emotional outcomes. Here, we investigated sex-dependent patterns in the association between antenatal maternal trait anxiety, methylation of the brain-derived neurotrophic factor gene (BDNF DNAm), and infant negative emotionality (NE). Mother-infant dyads (N = 276) were recruited at delivery. Maternal trait anxiety, as a marker of antenatal chronic stress exposure, was assessed soon after delivery using the Stait-Trait Anxiety Inventory (STAI-Y). Infants' BDNF DNAm at birth was assessed in 11 CpG sites in buccal cells whereas infants' NE was assessed at 3 (N = 225) and 6 months (N = 189) using the Infant Behavior Questionnaire-Revised (IBQ-R). Hierarchical linear analyses showed that higher maternal antenatal anxiety was associated with greater 6-month-olds' NE. Furthermore, maternal antenatal anxiety predicted greater infants' BDNF DNAm in five CpG sites in males but not in females. Higher methylation at these sites was associated with greater 3-to-6-month NE increase, independently of infants' sex. Maternal antenatal anxiety emerged as a risk factor for infant's NE. BDNF DNAm might mediate this effect in males. These results may inform the development of strategies to promote mothers and infants' emotional well-being.
Collapse
Affiliation(s)
- Sarah Nazzari
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Serena Grumi
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabiana Mambretti
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Marco Villa
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Roberto Giorda
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Matteo Bordoni
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Orietta Pansarasa
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Renato Borgatti
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Livio Provenzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
4
|
Chen Y, Wang H. The changes in adrenal developmental programming and homeostasis in offspring induced by glucocorticoids exposure during pregnancy. VITAMINS AND HORMONES 2024; 124:463-490. [PMID: 38408809 DOI: 10.1016/bs.vh.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Clinically, synthetic glucocorticoids are often used to treat maternal and fetal related diseases, such as preterm birth and autoimmune diseases. Although its clinical efficacy is positive, it will expose the fetus to exogenous glucocorticoids. Adverse environments during pregnancy (e.g., exogenous glucocorticoids exposure, malnutrition, infection, hypoxia, and stress) can lead to fetal overexposure to endogenous maternal glucocorticoids. Basal glucocorticoids levels in utero are crucial in determining fetal tissue maturation and its postnatal fate. As the synthesis and secretion organ of glucocorticoids, the adrenal development is crucial for the growth and development of the body. Studies have found that glucocorticoids exposure during pregnancy could cause abnormal fetal adrenal development, which could last after birth or even adulthood. As the key organ of fetal-originated adult disease, the adrenal developmental programming has a profound impact on the health of offspring, which can lead to many chronic diseases in adulthood. However, the aberrant adrenal development in offspring caused by glucocorticoids exposure during pregnancy and its intrauterine programming mechanism have not been systematically clarified. Therefore, this review summarizes recent research progress on the short and long-term hazards of aberrant adrenal development induced by glucocorticoids exposure during pregnancy, which is of great significance for the analysis of aberrant adrenal development and clarify the intrauterine origin mechanism of fetal-originated adult disease.
Collapse
Affiliation(s)
- Yawen Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China; Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, P.R. China.
| |
Collapse
|
5
|
Volqvartz T, Andersen HHB, Pedersen LH, Larsen A. Obesity in pregnancy-Long-term effects on offspring hypothalamic-pituitary-adrenal axis and associations with placental cortisol metabolism: A systematic review. Eur J Neurosci 2023; 58:4393-4422. [PMID: 37974556 DOI: 10.1111/ejn.16184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Obesity, affecting one in three pregnant women worldwide, is not only a major obstetric risk factor. The resulting low-grade inflammation may have a long-term impact on the offspring's HPA axis through dysregulation of maternal, placental and fetal corticosteroid metabolism, and children born of obese mothers have increased risk of diabetes and cardiovascular disease. The long-term effects of maternal obesity on offspring neurodevelopment are, however, undetermined and could depend on the specific effects on placental and fetal cortisol metabolism. This systematic review evaluates how maternal obesity affects placental cortisol metabolism and the offspring's HPA axis. Pubmed, Embase and Scopus were searched for original studies on maternal BMI, obesity, and cortisol metabolism and transfer. Fifteen studies were included after the screening of 4556 identified records. Studies were small with heterogeneous exposures and outcomes. Two studies found that maternal obesity reduced placental HSD11β2 activity. In one study, umbilical cord blood cortisol levels were affected by maternal BMI. In three studies, an altered cortisol response was consistently seen among offspring in childhood (n = 2) or adulthood (n = 1). Maternal BMI was not associated with placental HSD11β1 or HSD11β2 mRNA expression, or placental HSD11β2 methylation. In conclusion, high maternal BMI is associated with reduced placental HSD11β2 activity and a dampened cortisol level among offspring, but the data is sparse. Further investigations are needed to clarify whether the HPA axis is affected by prenatal factors including maternal obesity and investigate if adverse effects can be ameliorated by optimising the intrauterine environment.
Collapse
Affiliation(s)
- Tabia Volqvartz
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Lars Henning Pedersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, Aarhus, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
6
|
Ushida T, Nakatochi M, Kobayashi Y, Nakamura N, Fuma K, Iitani Y, Imai K, Sato Y, Hayakawa M, Kajiyama H, Kotani T. Antenatal corticosteroids and outcomes of small for gestational age infants born at 24-31 gestational weeks: a population-based propensity score matching analysis. Arch Gynecol Obstet 2023; 308:1463-1471. [PMID: 36352162 DOI: 10.1007/s00404-022-06834-4] [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: 08/29/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE To evaluate the effect of antenatal corticosteroid (ACS) treatment on neonatal outcomes in small for gestational age (SGA) infants born at 24-31 gestational weeks compared with non-SGA infants. METHODS A population-based retrospective study was conducted that analyzed clinical data from the Neonatal Research Network of Japan database, which enrolls neonates born at < 32 gestational weeks and weighing 1500 g or less (n = 22,414). Propensity score matching (with the ratio of ACS to no-ACS groups of 1:1) was performed in SGA (n = 7028) and non-SGA (n = 15,386) infants, respectively. Univariate logistic and interaction analyses were performed to compare the short-term neonatal outcomes of infants with and without ACS treatment in utero. RESULTS In the SGA and non-SGA infants, ACS treatment significantly reduced in-hospital mortality (odds ratio 0.67 95% confidence interval [0.50-0.88] and 0.62 [0.50-0.78], respectively), respiratory distress syndrome (0.77 [0.69-0.87] and 0.63 [0.58-0.68], respectively), and composite adverse outcomes (0.73 [0.58-0.91] and 0.57 [0.50-0.65], respectively). ACS treatment also significantly reduced intraventricular hemorrhage (grade III/IV), periventricular leukomalacia, and sepsis in the non-SGA infants, but not in the SGA infants. However, interaction analyses revealed no significant differences between the SGA and non-SGA infants in the efficacy of ACS treatment on short-term outcomes except for respiratory distress syndrome. CONCLUSIONS ACS treatment was associated with beneficial effects on mortality, respiratory distress syndrome, and adverse composite outcomes in extremely and very preterm SGA infants, with similar efficacy on all neonatal outcomes except for respiratory distress syndrome observed in the non-SGA infants.
Collapse
Affiliation(s)
- Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan.
- Division of Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yumiko Kobayashi
- Data Science Division, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Noriyuki Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Kazuya Fuma
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yukako Iitani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Division of Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
7
|
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
|
8
|
Dufour J, Sabry R, Khokhar JY, Favetta LA. Delta-9 tetrahydrocannabinol (THC) effects on the cortisol stress response in bovine granulosa cells. Toxicol In Vitro 2023; 88:105549. [PMID: 36596389 DOI: 10.1016/j.tiv.2022.105549] [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: 08/17/2022] [Revised: 12/01/2022] [Accepted: 12/30/2022] [Indexed: 01/02/2023]
Abstract
Maternal stress can result in changes in the hypothalamic-pituitary-adrenal (HPA) axis and lead to stress-related behaviours in offspring. Under physiological conditions, delta-9 tetrahydrocannabinol (THC) appears to be detrimental for fertility. However, cannabis is also commonly used for stress-relief. THC acts on the endocannabinoid receptors in granulosa cells (GCs), which affect oocyte competency. The objective of this study was to evaluate the effects of THC on in vitro bovine granulosa cell viability, apoptosis, and stress response pathway. GCs were cultured in vitro in the presence of clinically relevant therapeutic and recreational plasma doses of THC. Cortisol doses reflecting normal and elevated plasma levels were used to evaluate the effects of THC under induced stress in vitro. No effect of THC was observed on cell viability or apoptosis. High and low cortisol concentrations caused significant increases in 11β-HSD1 mRNA expression (n = 6, p < 0.0001). Interestingly, when combined with high [THC], there was a significant decrease in 11β-HSD1 expression compared to high and low cortisol treatments alone (p < 0.001, p < 0.05). GR expression was unaffected by cortisol treatments, and low [THC] treatment maintained increased expression in the presence of high and low cortisol treatments (n = 6, p < 0.01, p < 0.0001). Our findings represent a foundation to obtain useful data for evaluating THC potential therapeutic benefit.
Collapse
Affiliation(s)
- Jaustin Dufour
- Reproductive Health and Biotechnology Laboratory, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Reem Sabry
- Reproductive Health and Biotechnology Laboratory, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jibran Y Khokhar
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - Laura A Favetta
- Reproductive Health and Biotechnology Laboratory, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
9
|
Lu Z, Guo Y, Xu D, Xiao H, Dai Y, Liu K, Chen L, Wang H. Developmental toxicity and programming alterations of multiple organs in offspring induced by medication during pregnancy. Acta Pharm Sin B 2023; 13:460-477. [PMID: 36873163 PMCID: PMC9978644 DOI: 10.1016/j.apsb.2022.05.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/05/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022] Open
Abstract
Medication during pregnancy is widespread, but there are few reports on its fetal safety. Recent studies suggest that medication during pregnancy can affect fetal morphological and functional development through multiple pathways, multiple organs, and multiple targets. Its mechanisms involve direct ways such as oxidative stress, epigenetic modification, and metabolic activation, and it may also be indirectly caused by placental dysfunction. Further studies have found that medication during pregnancy may also indirectly lead to multi-organ developmental programming, functional homeostasis changes, and susceptibility to related diseases in offspring by inducing fetal intrauterine exposure to too high or too low levels of maternal-derived glucocorticoids. The organ developmental toxicity and programming alterations caused by medication during pregnancy may also have gender differences and multi-generational genetic effects mediated by abnormal epigenetic modification. Combined with the latest research results of our laboratory, this paper reviews the latest research progress on the developmental toxicity and functional programming alterations of multiple organs in offspring induced by medication during pregnancy, which can provide a theoretical and experimental basis for rational medication during pregnancy and effective prevention and treatment of drug-related multiple fetal-originated diseases.
Collapse
Affiliation(s)
- Zhengjie Lu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Dan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Hao Xiao
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Kexin Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Liaobin Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| |
Collapse
|
10
|
Sze Y, Fernandes J, Kołodziejczyk ZM, Brunton PJ. Maternal glucocorticoids do not directly mediate the effects of maternal social stress on the fetus. J Endocrinol 2022; 255:143-158. [PMID: 36256689 PMCID: PMC9716396 DOI: 10.1530/joe-22-0226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
Stress during pregnancy negatively affects the fetus and increases the risk for affective disorders in adulthood. Excess maternal glucocorticoids are thought to mediate fetal programming; however, whether they exert their effects directly or indirectly remains unclear. During pregnancy, protective mechanisms including maternal hypothalamic-pituitary-adrenal (HPA) axis hyporesponsiveness and placental 11β-hydroxysteroid dehydrogenase (11βHSD) type 2, which inactivates glucocorticoids, limit mother-to-fetus glucocorticoid transfer. However, whether repeated stress negatively impacts these mechanisms is not known. Pregnant rats were exposed to repeated social stress on gestational days (GD) 16-20 and several aspects of HPA axis and glucocorticoid regulation, including concentrations of glucocorticoids, gene expression for their receptors (Nr3c1, Nr3c2), receptor chaperones (Fkbp51, Fkbp52) and enzymes that control local glucocorticoid availability (Hsd11b1, Hsd11b2), were investigated in the maternal, placental and fetal compartments on GD20. The maternal HPA axis was activated following stress, though the primary driver was vasopressin, rather than corticotropin-releasing hormone. Despite the stress-induced increase in circulating corticosterone in the dams, only a modest increase was detected in the circulation of female fetuses, with no change in the fetal brain of either sex. Moreover, there was no change in the expression of genes that mediate glucocorticoid actions or modulate local concentrations in the fetal brain. In the placenta labyrinth zone, stress increased Hsd11b2 expression only in males and Fkbp51 expression only in females. Our results indicate that any role glucocorticoids play in fetal programming is likely indirect, perhaps through sex-dependent alterations in placental gene expression, rather than exerting effects via direct crossover into the fetal brain.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Joana Fernandes
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | | | - Paula J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
- Zhejiang University-University of Edinburgh Institute, International Campus, Haining, Zhejiang, P.R. China
- Correspondence should be addressed to P J Brunton:
| |
Collapse
|
11
|
Musana J, Cohen CR, Kuppermann M, Gerona R, Wanyoro A, Aguilar D, Santos N, Temmerman M, Weiss SJ. Obstetric risk in pregnancy interacts with hair cortisone levels to reduce gestational length. Front Glob Womens Health 2022; 3:878538. [PMID: 35936818 PMCID: PMC9354598 DOI: 10.3389/fgwh.2022.878538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/30/2022] [Indexed: 11/29/2022] Open
Abstract
Background Maternal psychological stress has been linked to preterm birth. However, the differential contribution of psychological stress versus stress hormones is not clear. Studies focus primarily on perceived stress and cortisol, with few assessing its inter-convertible hormone cortisone. Furthermore, little is known about the potential moderating roles of obstetric risk and fetal sex in the relationship between maternal stress and gestational length. This gap in knowledge is particularly evident for rural women who typically experience chronic multiple stressors during pregnancy. We explored the relationship of hormonal and psychological stress to gestational length and the effects of obstetric risks and fetal sex on this relationship among Kenyan pregnant women. Methods The sample included 130 women recruited between 22 to 28 weeks gestation. They completed a clinical and sociodemographic questionnaire together with the Perceived Stress Scale and provided a hair sample for cortisol and cortisone assay. Women underwent an ultrasound to assess weeks of gestation. At delivery, their pregnancy-related health problems were identified using information extracted from medical records to compile each woman's number of pregnancy risks on the Obstetric Medical Risk Index (OMRI). Results Perceived stress and hair cortisol were not significant predictors of gestational length. However, a greater number of obstetric risks on the OMRI was associated with shorter gestational length. This effect was further explained by the interaction between obstetric risk and hair cortisone (B = 0.709, p = 0.02). Hair cortisone levels of mothers who had a shorter gestation were significantly higher in mothers with 2 or more risks on the OMRI but not among mothers with only one or no risks (t = 2.39, p = 0.02). Fetal sex had no relationship to gestational length and also had no moderating effect on the relationship between any stress-related metric and gestational length. Conclusion Cortisone levels may increase in anticipation of shorter gestation as a compensatory response to increased obstetric risk. Elevated cortisone may be a more sensitive marker of risk for early delivery than cortisol or psychological stress, with salience for both the male and female fetus.
Collapse
Affiliation(s)
- Joseph Musana
- Department of Obstetrics and Gynaecology, Aga Khan University Hospital, Nairobi, Kenya
- *Correspondence: Joseph Musana
| | - Craig R. Cohen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Miriam Kuppermann
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Roy Gerona
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Anthony Wanyoro
- Department of Obstetrics and Gynaecology, Kenyatta University, Nairobi, Kenya
| | - David Aguilar
- Clinical Toxicology and Environmental Biomonitoring Lab, University of California, San Francisco, San Francisco, CA, United States
| | - Nicole Santos
- Institute for Global Health Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Marleen Temmerman
- Department of Obstetrics and Gynaecology, Aga Khan University Hospital, Nairobi, Kenya
| | - Sandra J. Weiss
- Department of Community Health Systems, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
12
|
Associations between maternal awakening salivary cortisol levels in mid-pregnancy and adverse birth outcomes. Arch Gynecol Obstet 2022; 306:1989-1999. [PMID: 35320387 DOI: 10.1007/s00404-022-06513-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/06/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Elevated levels of maternal cortisol have been hypothesized as the intermediate process between symptoms of depression and psychosocial stress during pregnancy and adverse birth outcomes. Therefore, we examined associations between cortisol levels in the second trimester of pregnancy and risks of three common birth outcomes in a nested case-control study. METHODS This study was embedded in the PRIDE Study (n = 3,019), from which we selected all cases with preterm birth (n = 64), low birth weight (n = 49), and small-for-gestational age (SGA; n = 65), and 260 randomly selected controls, among the participants who provided a single awakening saliva sample in approximately gestational week 19 in 2012-2016. Multivariable linear and logistic regression was performed to assess the associations between continuous and categorized cortisol levels and the selected outcomes. RESULTS We did not observe any associations between maternal cortisol levels and preterm birth and low birth weight. However, high cortisol levels (≥ 90th percentile) seemed to be associated with SGA (adjusted odds ratio 2.1, 95% confidence interval 0.9-4.8), in particular among girls (adjusted odds ratio 3.7, 95% confidence interval 1.1-11.9, based on eight exposed cases) in an exploratory analysis. CONCLUSION The results of this study showed no suggestions of associations between maternal awakening cortisol levels in mid-pregnancy and adverse birth outcomes, except for an increased risk of SGA.
Collapse
|
13
|
Prenatal ethanol exposure induces dynamic changes of expression and activity of hepatic cytochrome P450 isoforms in male rat offspring. Reprod Toxicol 2022; 109:101-108. [PMID: 35301062 DOI: 10.1016/j.reprotox.2022.03.002] [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: 06/27/2021] [Revised: 03/05/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023]
Abstract
This study aimed at determining the effect of prenatal ethanol exposure (PEE) on the expression and activity of cytochrome P450 (CYP) isozymes at different life stages of male rat offspring. Pregnant Wistar rats were administered with ethanol (4 g/kg/d) intragastrically from gestational day (GD) 9-20. Male offspring's gene and activity of CYP isozymes were analyzed on GD 20 (only expression), postnatal day (PD) 84 and 196. Using aniline as probe, we compared the enzyme kinetics of hepatic CYP2E1 between two groups. Expression of CYP isozymes was examined in rat primary hepatocytes and human hepatic cell lines treated with ethanol or/and glucocorticoid. Gene level of Cyp1a2, 2b1, 2d1, 2e1, 3a1 and aryl hydrocarbon receptor were increased in PEE group on GD 20 and PD 84 and Cyp2e1 still exhibited an increasing trend on PD 196 compared with the control. PEE inhibited CYP2D1 and 2E1 activities in male offspring on PD 84. CYP activities in two groups became the same level on PD 196. PEE induced an opposite change in gene and protein level of hepatic CYP2E1 before and after birth. In consistent with lower protein level, aniline metabolism in PEE was weaker in liver microsome. Both single and combined use of ethanol or/and glucocorticoid increased CYPs expression in vitro. In conclusion, PEE programmed a higher gene and lower protein level of CYPs in male offspring, which dwindled with age. Impairment of protein levels and enzyme activities of CYPs may affect individual metabolism of endogenous and exogenous substances in early adulthood.
Collapse
|
14
|
Maternal Signatures of Cortisol in First Trimester Small-for-Gestational Age. Reprod Sci 2022; 29:1498-1505. [PMID: 35001327 DOI: 10.1007/s43032-021-00822-w] [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: 07/06/2021] [Accepted: 12/03/2021] [Indexed: 10/19/2022]
Abstract
The objective of this study was to identify predictable maternal serum signatures of cortisol metabolism during the first trimester of women who are expected to deliver small-for-gestational-age (SGA) neonates. This prospective cohort study included 112 pregnant women (with and without SGA, n = 56 each). Maternal serum samples were collected at 10-14 gestational weeks to quantify the levels of cortisol and its precursors and metabolites by liquid chromatography-mass spectrometry. Increased maternal serum levels of tetrahydrocortisol (11.82 ± 8.16 ng/mL vs. 7.51 ± 2.90 ng/mL, P < 0.005) and decreased 21-deoxycortisol (2.98 ± 1.36 ng/mL vs. 4.33 ± 2.06 ng/mL, P < 0.0001) were observed in pregnant women carrying SGA fetus. In conjunction with individual steroid levels, metabolic ratios corresponding to the activity of related enzymes were calculated. In addition to increased tetrahydrocortisol/cortisol ratio (P < 0.006), the SGA group showed a significant increase in the two metabolic ratios including cortisol/11-deoxycortisol (P < 0.03) and cortisol/21-deoxycortisol (P < 0.0003). The receiver operating characteristic (ROC) curve generated in combination with three variables of 21-deoxycortisol concentration and two metabolic ratios of cortisol/21-deoxycortisol and tetrahydrocortisol/cortisol resulted in an area under the ROC curve = 0.824 (95% confidence interval, 0.713-0.918). A significant decrease in maternal serum levels of 21-deoxycortisol and an increase in two metabolic ratios of cortisol/21-deoxycortisol and tetrahydrocortisol/cortisol, indicating cortisol biosynthetic rate, represent potential biomarkers for the prediction of SGA in the first trimester.
Collapse
|
15
|
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: 82] [Impact Index Per Article: 20.5] [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
|
16
|
Stoye DQ, Sullivan G, Galdi P, Kirschbaum C, Lamb GJ, Black GS, Evans MJ, Boardman JP, Reynolds RM. Perinatal determinants of neonatal hair glucocorticoid concentrations. Psychoneuroendocrinology 2021; 128:105223. [PMID: 33878601 PMCID: PMC8155393 DOI: 10.1016/j.psyneuen.2021.105223] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/21/2021] [Accepted: 04/05/2021] [Indexed: 12/15/2022]
Abstract
Adult hair glucocorticoid concentrations reflect months of hypothalamic-pituitary-adrenal axis activity. However, little is known about the determinants of neonatal hair glucocorticoids. We tested associations between perinatal exposures and neonatal hair glucocorticoids. Cortisol and cortisone were measured by LC-MS/MS in paired maternal and infant hair samples collected within 10 days of birth (n = 49 term, n = 47 preterm), with neonatal samples collected at 6-weeks in n = 54 preterm infants. We demonstrate cortisol accumulation in hair increases with fetal maturity, with hair cortisol being higher in term than preterm born infants after delivery (median 401 vs 106 pg/mg; p < 0.001). In term born infants, neonatal hair cortisol is positively associated with maternal hair cortisol concentration (β = 0.240, p = 0.045) and negatively associated with birthweight z-score (β = -0.340, p = 0.006). Additionally, being born without maternal labour is associated with lower hair cortisol concentrations (β = -0.489, p < 0.001) and a lower ratio of cortisol to cortisone (β = -0.484, p = 0.001). In preterm infants, histological chorioamnionitis is associated with a higher cortisol to cortisone ratio in hair (β = 0.459, p = 0.001). In samples collected 6 weeks after preterm birth, hair cortisol concentration is associated with cortisol hair concentrations measured after birth (β = 0.523, p < 0.001), chorioamnionitis (β = 0.250, p = 0.049) and postnatal exposures including intravenous hydrocortisone therapy (β = 0.343, p < 0.007) and neonatal sepsis (β = 0.290, p = 0.017). In summary, neonatal hair cortisol is associated with birth gestation, maternal hair cortisol concentration and fetal growth. Additionally, exposures at delivery are important determinants of hair cortisol, and should be considered in the design of future research investigating how neonatal hair cortisol relates to prenatal exposures or fetal development.
Collapse
Affiliation(s)
- David Q Stoye
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Gemma Sullivan
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Paola Galdi
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | | - Gillian J Lamb
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Gill S Black
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Margaret J Evans
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - James P Boardman
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Rebecca M Reynolds
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK; Centre for Cardiovascular Science, University of Edinburgh, UK.
| |
Collapse
|
17
|
Qi Y, Li B, Wen Y, Yang X, Chen B, He Z, Zhao Z, Magdalou J, Wang H, Chen L. H3K9ac of TGFβRI in human umbilical cord: a potential biomarker for evaluating cartilage differentiation and susceptibility to osteoarthritis via a two-step strategy. Stem Cell Res Ther 2021; 12:163. [PMID: 33663609 PMCID: PMC7934528 DOI: 10.1186/s13287-021-02234-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/15/2021] [Indexed: 11/10/2022] Open
Abstract
Background Epidemiological investigation and our previous reports indicated that osteoarthritis had a fetal origin and was closely associated with intrauterine growth retardation (IUGR). Human Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) could be programmable to “remember” early-life stimuli. Here, we aimed to explore an early-warning biomarker of fetal-originated adult osteoarthritis in the WJ-MSCs. Methods Firstly, two kinds of WJ-MSCs were applied to evaluate their chondrogenic potential in vitro through inducing chondrogenic differentiation as the first step of our strategy, one from newborns with IUGR and the other from normal newborns but treated with excessive cortisol during differentiation to simulate the excessive maternal glucocorticoid in the IUGR newborns. As for the second step of the strategy, the differentiated WJ-MSCs were treated with interleukin 1β (IL-1β) to mimic the susceptibility to osteoarthritis. Then, the expression and histone acetylation levels of transforming growth factor β (TGFβ) signaling pathway and the expression of histone deacetylases (HDACs) were quantified, with or without cortisol receptor inhibitor RU486, or HDAC4 inhibitor LMK235. Secondly, the histone acetylation and expression levels of TGFβRI were further detected in rat cartilage and human umbilical cord from IUGR individuals. Results Glycosaminoglycan content and the expression levels of chondrogenic genes were decreased in the WJ-MSCs from IUGR, and the expression levels of chondrogenic genes were further reduced after IL-1β treatment, while the expression levels of catabolic factors were increased. Then, serum cortisol level from IUGR individuals was found increased, and similar changes were observed in normal WJ-MSCs treated with excessive cortisol. Moreover, the decreased histone 3 lysine 9 acetylation (H3K9ac) level of TGFβRI and its expression were observed in IUGR-derived WJ-MSCs and normal WJ-MSCs treated with excessive cortisol, which could be abolished by RU486 and LMK235. At last, the decreased H3K9ac level of TGFβRI and its expression were further confirmed in the cartilage of IUGR rat offspring and human umbilical cords from IUGR newborn. Conclusions WJ-MSCs from IUGR individuals displayed a poor capacity of chondrogenic differentiation and an increased susceptibility to osteoarthritis-like phenotype, which was attributed to the decreased H3K9ac level of TGFβRI and its expression induced by high cortisol through GR/HDAC4. The H3K9ac of TGFβRI in human umbilical cord could be a potential early-warning biomarker for predicting neonatal cartilage dysplasia and osteoarthritis susceptibility. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02234-8.
Collapse
Affiliation(s)
- Yongjian Qi
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Bin Li
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yinxian Wen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Xu Yang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Biao Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Zheng He
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Zhe Zhao
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jacques Magdalou
- UMR 7561 CNRS-Université de Lorraine, Faculté de Médicine, Vandoeuvre-lès-Nancy, France
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China. .,Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| | - Liaobin Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China. .,Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
18
|
Jahnke JR, Terán E, Murgueitio F, Cabrera H, Thompson AL. Maternal stress, placental 11β-hydroxysteroid dehydrogenase type 2, and infant HPA axis development in humans: Psychosocial and physiological pathways. Placenta 2020; 104:179-187. [PMID: 33360746 DOI: 10.1016/j.placenta.2020.12.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/24/2020] [Accepted: 12/15/2020] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Prenatal stress is known to influence fetal hypothalamic-pituitary-adrenal axis (HPA axis) development. Placental 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2) is a central gene in this pathway, but little is known about what influences its functioning. We assess how maternal distress influences HSD11B2 functioning, and how HSD11B2 in turn, is associated with infant HPA axis development. METHODS Data come from 24 mother-infant dyads on the Galápagos Islands. Using adjusted linear regression models, we assess the effects of maternal psychosocial (stress and depressive symptoms, measured by the Perceived Stress Scale and the Patient Health Questionnaire-8, respectively) and physiological (HPA axis dysregulation) distress during pregnancy on HSD11B2 methylation and expression and then test how these HSD11B2 measures influence infant HPA axis development. RESULTS Maternal HPA axis dysregulation during pregnancy is associated with lower placental HSD11B2 expression, which is associated with an exaggerated cortisol reactivity in infants. Sex-specific analyses revealed that maternal depressive symptoms may influence the functioning of placental HSD11B2 differently in girls (n = 11, 46%) than in boys (n = 13, 54%), though the sample size was small. DISCUSSION These results support a disrupted adaptive framework, in which the ability to upregulate HSD11B2 expression in response to acute stress diminishes as maternal stress becomes chronic. In this model, chronic stress may exhaust the protective mechanism of HSD11B2, leaving the infant vulnerable to high levels of maternal cortisol, which could injure the fetal HPA axis and disrupt long-term neurobehavioral and metabolic development. While larger studies will be needed to confirm these findings, this study offers exploratory results on the effects of maternal distress on both HSD11B2 methylation and expression and the effect of HSD11B2 on offspring HPA axis development.
Collapse
Affiliation(s)
- Johanna R Jahnke
- Department of Anthropology, 301 Alumni Building CB #3115, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA; Carolina Population Center, 123 West Franklin Street, Chapel Hill, NC, 27516, USA.
| | - Enrique Terán
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito. Hospital de los Valles, Ed. Especialidades Médicas, PB, Av. Interoceánica Km. 12 ½; y Av. Florencia, Casilla Postal 17-1200-841, Quito, Ecuador; Galapagos Science Center. Avenida Alsacio Northia, Puerto Baquerizo Moreno, Ecuador San Cristobal, Galapagos Archipelago, Ecuador.
| | - Francisca Murgueitio
- Hospital Oskar Jandl, Ministerio de Salud Publica. San Cristóbal, Galápagos Archipiélago, Ecuador.
| | - Holger Cabrera
- Hospital Oskar Jandl, Ministerio de Salud Publica. San Cristóbal, Galápagos Archipiélago, Ecuador.
| | - Amanda L Thompson
- Department of Anthropology, 301 Alumni Building CB #3115, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA; Carolina Population Center, 123 West Franklin Street, Chapel Hill, NC, 27516, USA; Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill 170 Rosenau Hall, CB #7400, 135 Dauer Drive, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
19
|
Xu R, Zhu Z, Tang W, Zhou Q, Zeng S. Inferior Adrenal Artery PI in Fetuses with IUGR: Value Indicating Early Blood Redistribution and Steroidogenic Response. J Clin Endocrinol Metab 2020; 105:5908703. [PMID: 32946562 DOI: 10.1210/clinem/dgaa610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/16/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To characterize the inferior adrenal artery (IAA) pulsatility index (PI) in intrauterine growth-restricted (IUGR) fetuses without brain sparing. METHODS Twenty-three IUGR fetuses with a normal Doppler cerebroplacental ratio (CPR) and 23 normal controls were included in this prospective cross-sectional study. The PI of the IAA was recorded using routine transabdominal Doppler ultrasound. The differences in Doppler characteristics, perinatal outcomes, and steroidogenesis in the umbilical vein at birth (adrenocorticotropic hormone [ACTH] and cortisol [F] levels) were compared between the 2 groups. The correlations between IAA-PI and steroidogenesis were assessed in the IUGR group. RESULTS IAA-PI was significantly lower in IUGR fetuses than in normal controls (0.85 vs 1.18 at first scan, 0.78 vs 0.92 at last scan; both P < 0.001). The plasma F and ACTH levels in IUGR cases were significantly higher than those of the normal controls (18.2 vs 12.4 µg/dL and 280.5 vs 125.6 pg/mL for F and ACTH, respectively; both P < 0.001). There were negative correlations between IAA-PI and plasma F values and between IAA-PI and ACTH values in the IUGR group (r = -0.774 and -0.82 at first scan, r = -0.525 and -0.45 at last scan, respectively; P < 0.001). CONCLUSION Increased adrenal gland blood flow with concomitant increases in ACTH and F levels were observed in IUGR fetuses. IAA-PI is useful to assess early blood redistribution and may be beneficial for evaluating the steroidogenic response in high-risk pregnancies.
Collapse
Affiliation(s)
- Ran Xu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziling Zhu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenjuan Tang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qichang Zhou
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shi Zeng
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
Shi XT, Zhu HL, Xiong YW, Liu WB, Zhou GX, Cao XL, Yi SJ, Dai LM, Zhang C, Gao L, Xu DX, Wang H. Cadmium down-regulates 11β-HSD2 expression and elevates active glucocorticoid level via PERK/p-eIF2α pathway in placental trophoblasts. CHEMOSPHERE 2020; 254:126785. [PMID: 32334250 DOI: 10.1016/j.chemosphere.2020.126785] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/30/2020] [Accepted: 04/12/2020] [Indexed: 06/11/2023]
Abstract
Fetal overexposure to active glucocorticoid (GC) is the major cause for fetal growth restriction (FGR). This study investigated the influences of cadmium (Cd) exposure on active GC and its mechanism in placental trophoblasts. Pregnant mice were exposed to CdCl2 (4.5 mg/kg, i.p.). Human JEG-3 cells were treated with CdCl2 (0-20 μM). Prenatal Cd exposure significantly increased active GC level in amniotic fluid and placenta. Similarly, Cd treatment also elevated active GC level in medium. Expectedly, the expression of 11β-HSD2 protein was markedly downregulated in Cd-exposed placental trophoblasts. We further found that Cd activated the PERK/p-eIF2α signaling pathway in placental trophoblasts. Mechanistically, PERK siRNA pretreatment completely blocked PERK/p-eIF2α signaling, and thereby restoring Cd-downregulated 11β-HSD2 protein expression in human placental trophoblasts. We further found that N-acetylcysteine, a well-known antioxidant, obviously reversed Cd-downregulated 11β-HSD2 protein expression by inhibiting p-PERK/p-eIF2α signaling in placental trophoblasts. Overall, our data suggest that Cd activates the PERK/p-eIF2α signaling, down-regulates the protein expression of 11β-HSD2, and thereby elevating active GC level in placental trophoblast.
Collapse
Affiliation(s)
- Xue-Ting Shi
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Hua-Long Zhu
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Yong-Wei Xiong
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Wei-Bo Liu
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Guo-Xiang Zhou
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Xue-Lin Cao
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Song-Jia Yi
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Li-Min Dai
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Lan Gao
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China.
| |
Collapse
|
21
|
Freedman R, Hunter SK, Noonan K, Wyrwa A, Christians U, Law AJ, Hoffman MC. Maternal Prenatal Depression in Pregnancies With Female and Male Fetuses and Developmental Associations With C-reactive Protein and Cortisol. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 6:310-320. [PMID: 33060035 DOI: 10.1016/j.bpsc.2020.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/01/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Prenatal depression has lasting effects on development in offspring, including later mental illness risk. Maternal responses to depression include inflammation and hypothalamic-pituitary-adrenal axis stimulation. Effects on development of cerebral inhibitory neurocircuits may differ for female and male fetuses. METHODS Mothers (N = 181) were assessed periodically, beginning at 16 weeks' gestation, using the Center for Epidemiologic Studies-Depression Scale. Maternal prenatal C-reactive protein and hair cortisol and cortisone levels were determined. Cortisone was determined in neonatal hair. Development of cerebral inhibitory neurocircuits was assessed in 162 1-month-old newborns by inhibition of P50 electrophysiological responses to repeated sounds. RESULTS Maternal depression was associated with decreased newborn P50 inhibition in both sexes. Maternal C-reactive protein levels were significantly associated with depression only in pregnancies with male fetuses and with decreased newborn P50 inhibition only in male newborns. Maternal cortisol levels were significantly associated with depression only in pregnancies with female fetuses and with decreased newborn P50 inhibition only in female newborns. In pregnancies with male fetuses compared with pregnancies with female fetuses, cortisol was more robustly metabolized to cortisone, which does not activate cortisol receptors. CONCLUSIONS This study finds sex-specific associations of C-reactive protein and cortisol levels with prenatal depression in women and with decreased development of newborn P50 inhibition. Sex-based differences in maternal response to depression with inflammation or cortisol and their developmental effects may reflect evolutionary influences to promote survival in adversity. Decreased newborn P50 inhibition is associated with later childhood behavioral problems, and decreased P50 inhibition is a pathophysiological feature of several mental illnesses.
Collapse
Affiliation(s)
- Robert Freedman
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado.
| | - Sharon K Hunter
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - Kathleen Noonan
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - Anna Wyrwa
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - Uwe Christians
- Department of Psychiatry, iC42 Clinical Research and Development, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - Amanda J Law
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado; Department of Anesthesiology, Department of Cell and Developmental Biology, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| | - M Camille Hoffman
- Institute for Children's Mental Disorders, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado; Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Anschutz Medical Center F-546, Aurora, Colorado
| |
Collapse
|
22
|
Siemiątkowska A, Kosicka K, Szpera-Goździewicz A, Krzyścin M, Bręborowicz GH, Główka FK. Cortisol metabolism in pregnancies with small for gestational age neonates. Sci Rep 2019; 9:17890. [PMID: 31784640 PMCID: PMC6884581 DOI: 10.1038/s41598-019-54362-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 11/12/2019] [Indexed: 11/27/2022] Open
Abstract
Small for gestational age (SGA) newborns are often born from hypertensive pregnancies. This study aimed to compare the systemic metabolism of cortisol (F) in pregnancies with SGA and appropriate for gestational age (AGA) infants, considering both the normotensive (NT) and hypertensive patients. We hypothesized that the disturbances in systemic metabolism of F in pre-eclampsia (PE) might be attributed not to hypertension only, but to SGA. The study included 117 pregnants in the third trimester, divided into groups: NT pregnancy and SGA neonate (SGA-NT); NT pregnancy and AGA neonate (AGA-NT; controls), and respective groups with PE: SGA-PE and AGA-PE. We assessed the glucocorticoid balance with the function of enzymes involved in systemic metabolism of F: 11β-hydroxysteroid dehydrogenase type 1 and 2 (11β-HSD1 and 11β-HSD2), 5α- and 5β-reductase. The enzymes' functions were estimated with the levels of F, cortisone (E), and their metabolites in plasma or urine, which we measured with HPLC-FLD and HPLC-MS/MS. The plasma F/E and urinary free F/E (UFF/UFE) ratios correlated significantly only in patients with the normal function of 5α- and 5β-reductase. The increased function of 11β-HSD2 was noted in all pre-eclamptic pregnancies. Increased function of 5α- and 5β-reductase was specific only for SGA-PE pregnancies, and the function of 5α-reductase was dependent on fetal sex. The SGA-NT pregnancies with male fetuses trended towards the higher function of renal 11β-HSD2 and 5β-reductase; SGA-NT pregnancies with female fetuses lacked any systemic glucocorticoid imbalance. In conclusion, systemic metabolism of F is the most intensive in pre-eclamptic pregnancies complicated by SGA with female fetuses. Our study supports the hypothesis about the different origins of PE and idiopathic intrauterine growth restriction and suggests the sex-specific mechanisms responsible for fetal growth restriction.
Collapse
Affiliation(s)
- Anna Siemiątkowska
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781, Poznań, Poland
| | - Katarzyna Kosicka
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781, Poznań, Poland.
| | - Agata Szpera-Goździewicz
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznań, Poland
| | - Mariola Krzyścin
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznań, Poland
| | - Grzegorz H Bręborowicz
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznań, Poland
| | - Franciszek K Główka
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781, Poznań, Poland
| |
Collapse
|
23
|
Wright N, Pickles A, Braithwaite EC, Sharp H, Hill J. Sex-dependent associations between maternal prenatal cortisol and child callous-unemotional traits: Findings from the Wirral Child Health and Development Study. Psychoneuroendocrinology 2019; 109:104409. [PMID: 31446327 PMCID: PMC6857434 DOI: 10.1016/j.psyneuen.2019.104409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/12/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Elevated maternal glucocorticoids during pregnancy may impact on fetal development in a sex-dependent way, leading to increased amygdala activation and increased risk for internalising disorders in females. Based on evidence implicating reduced amygdala activation in callous-unemotional (CU) traits, we predicted that elevated maternal cortisol in pregnancy would be associated with lower CU traits and elevated anxious-depressed symptoms, only in girls. METHODS Participants were 225 members of a stratified subsample within an epidemiological longitudinal cohort (WCHADS). Salivary cortisol was measured over two days at 32 weeks gestation (on waking, 30-min post-waking and during the evening) and the log of the area under the curve (LogAUC) was calculated as an index of diurnal cortisol. Mothers reported on child CU traits and anxious-depressed symptoms at 2.5, 3.5 and 5.0 years of age. RESULTS As predicted there was a sex of child by cortisol interaction (p < .001) whereby elevated maternal cortisol was associated with lower child CU traits, explaining 25% of the variance, in girls, but not in boys. This effect remained when controlling for relevant confounders and anxious-depressed symptoms. By contrast, elevated maternal cortisol did not predict higher anxious-depressed symptoms in girls. CONCLUSIONS The study adds to growing evidence for sex-dependent effects of elevated maternal cortisol during pregnancy on early child psychopathology, consistent with mediation by elevated amygdala activation. The conditions under which, in girls, this is associated with heightened responsiveness to others' distress characteristic of low CU traits, or with increased affective symptoms, require further study.
Collapse
Affiliation(s)
- Nicola Wright
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK.
| | - Andrew Pickles
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK.
| | | | - Helen Sharp
- Department of Psychological Sciences, Institute of Life and Human Sciences, Liverpool, UK.
| | - Jonathan Hill
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK.
| |
Collapse
|
24
|
Vitellius G, Delemer B, Caron P, Chabre O, Bouligand J, Pussard E, Trabado S, Lombes M. Impaired 11β-Hydroxysteroid Dehydrogenase Type 2 in Glucocorticoid-Resistant Patients. J Clin Endocrinol Metab 2019; 104:5205-5216. [PMID: 31225872 DOI: 10.1210/jc.2019-00800] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/17/2019] [Indexed: 02/13/2023]
Abstract
CONTEXT Six patients carrying heterozygous loss-of-function mutations of glucocorticoid (GC) receptor (GR) presented with hypercortisolism, associated with low kalemia, low plasma renin, and aldosterone levels, with or without hypertension, suggesting a pseudohypermineralocorticism whose mechanisms remain unclear. We hypothesize that an impaired activity of the 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2; encoded by the HSD11B2 gene), catalyzing cortisol (F) inactivation, may account for an inappropriate activation of a renal mineralocorticoid signaling pathway in these GC-resistant patients. OBJECTIVE We aim at studying the GR-mediated regulation of HSD11B2. DESIGN The HSD11B2 promoter was subcloned and luciferase reporter assays evaluated GR-dependent HSD11B2 regulation, and 11β-HSD2 expression/activity was studied in human breast cancer MCF7 cells, endogenously expressing this enzyme. RESULTS Transfection assays revealed that GR transactivated the long (2.1-kbp) HSD11B2 promoter construct, whereas a defective 501H GR mutant was unable to stimulate luciferase activity. GR-mediated transactivation of the HSD11B2 gene was inhibited by the GR antagonist RU486. A threefold increase in HSD11B2 mRNA levels was observed after dexamethasone (DXM) treatment of MCF7 cells, inhibited by RU486 or by actinomycin, supporting a GR-dependent transcription. Chromatin immunoprecipitation further demonstrated a DXM-dependent GR recruitment onto the HSD11B2 promoter. 11β-HSD2 activity, evaluated by the cortisone/F ratio, quantified by liquid chromatography/tandem mass spectrometry, was 10-fold higher in the supernatant of DXM-treated cells than controls, consistent with a GR-dependent stimulation of 11β-HSD2 catalytic activity. CONCLUSION Collectively, we demonstrate that 11β-HSD2 expression and activity are transcriptionally regulated by GR. In the context of GR haploinsufficiency, these findings provide evidence that defective GR signaling may account for apparent mineralocorticoid excess in GC-resistant patients.
Collapse
Affiliation(s)
- Géraldine Vitellius
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche_S U1185, Faculty of Medicine at Université Paris-Sud, University Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Brigitte Delemer
- Service d'Endocrinologie-Diabète-Nutrition, Hôpital Robert Debré, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Philippe Caron
- Service d'Endocrinologie, Pôle Cardio-Vasculaire et Métabolique, Centre Hospitalier Universitaire de Larrey, Toulouse, France
| | - Olivier Chabre
- Endocrinologie, Pavillon des Écrins, Centre Hospitalier Universitaire de Grenoble, La Tronche, Grenoble, France
| | - Jérôme Bouligand
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche_S U1185, Faculty of Medicine at Université Paris-Sud, University Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpitaux Universitaires Paris-Sud, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire de Bicêtre, France
| | - Eric Pussard
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche_S U1185, Faculty of Medicine at Université Paris-Sud, University Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpitaux Universitaires Paris-Sud, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire de Bicêtre, France
| | - Séverine Trabado
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche_S U1185, Faculty of Medicine at Université Paris-Sud, University Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpitaux Universitaires Paris-Sud, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire de Bicêtre, France
| | - Marc Lombes
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche_S U1185, Faculty of Medicine at Université Paris-Sud, University Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
| |
Collapse
|
25
|
Ae-Ngibise KA, Wylie BJ, Boamah-Kaali E, Jack DW, Oppong FB, Chillrud SN, Gyaase S, Kaali S, Agyei O, Kinney PL, Mujtaba M, Wright RJ, Asante KP, Lee AG. Prenatal maternal stress and birth outcomes in rural Ghana: sex-specific associations. BMC Pregnancy Childbirth 2019; 19:391. [PMID: 31664941 PMCID: PMC6819589 DOI: 10.1186/s12884-019-2535-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 09/24/2019] [Indexed: 01/08/2023] Open
Abstract
Background In developed countries, prenatal maternal stress has been associated with poor fetal growth, however this has not been evaluated in rural sub-Saharan Africa. We evaluated the effect of prenatal maternal stress on fetal growth and birth outcomes in rural Ghana. Methods Leveraging a prospective, rural Ghanaian birth cohort, we ascertained prenatal maternal negative life events, categorized scores as 0-2 (low stress; referent), 3-5 (moderate), and > 5 (high) among 353 pregnant women in the Kintampo North Municipality and Kintampo South District located within the middle belt of Ghana. We employed linear regression to determine associations between prenatal maternal stress and infant birth weight, head circumference, and length. We additionally examined associations between prenatal maternal stress and adverse birth outcome, including low birth weight, small for gestational age, or stillbirth. Effect modification by infant sex was examined. Results In all children, high prenatal maternal stress was associated with reduced birth length (β = − 0.91, p = 0.04; p-value for trend = 0.04). Among girls, moderate and high prenatal maternal stress was associated with reduced birth weight (β = − 0.16, p = 0.02; β = − 0.18, p = 0.04 respectively; p-value for trend = 0.04) and head circumference (β = − 0.66, p = 0.05; β = − 1.02, p = 0.01 respectively; p-value for trend = 0.01). In girls, high prenatal stress increased odds of any adverse birth outcome (OR 2.41, 95% CI 1.01-5.75; p for interaction = 0.04). Sex-specific analyses did not demonstrate significant effects in boys. Conclusions All infants, but especially girls, were vulnerable to effects of prenatal maternal stress on birth outcomes. Understanding risk factors for impaired fetal growth may help develop preventative public health strategies. Trial registration NCT01335490 (prospective registration). Date of Registration: April 14, 2011. Status of Registration: Completed.
Collapse
Affiliation(s)
- Kenneth Ayuurebobi Ae-Ngibise
- Ghana Health Service, Kintampo Health Research Centre, Brong Ahafo Region, Kintampo, Ghana.,School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Blair J Wylie
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ellen Boamah-Kaali
- Ghana Health Service, Kintampo Health Research Centre, Brong Ahafo Region, Kintampo, Ghana
| | - Darby W Jack
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Felix Boakye Oppong
- Ghana Health Service, Kintampo Health Research Centre, Brong Ahafo Region, Kintampo, Ghana
| | - Steven N Chillrud
- Lamont-Doherty Earth Observatory at Columbia University, Palisades, NY, USA
| | - Stephaney Gyaase
- Ghana Health Service, Kintampo Health Research Centre, Brong Ahafo Region, Kintampo, Ghana
| | - Seyram Kaali
- Ghana Health Service, Kintampo Health Research Centre, Brong Ahafo Region, Kintampo, Ghana
| | - Oscar Agyei
- Ghana Health Service, Kintampo Health Research Centre, Brong Ahafo Region, Kintampo, Ghana
| | - Patrick L Kinney
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Mohammed Mujtaba
- Ghana Health Service, Kintampo Health Research Centre, Brong Ahafo Region, Kintampo, Ghana
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Pediatrics, Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kwaku Poku Asante
- Ghana Health Service, Kintampo Health Research Centre, Brong Ahafo Region, Kintampo, Ghana
| | - Alison G Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA.
| |
Collapse
|
26
|
Petrullo L, Hinde K, Lu A. Steroid hormone concentrations in milk predict sex-specific offspring growth in a nonhuman primate. Am J Hum Biol 2019; 31:e23315. [PMID: 31468643 DOI: 10.1002/ajhb.23315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/09/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES In humans and other mammals, maternal hormones are transferred to offspring during lactation via milk and may regulate postnatal development, including the pace of early growth. Here, we used a nonhuman primate model to test the hypotheses that milk cortisol and dehydroepiandrosterone-sulfate (DHEAS) concentrations reflect maternal characteristics, and that changes in these hormones across lactation are associated with early postnatal growth rates. METHODS Demographic information, morphometrics, and milk samples were collected from rhesus macaque mothers and their infants at the California National Primate Research Center in Davis, California. Using linear models, we examined the relationship between maternal traits and milk hormone concentrations (N = 104 females) and explored the effect of milk hormones on the rate of offspring growth (N = 72 mother-infant dyads), controlling for available milk energy. RESULTS Contrary to previous studies, we found that milk cortisol concentrations were categorically higher in multiparous females than in primiparous females. However, milk DHEAS concentrations decreased with maternal parity. Neither milk cortisol nor DHEAS were related to maternal rank. Finally, changes in milk hormones predicted offspring growth in a sex-specific and temporal manner: increases in cortisol from peak to late lactation predicted faster female growth, and increases in DHEAS concentrations from early to peak and peak to late lactation predicted faster male growth. CONCLUSIONS Our findings shed light on how hormonal components of milk have sex-specific effects on offspring growth during early postnatal life with varying temporal windows of sensitivity.
Collapse
Affiliation(s)
- Lauren Petrullo
- Interdepartmental Doctoral Program in Anthropological Sciences, Stony Brook University, Stony Brook, New York
| | - Katie Hinde
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona.,School for Human Evolution and Social Change, Arizona State University, Tempe, Arizona.,Brain, Mind, and Behavior Unit, California National Primate Research Center, Davis, California
| | - Amy Lu
- Department of Anthropology, Stony Brook University, Stony Brook, New York
| |
Collapse
|
27
|
Jin S, Sun X, Liu Q, Liang H, Li C, Mao Z, Song C, Xia W, Li Y, Xu S. Steroid Hormones in Cord Blood Mediate the Association Between Maternal Prepregnancy BMI and Birth Weight. Obesity (Silver Spring) 2019; 27:1338-1346. [PMID: 31207092 DOI: 10.1002/oby.22524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/19/2019] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Maternal overweight has been associated with increasing offspring birth weight, but epidemiological data on potential biological mechanisms are limited. This study aimed to examine whether steroid hormones mediate the association between maternal prepregnancy BMI (pre-BMI) and birth weight. METHODS This study involving 2,039 participants was conducted from an ongoing cohort study in Wuhan, China. Mediation analysis was used to identify the extent to which steroid hormones mediated associations. RESULTS Each one-unit increase in pre-BMI was significantly associated with lower log2 -transformed cord blood levels of cortisol and corticosterone. Levels of cortisol and corticosterone were also negatively associated with birth weight. It was estimated that corticosterone mediated 3.48% of the association between pre-BMI and birth weight, and no significant mediation effect was observed in cortisol. After stratification by maternal gestational weight gain (GWG; within or in excess of the Institute of Medicine [IOM] guidelines), the associations of pre-BMI with cortisol and corticosterone levels were significant in the women with GWG > IOM but not in women with GWG ≤ IOM. When the mediation analysis in the women with GWG > IOM was limited, the mediation effects of cord blood cortisol and corticosterone were both significant (P < 0.05). CONCLUSIONS Cord blood cortisol and corticosterone partially mediate the association of increased maternal pre-BMI with higher birth weight.
Collapse
Affiliation(s)
- Shuna Jin
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaojie Sun
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Liu
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huifang Liang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunhui Li
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhenxing Mao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Chengwu Song
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
28
|
Huang S, Dong W, Jiao Z, Liu J, Li K, Wang H, Xu D. Prenatal Dexamethasone Exposure Induced Alterations in Neurobehavior and Hippocampal Glutamatergic System Balance in Female Rat Offspring. Toxicol Sci 2019; 171:369-384. [PMID: 31518422 DOI: 10.1093/toxsci/kfz163] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epidemiological investigations have suggested that periodic use of dexamethasone during pregnancy is a risk factor for abnormal behavior in offspring, but the potential mechanism remains unclear. In this study, we investigated the changes in the glutamatergic system and neurobehavior in female offspring with prenatal dexamethasone exposure (PDE) to explore intrauterine programing mechanisms. Compared with the control group, rat offspring with PDE exhibited spatial memory deficits and anxiety-like behavior. The expression of hippocampal glucocorticoid receptors (GR) and histone deacetylase 2 (HDAC2) increased, whereas histone H3 lysine 14 acetylation (H3K14ac) of brain-derived neurotrophic factor (BDNF) exon IV (BDNF IV) and expression of BDNF decreased. The glutamatergic system also changed. We further observed that changes in the fetal hippocampus were consistent with those in adult offspring. In vitro, the administration of 0.5 μM dexamethasone to the H19-7 fetal hippocampal neuron cells directly led to a cascade of changes in the GR/HDAC2/BDNF pathway, whereas the GR antagonist RU486 and the HDAC2 inhibitor romidepsin (Rom) reversed changes caused by dexamethasone to the H3K14ac level of BDNF IV and to the expression of BDNF. The increase in HDAC2 can be reversed by RU486, and the changes in the glutamatergic system can be partially reversed after supplementation with BDNF. It is suggested that PDE increases the expression of HDAC2 by activating GR, reducing the H3K14ac level of BDNF IV, inducing alterations in neurobehavior and hippocampal glutamatergic system balance. The findings suggest that BDNF supplementation and glutamatergic system improvement are potential therapeutic targets for the fetal origins of abnormal neurobehavior.
Collapse
Affiliation(s)
- Songqiang Huang
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Wanting Dong
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Zhexiao Jiao
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Jie Liu
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Ke Li
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, Hubei Province, China
| | - Hui Wang
- *Department of Pharmacology, School of Basic Medical Sciences
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University
| | - Dan Xu
- *Department of Pharmacology, School of Basic Medical Sciences
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University
| |
Collapse
|
29
|
Fetal growth restriction is associated with an altered cardiopulmonary and cerebral hemodynamic response to surfactant therapy in preterm lambs. Pediatr Res 2019; 86:47-54. [PMID: 30982059 DOI: 10.1038/s41390-019-0398-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/03/2019] [Accepted: 04/07/2019] [Indexed: 11/08/2022]
Abstract
BACKGROUND Efficacy of surfactant therapy in fetal growth restricted (FGR) preterm neonates is unknown. METHODS Twin-bearing ewes underwent surgery at 105 days gestation to induce FGR in one twin by single umbilical artery ligation. At 123-127 days, catheters and flow probes were implanted in pulmonary and carotid arteries to measure flow and pressure. Lambs were delivered, intubated and mechanically ventilated. At 10 min, surfactant (100 mg kg-1) was administered. Ventilation, oxygenation, and hemodynamic responses were recorded for 1 h before euthanasia at 120 min. Lung tissue and bronchoalveolar lavage fluid was collected for analysis of surfactant protein mRNA and phosphatidylcholines (PCs). RESULTS FGR preterm lambs were 26% lighter than appropriate for gestational age (AGA) lambs and had baseline differences in lung mechanics and pulmonary blood flows. Surfactant therapy reduced ventilator and oxygen requirements and improved lung mechanics in both groups, although a more rapid improvement in compliance and tidal volume was observed in AGA lambs. Surfactant administration was associated with decreased mean pulmonary and carotid blood flow in FGR but not AGA lambs. No major differences in surfactant protein mRNA or PC levels were noted. CONCLUSIONS Surfactant therapy was associated with an altered pulmonary and cerebral hemodynamic response in preterm FGR lambs.
Collapse
|
30
|
Jayasuriya NA, Hughes AE, Sovio U, Cook E, Charnock-Jones DS, Smith GCS. A Lower Maternal Cortisol-to-Cortisone Ratio Precedes Clinical Diagnosis of Preterm and Term Preeclampsia by Many Weeks. J Clin Endocrinol Metab 2019; 104:2355-2366. [PMID: 30768664 PMCID: PMC6500797 DOI: 10.1210/jc.2018-02312] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/16/2019] [Indexed: 12/26/2022]
Abstract
CONTEXT Previous studies have shown reduced placental levels of 11-β-hydroxysteroid dehydrogenase type 2 (11βHSD2) in preeclampsia (PE). However, it is unknown if the maternal cortisol-to-cortisone ratio is predictive of placental complications of pregnancy. OBJECTIVE To determine the relationship between the maternal serum cortisol-to-cortisone ratio at different stages of pregnancy and the risk of PE or fetal growth restriction (FGR). DESIGN Women from the Pregnancy Outcome Prediction Study experiencing PE (n = 194) or FGR (n = 185), plus a random sample of healthy controls (n = 279), were studied. Steroids were measured at ∼12, ∼20, ∼28, and ∼36 weeks of gestational age (wkGA). Separate analyses were performed for outcomes with term or preterm delivery. Associations were modeled using logistic regression. RESULTS At 28 wkGA, the cortisol-to-cortisone ratio was negatively associated (OR per 1 SD increase, 95% CI)] with preterm PE (OR 0.33, 95% CI 0.19 to 0.57), term PE (OR 0.61, 95% CI 0.49 to 0.76), and preterm FGR (OR 0.50, 95% CI 0.29 to 0.85). At 36 wkGA, the cortisol-to-cortisone ratio was negatively associated with term PE (OR 0.42, 95% CI 0.32 to 0.55) but not term FGR (OR 1.07, 95% CI 0.87 to 1.31). Associations were not materially affected by adjustment for maternal characteristics. CONCLUSIONS A lower maternal serum cortisol-to-cortisone ratio precedes clinical manifestation of PE and preterm FGR by many weeks, despite previous reports of reduced levels of placental 11βHSD2 in these conditions. Our observations implicate enhanced maternal 11βHSD2 activity or reduced 11βHSD type 1 activity in the pathophysiology of PE.
Collapse
Affiliation(s)
- Nimesh A Jayasuriya
- University of Glasgow School of Medicine, Glasgow, United Kingdom
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Alice E Hughes
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ulla Sovio
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Emma Cook
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - D Stephen Charnock-Jones
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Gordon C S Smith
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- Correspondence and Reprint Requests: Gordon C. S. Smith, DSc, FMedSci, Department of Obstetrics and Gynaecology, University of Cambridge, Rosie Hospital, Cambridge, CB2 0SW, United Kingdom. E-mail:
| |
Collapse
|
31
|
Portella AK, Paquet C, Bischoff AR, Molle RD, Faber A, Moore S, Arora N, Levitan R, Silveira PP, Dube L. Multi-behavioral obesogenic phenotypes among school-aged boys and girls along the birth weight continuum. PLoS One 2019; 14:e0212290. [PMID: 30789933 PMCID: PMC6383887 DOI: 10.1371/journal.pone.0212290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/30/2019] [Indexed: 11/18/2022] Open
Abstract
Evidence shows that extremes of birth weight (BW) carry a common increased risk for the development of adiposity and related cardiovascular diseases, but little is known about the role of obesogenic behaviors in this process. Moreover, no one has empirically examined whether the relationship between BW, obesogenic behaviors and BMI along the full low-to-high birthweight continuum reflects the U-shape pattern expected from common risk at both BW extremes. Our objective was to characterize physical activity, screen time, and eating behavior and their relationship to BMI as a function of BW among school-aged boys and girls. In this cross-sectional study, 460 children aged 6 to 12 years (50% boys) from Montreal, Canada provided information on sleeping time, screen time, physical activity levels, eating behavior (emotional, external and restrained eating) and anthropometrics (height, weight, BW) through parent reported questionnaires. BMI was normalized using WHO Standards (zBMI), and BW expressed as ratio using Canadian population standards (BW for gestational age and sex). Analyses were conducted using generalized linear models with linear and quadratic terms for BW, stratified by sex and adjusted for age, ethnicity and household income. In boys, physical activity and screen time showed U-shaped associations with BW, while physical activity had an inverted U-shaped in girls. Emotional and restrained eating had positive linear relations with BW in boys and girls. Sleep time and external eating were not associated with BW. A U-shaped relationship between BW and zBMI was found in boys but no association was found in girls. Only sleep (in boys and girls), and emotional eating (girls only) were related to zBMI and mediation of the BW-zBMI relationship was only supported for emotional eating. In conclusion, BW relates to obesogenic behaviors and BMI in both non-linear and linear ways, and these associations differed by sex.
Collapse
Affiliation(s)
- Andre Krumel Portella
- Desautels Faculty of Management, McGill Center for the Convergence of Health and Economics, McGill University, Montreal, QC, Canada
- PostGraduate Program in Pediatrics, Universidade Federal de Ciencias da Saude de Porto Alegre, Porto Alegre, RS, Brasil
- * E-mail:
| | - Catherine Paquet
- School of Health Sciences, Centre for Population Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Adrianne Rahde Bischoff
- Division of Neonatology, Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Roberta Dalle Molle
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Aida Faber
- Desautels Faculty of Management, McGill Center for the Convergence of Health and Economics, McGill University, Montreal, QC, Canada
| | - Spencer Moore
- Arnold School of Public Health, University of South Carolina, Columbia, SC, United States of America
| | | | - Robert Levitan
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Patricia Pelufo Silveira
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Sackler Institute for Epigenetics & Psychobiology, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Laurette Dube
- Desautels Faculty of Management, McGill Center for the Convergence of Health and Economics, McGill University, Montreal, QC, Canada
| |
Collapse
|
32
|
Iwata S, Kinoshita M, Okamura H, Tsuda K, Saikusa M, Harada E, Saitoh S, Iwata O. Intrauterine growth and the maturation process of adrenal function. PeerJ 2019; 7:e6368. [PMID: 30746307 PMCID: PMC6368969 DOI: 10.7717/peerj.6368] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/28/2018] [Indexed: 11/20/2022] Open
Abstract
BACKGROUNDS Environmental factors during early life alter the hypothalamus-pituitary-adrenal (HPA) axis regulation and increase the risk of diseases in later life. However, adrenal function at each developmental stage has not fully been investigated in relation to pathological antenatal conditions. Cortisol levels of newborns with intrauterine growth restriction (IUGR) are elevated during the neonatal period; however, when studied during early childhood, cortisol levels are reduced compared with their peers, suggesting that the HPA axis regulation might be altered from activation to suppression, the timing of which remains uncertain. AIM The aim of this study was to assess the presence of an interaction between intrauterine growth and postnatal age on cortisol levels in newborns hospitalised at a neonatal intensive care unit. METHODS We performed a secondary analysis using a dataset from saliva samples of 62 newborns collected between 30 and 40 weeks corrected age. Interactions between postnatal age and clinical variables with regard to cortisol levels were assessed. RESULTS The z-score of the birth weight and IUGR showed significant interactions with postnatal age on cortisol levels; cortisol levels were higher ≤5 days of birth and lower >14 days of birth than those in their peers without IUGR. CONCLUSION The adrenal function of newborns with IUGR might be altered from activation to suppression within the first several weeks of life. Longitudinal studies need to address when/how IUGR alters adrenal functions, and how these responses are associated with diseases during adulthood.
Collapse
Affiliation(s)
- Sachiko Iwata
- Center for Human Development and Family Science, Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Masahiro Kinoshita
- Centre for Developmental and Cognitive Neuroscience, Department of Paediatrics and Child Health, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hisayoshi Okamura
- Centre for Developmental and Cognitive Neuroscience, Department of Paediatrics and Child Health, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kennosuke Tsuda
- Center for Human Development and Family Science, Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Mamoru Saikusa
- Centre for Developmental and Cognitive Neuroscience, Department of Paediatrics and Child Health, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Eimei Harada
- Centre for Developmental and Cognitive Neuroscience, Department of Paediatrics and Child Health, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shinji Saitoh
- Center for Human Development and Family Science, Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Osuke Iwata
- Center for Human Development and Family Science, Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| |
Collapse
|
33
|
Molenaar NM, Tiemeier H, van Rossum EFC, Hillegers MHJ, Bockting CLH, Hoogendijk WJG, van den Akker EL, Lambregtse-van den Berg MP, El Marroun H. Prenatal maternal psychopathology and stress and offspring HPA axis function at 6 years. Psychoneuroendocrinology 2019; 99:120-127. [PMID: 30223193 DOI: 10.1016/j.psyneuen.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Intrauterine exposures such as maternal psychopathology and stress are known to influence the physical and mental health of the offspring. One of the proposed pathways underlying these associations is dysregulated hypothalamic-pituitary-adrenal (HPA) axis activity in the offspring. This study examined the relation of perinatal maternal symptoms of psychopathology and stress with offspring HPA axis activity at 6 years as measured by hair cortisol and cortisone concentrations. METHODS The study was part of the population-based Generation R Study, a prospective population-based cohort from fetal life onwards. 2546 children and their mothers formed the study population. Perinatal maternal psychopathology and stress were assessed by questionnaires in the second and third trimester. Principal components for both psychopathology and stress were created to reduce the number of explanatory variables. Child hair samples for cortisol and cortisone measurements were collected at the age of 6. Linear regression analysis, adjusted for covariates, was used to examine associations between maternal psychopathology and stress and child hair cortisol and cortisone levels. RESULTS The maternal psychopathology principal component was associated with higher child hair cortisone (adjusted B = 0.24, 95%CI 0.08;0.40, p-value < 0.01). Effect estimates of the individual dimensions ranged from 0.97 (95%CI 0.21;1.73, p-value = 0.01) for interpersonal sensitivity to 1.67 (95%CI 0.86;2.47, p-value < 0.01) for paranoid ideation. In addition, children exposed to intrauterine stress, as measured by the principal component, had higher hair cortisone levels (adjusted B = 0.54, 95%CI 0.21;0.88, p-value < 0.01). Exposure to maternal psychopathology and stress was not associated with offspring hair cortisol. Stratification by child sex resulted in associations between maternal symptoms of psychopathology during pregnancy and child hair cortisone levels in boys and associations between maternal symptoms of stress during pregnancy and child hair cortisone levels in girls. CONCLUSION Our results suggest that maternal psychopathology and stress during pregnancy are associated with long-term HPA axis activity of the offspring. The association of maternal psychopathology and stress during pregnancy with offspring hair cortisone levels is a novel finding. Future studies should examine whether these psychophysiological differences between exposed and non-exposed children underlie offspring morbidity associated with maternal psychopathology and stress during pregnancy.
Collapse
Affiliation(s)
- N M Molenaar
- The Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H Tiemeier
- The Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands; The Department of Social and Behavioral Sciences, Harvard TH Chan School of Public Health, Boston, USA
| | - E F C van Rossum
- The Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - M H J Hillegers
- The Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - C L H Bockting
- The Department of Psychiatry, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - W J G Hoogendijk
- The Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - E L van den Akker
- The Department of Pediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - M P Lambregtse-van den Berg
- The Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands; The Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - H El Marroun
- The Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands; The Department of Pediatrics, Erasmus Medical Center, Rotterdam, The Netherlands; The Department of Psychology, Education and Child Studies, Erasmus School of Social and Behavioural Sciences, Rotterdam, The Netherlands.
| |
Collapse
|
34
|
He Z, Zhang J, Huang H, Yuan C, Zhu C, Magdalou J, Wang H. Glucocorticoid-activation system mediated glucocorticoid-insulin-like growth factor 1 (GC-IGF1) axis programming alteration of adrenal dysfunction induced by prenatal caffeine exposure. Toxicol Lett 2018; 302:7-17. [PMID: 30528684 DOI: 10.1016/j.toxlet.2018.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 02/08/2023]
Abstract
Glucocorticoids play a major factor in fetal maturation and fate decision after birth. We have previously demonstrated that prenatal caffeine exposure (PCE) resulted in adrenal dysplasia. However, its molecular mechanism has not been clarified. In the present study, a rat model of intrauterine growth retardation (IUGR) was established by PCE, and offspring were sacrificed. Moreover, NCI-H295 A cells were used to confirm glucocorticoid-related molecular mechanism. Results showed that PCE fetal weight decreased, and the IUGR rate increased, while serum corticosterone levels increased but insulin-like growth factor 1 (IGF1) levels decreased. Fetal adrenals exhibited an activated glucocorticoid-activation system, and the downregulated expression of IGF1 signal pathway and steroidal synthetases. For adult rats, there was no significant change in the glucocorticoid-activation system in the PCE group, the IGF1 signal pathway showed increased trend, and the expression levels of adrenal steroidal synthetases were close to normal. The data in vitro showed that the cortisol of 1200 nM can inhibit the expression of adrenocortical cell steroidal synthetases and IGF1 signal pathway when compared with the control. Meanwhile, the glucocorticoid-activation system was activated while GR inhibitor mifepristone can reverse the effect of cortisol. Furthermore, cortisol can also promote GR into the nucleus after its activation. Based on these findings, we speculated that high concentrations of glucocorticoid in utero led to GR in the nucleus through its activation and then inhibited the IGF1 signaling pathway by activating the glucocorticoid-activation system, which could further downregulate steroid synthesis.
Collapse
Affiliation(s)
- Zheng He
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Jinzhi Zhang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Hegui Huang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Chao Yuan
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Chunyan Zhu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Jacques Magdalou
- UMR 7365 CNRS-Université de Lorraine, Faculté de Médecine, Vandœuvre-lès-Nancy, France
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China.
| |
Collapse
|
35
|
Soo JY, Wiese MD, Berry MJ, McMillen IC, Morrison JL. Intrauterine growth restriction may reduce hepatic drug metabolism in the early neonatal period. Pharmacol Res 2018; 134:68-78. [PMID: 29890254 DOI: 10.1016/j.phrs.2018.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/23/2018] [Accepted: 06/04/2018] [Indexed: 11/26/2022]
Abstract
The effects of intrauterine growth restriction (IUGR) extend well into postnatal life. IUGR is associated with an increased risk of adverse health outcomes, which often leads to greater medication usage. Many medications require hepatic metabolism for activation or clearance, but hepatic function may be altered in IUGR fetuses. Using a sheep model of IUGR, we determined the impact of IUGR on hepatic drug metabolism and drug transporter expression, both important mediators of fetal drug exposure, in late gestation and in neonatal life. In the late gestation fetus, IUGR decreased the gene expression of uptake drug transporter OATPC and increased P-glycoprotein protein expression in the liver, but there was no change in the activity of the drug metabolising enzymes CYP3A4 or CYP2D6. In contrast, at 3 weeks of age, CYP3A4 activity was reduced in the livers of lambs born with low birth weight (LBW), indicating that LBW results in changes to drug metabolising capacity in neonatal life. Together, these results suggest that IUGR may reduce hepatic drug metabolism in fetal and neonatal life through different mechanisms.
Collapse
Affiliation(s)
- Jia Yin Soo
- Early Origins of Adult Health Research Group, Adelaide, SA, 5001, Australia; School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia
| | - Michael D Wiese
- School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia
| | - Mary J Berry
- Centre for Translational Physiology, Wellington, New Zealand; Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | | | - Janna L Morrison
- Early Origins of Adult Health Research Group, Adelaide, SA, 5001, Australia; School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia.
| |
Collapse
|
36
|
Ryu RJ, Easterling TR, Caritis SN, Venkataramanan R, Umans JG, Ahmed MS, Clark S, Kantrowitz-Gordon I, Hays K, Bennett B, Honaker MT, Thummel KE, Shen DD, Hebert MF. Prednisone Pharmacokinetics During Pregnancy and Lactation. J Clin Pharmacol 2018; 58:1223-1232. [PMID: 29733485 DOI: 10.1002/jcph.1122] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/18/2018] [Indexed: 11/09/2022]
Abstract
To evaluate the steady-state pharmacokinetics of prednisone and its metabolite prednisolone in pregnant and lactating female subjects, 19 subjects received prednisone (4-40 mg/day orally) in early (n = 3), mid (n = 9), and late (n = 13) pregnancy as well as postpartum with (n = 2) and without (n = 5) lactation. Serial blood and urine samples were collected over 1 dosing interval. Prednisone and its metabolite, prednisolone, steady-state noncompartmental pharmacokinetic parameters were estimated. During pregnancy, prednisone apparent oral clearance increased with dose (35.1 ± 11.4 L/h with 5 mg, 52.6 ± 5.2 L/h with 10 mg, and 64.3 ± 6.9 L/h with 20 mg, P = .001). Similarly, unbound prednisone apparent oral clearance increased with dose. In addition, prednisolone renal clearance increased with dose (0.3 ± 0.3 L/h with 5 mg, 0.5 ± 0.4 L/h with 10 mg, and 1.3 ± 1.1 L/h with 20 mg, P = .002). Higher prednisone (r = 0.57, P ≤ .05) and prednisolone (r = 0.75, P ≤ .05) concentrations led to a higher percentage of unbound drug. Breast-milk/plasma area under the concentration-time curve ratios were 0.5-0.6 for prednisone and 0.02-0.03 for prednisolone. Relative infant doses were 0.35% to 0.53% and 0.09% to 0.18%, for prednisone and prednisolone, respectively. Prednisone and prednisolone exhibit dose- and concentration-dependent pharmacokinetics during pregnancy, and infant exposure to these agents via breast milk is minimal.
Collapse
Affiliation(s)
- Rachel J Ryu
- Department of Pharmacy, University of Washington, Seattle, WA, USA
| | - Thomas R Easterling
- Department of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
| | - Steve N Caritis
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Raman Venkataramanan
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jason G Umans
- MedStar Health Research Institute, and the Georgetown-Howard, Hyattsville, MD, USA.,Universities Center for Clinical and Translational Science, Washington, DC, USA
| | - Mahmoud S Ahmed
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Shannon Clark
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Karen Hays
- Department of Pharmacy, University of Washington, Seattle, WA, USA
| | - Brooke Bennett
- Department of Pharmacy, University of Washington, Seattle, WA, USA
| | - Matthew T Honaker
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Kenneth E Thummel
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Danny D Shen
- Department of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Mary F Hebert
- Department of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
| |
Collapse
|
37
|
Gifford RM, Reynolds RM. Sex differences in early-life programming of the hypothalamic-pituitary-adrenal axis in humans. Early Hum Dev 2017; 114:7-10. [PMID: 28927573 DOI: 10.1016/j.earlhumdev.2017.09.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increasing evidence supports fetal glucocorticoid exposure with associated altered offspring hypothalamic-pituitary-adrenal (HPA) axis activity as a key mechanism linking early life events with later life disease. Alterations in HPA axis activity are linked to a range of cardiometabolic and psychiatric diseases. As many of these diseases manifest sex differences in presentation we review the evidence for programmed sex-differences in the HPA axis. Available literature suggests vulnerability of the female HPA axis to prenatal stressors with female offspring demonstrating increased HPA axis reactivity. This may be due to changes in placental glucocorticoid metabolism leading to increased fetal glucocorticoid exposure. We discuss the potential consequences of increased vulnerability of the female HPA axis for later life health and consider the underlying mechanisms. Further studies are needed to determine whether sex-differences in early-life programming of the HPA axis represent a pathway underpinning the sex-differences in common cardiometabolic and psychiatric diseases.
Collapse
Affiliation(s)
- Robert M Gifford
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom.
| | - Rebecca M Reynolds
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom.
| |
Collapse
|
38
|
Stress and the HPA Axis: Balancing Homeostasis and Fertility. Int J Mol Sci 2017; 18:ijms18102224. [PMID: 29064426 PMCID: PMC5666903 DOI: 10.3390/ijms18102224] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/17/2017] [Accepted: 10/21/2017] [Indexed: 12/25/2022] Open
Abstract
An organism’s reproductive fitness is sensitive to the environment, integrating cues of resource availability, ecological factors, and hazards within its habitat. Events that challenge the environment of an organism activate the central stress response system, which is primarily mediated by the hypothalamic–pituitary–adrenal (HPA) axis. The regulatory functions of the HPA axis govern the cardiovascular and metabolic system, immune functions, behavior, and reproduction. Activation of the HPA axis by various stressors primarily inhibits reproductive function and is able to alter fetal development, imparting a biological record of stress experienced in utero. Clinical studies and experimental data indicate that stress signaling can mediate these effects through direct actions in the brain, gonads, and embryonic tissues. This review focuses on the mechanisms by which stress activation of the HPA axis impacts fertility and fetal development.
Collapse
|
39
|
Lazo-de-la-Vega-Monroy ML, Solís-Martínez MO, Romero-Gutiérrez G, Aguirre-Arzola VE, Wrobel K, Wrobel K, Zaina S, Barbosa-Sabanero G. 11 beta-hydroxysteroid dehydrogenase 2 promoter methylation is associated with placental protein expression in small for gestational age newborns. Steroids 2017; 124:60-66. [PMID: 28502862 DOI: 10.1016/j.steroids.2017.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 11/15/2022]
Abstract
Small for gestational age infants have greater risk of developing metabolic diseases in adult life. It has been suggested that low birth weight may result from glucocorticoid excess in utero, a key mechanism in fetal programming. The placental enzyme 11-beta hydroxysteroid dehydrogenase type 2 (11β-HSD2, HSD11B2 gene) acts as a barrier protecting the fetus from maternal corticosteroid deleterious effects. Low placental 11β-HSD2 transcription and activity have been associated with low birth weight, yet the mechanism regulating its protein expression is not fully understood. In the present study we aimed to analyze 11β-HSD2 protein expression in placentas of adequate and small for gestational age (AGA and SGA, respectively) newborns from healthy mothers, and to explore whether 11β-HSD2 protein expression could be modulated by DNA methylation. 11β-HSD2 protein levels were measured by western blot in placental biopsies from term AGA and SGA infants (n=10 per group). DNA methylation was profiled both globally and in the HSD11B2 promoter by liquid chromatography with UV detection and methylation-specific melting curve analysis, respectively. We found lower placental 11β-HSD2 protein expression and higher HSD11B2 promoter methylation in SGA compared to AGA. Promoter methylation was inversely correlated with both protein expression and, importantly, birth weight. No changes in global placental methylation were found. In conclusion, lower 11β-HSD2 protein expression is associated with higher HSD11B2 promoter methylation, correlating with birth weight in healthy pregnancy. Our data support the role of 11β-HSD2 in determining birth weight, providing evidence of its regulation by epigenetic mechanisms, which may affect postnatal metabolic disease risk.
Collapse
Affiliation(s)
| | | | | | | | - Katarzyna Wrobel
- Department of Chemistry, University of Guanajuato, Guanajuato, Mexico.
| | - Kazimierz Wrobel
- Department of Chemistry, University of Guanajuato, Guanajuato, Mexico.
| | - Silvio Zaina
- Medical Sciences Department, Health Sciences Division, University of Guanajuato, Leon Campus, Mexico.
| | - Gloria Barbosa-Sabanero
- Medical Sciences Department, Health Sciences Division, University of Guanajuato, Leon Campus, Mexico.
| |
Collapse
|
40
|
Iughetti L, Lucaccioni L, Ferrari F. Challenges in the development and growth of small for gestational age newborns. Expert Rev Endocrinol Metab 2017; 12:253-260. [PMID: 30058882 DOI: 10.1080/17446651.2017.1338137] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Being born with low birth weight (less than 2.500 gr) is considered a public health problem, with an overall world rate of about 14%. Low birth weight may result from a premature birth (< 37 weeks of gestation), from several causes of intra-uterine growth restriction or from a combination of both. Areas covered: We described how, if weight and/or length at birth are not adequate for gestational age, the newborn is considered 'small for gestational age' and may present several growth, hormonal and developmental peculiarities, possibly due to the growth restriction developed during pregnancy. Expert commentary: We provide a description of the possible mid-term consequences of being born small for gestational age and how to follow and provide care for these babies from a multidisciplinary point of view.
Collapse
Affiliation(s)
- Lorenzo Iughetti
- a Post-graduate School of Paediatrics, Department of Medical and Surgical Sciences of the Mother, Children and Adults , University of Modena and Reggio Emilia , Modena , Italy
| | - Laura Lucaccioni
- b Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences of the Mother, Children and Adults , University of Modena and Reggio Emilia , Modena , Italy
| | - Fabrizio Ferrari
- b Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences of the Mother, Children and Adults , University of Modena and Reggio Emilia , Modena , Italy
| |
Collapse
|
41
|
Class-Specific Histone Deacetylase Inhibitors Promote 11-Beta Hydroxysteroid Dehydrogenase Type 2 Expression in JEG-3 Cells. Int J Cell Biol 2017; 2017:6169310. [PMID: 28321257 PMCID: PMC5339487 DOI: 10.1155/2017/6169310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/17/2017] [Accepted: 01/24/2017] [Indexed: 12/13/2022] Open
Abstract
Exposure to maternal cortisol plays a crucial role in fetal organogenesis. However, fetal overexposure to cortisol has been linked to a range of short- and long-term adverse outcomes. Normally, this is prevented by the expression of an enzyme in the placenta called 11-beta hydroxysteroid dehydrogenase type 2 (11β-HSD2) which converts active cortisol to its inactive metabolite cortisone. Placental 11β-HSD2 is known to be reduced in a number of adverse pregnancy complications, possibly through an epigenetic mechanism. As a result, a number of pan-HDAC inhibitors have been examined for their ability to promote 11β-HSD2 expression. However, it is not known if the effects of pan-HDAC inhibition are a general phenomenon or if the effects are dependent upon a specific class of HDACs. Here, we examined the ability of pan- and class-specific HDAC inhibitors to regulate 11β-HSD2 expression in JEG3 cells. We find that pan-, class I, or class IIa HDAC inhibition promoted 11β-HSD2 expression and prevented cortisol or interleukin-1β-induced decrease in its expression. These results demonstrate that targeting a specific class of HDACs can promote 11β-HSD2 expression in JEG3 cells. This adds to the growing body of evidence suggesting that HDACs may be crucial in maintaining normal fetal development.
Collapse
|
42
|
Sex differences in early-life programming of the hypothalamic-pituitary-adrenal axis in humans suggest increased vulnerability in females: a systematic review. J Dev Orig Health Dis 2017; 8:244-255. [PMID: 28103963 DOI: 10.1017/s204017441600074x] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fetal glucocorticoid overexposure is a key mechanism linking early development with later-life disease. In humans, low birth weight associates with increased fasting cortisol, hypothalamic-pituitary-adrenal (HPA) axis reactivity, and with cardiovascular risk and cognitive decline. As there are sex differences in these adult diseases, we hypothesized that there may be sex differences in programming of the HPA axis in response to prenatal stressors. We conducted a systematic review following Meta-Analysis of Observational Studies in Epidemiology and Preferred Reporting Items for Systematic Reviews and Meta-Analysis. We searched Embase, MEDLINE and Web of Science from inception to 31 October 2016. We included studies related to sex differences, prenatal exposures and HPA axis. We excluded studies investigating specific disease states. The 23 included studies investigated the consequences of low birth weight, preterm birth and maternal stressors of asthma, psychosocial stress and glucocorticoid medications on HPA axis outcomes of placental glucocorticoid biology and offspring HPA axis function in early life and later life. Female offspring exposed to stressors had increased HPA axis reactivity compared with males. Furthermore, the female placenta increased its permeability to maternal glucocorticoids following maternal stress with changes in the expression of 11β-hydroxysteroid dehydrogenase enzymes in response to maternal glucocorticoid exposure or asthma. Among males there was some evidence of altered diurnal cortisol secretion. We conclude that although there is some evidence of male vulnerability leading to altered diurnal cortisol secretion, the female HPA axis is more vulnerable to programming, particularly in terms of its reactivity; this suggests a mechanism underlying sex differences in later-life diseases.
Collapse
|
43
|
Giesbrecht GF, Rash JA, Edwards HE, Wynne-Edwards KE. Full-term deliveries without antecedent labor reveal sex differences in umbilical cord glucocorticoid concentrations. Psychoneuroendocrinology 2016; 74:121-125. [PMID: 27608361 DOI: 10.1016/j.psyneuen.2016.08.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 08/10/2016] [Accepted: 08/31/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Previous studies have shown that pregnant women have higher salivary cortisol levels when the fetus is female. These findings suggest a basis for the sex differences observed in many offspring outcomes after exposure to in utero stress, but it is not known if fetal adrenal glucocorticoid synthesis differs by sex. METHODS Arterial and venous umbilical cord blood samples were collected immediately after scheduled cesarean delivery (n=52, 25 female). Cortisol and corticosterone concentrations were quantified by liquid chromatography coupled to tandem mass spectrometry. RESULTS Sex differences were observed for fetal arterial and venous cortisol and venous corticosterone, with higher levels present when the fetus was female. However, sex differences were not observed for fetal synthesis of cortisol, suggesting that the fetus does not control the differences observed in cord blood glucocorticoids. CONCLUSIONS The presence of sex differences in umbilical cord glucocorticoid concentrations in the absence of sex differences in glucocorticoid synthesis by the fetal adrenal gland suggests that these differences have a maternal or placental origin. Thus, the in utero glucocorticoids in circulation are sex-specific and may have developmental importance for sex differences in psychiatric and neurodevelopment disorders that display sex biases.
Collapse
Affiliation(s)
- Gerald F Giesbrecht
- Department of Pediatrics, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada; Department of Community Health Sciences, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada; Department of Psychology, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada.
| | - Joshua A Rash
- Department of Psychology, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada
| | - Heather E Edwards
- Allan Centre for Women & Department of Obstetrics and Gynecology, University of Calgary, Calgary, Alberta, Canada
| | - Katherine E Wynne-Edwards
- Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, AB T2N 4N1, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, AB T2N 4N1, Canada
| |
Collapse
|
44
|
Sexually dimorphic and interactive effects of prenatal maternal cortisol and psychological distress on infant cortisol reactivity. Dev Psychopathol 2016; 29:805-818. [PMID: 27426858 DOI: 10.1017/s0954579416000493] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In utero exposure to maternal psychological distress is a risk factor for developmental psychopathology, and these effects are believed to partially occur via dysregulation of the maternal and fetal hypothalamus-adrenal-pituitary axes. Nevertheless, only a few human studies have directly assessed the effects of prenatal cortisol exposure on infant cortisol reactivity, and none have investigated sex differences or potential interactions between prenatal cortisol and psychological distress. Here we report on a prospective longitudinal investigation (N = 236) of in utero exposure to maternal cortisol and distress in a relatively high socioeconomic status and low-risk population to determine whether these exposures interact in their effects on infant (M age = 3.0 months, range = 2.3-5.0 months, 51.9% male) cortisol reactivity and whether there are sex differences in these effects. Results revealed both sexually dimorphic and interactive effects of prenatal cortisol and distress, even after controlling for postnatal distress. In general, blunted reactivity in females was associated with exposure to high maternal distress and flattened patterns of diurnal maternal cortisol, whereas blunted reactivity in males was associated with exposure to steeper morning increases and daytime decreases in maternal cortisol. The findings suggest that sex differences in the effects of prenatal cortisol and distress on infant cortisol reactivity are a plausible mechanism by which maternal experiences during pregnancy contribute to sex differences in the development of psychopathology.
Collapse
|
45
|
Konstantakou P, Mastorakos G, Vrachnis N, Tomlinson JW, Valsamakis G. Dysregulation of 11beta-hydroxysteroid dehydrogenases: implications during pregnancy and beyond. J Matern Fetal Neonatal Med 2016; 30:284-293. [PMID: 27018008 DOI: 10.3109/14767058.2016.1171308] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glucococorticoids play a critical role in the developmental programing and fetal growth. Key molecules mediating and regulating tissue-specific glucocorticoid actions are 11beta-hydroxysteroid dehydrogenase (11beta-HSD) type 1 and 2 isozymes, both of which are expressed in the placenta and the fetal membranes. 11beta-HSD1 is implicated in the pathogenesis of metabolic syndrome and its dysregulation has been observed in pregnancy-related complications (pre-eclampsia, intrauterine growth restriction). Interestingly, preliminary clinical data have associated certain 11beta-HSD1 gene polymorphisms with hypertensive disorders in pregnancy, suggesting, if confirmed by further targeted studies, it's potential as a putative prognostic marker. Animal studies and observations in humans have confirmed that 11beta-HSD2 insufficiency is related with pregnancy adversity (pre-eclampsia, intrauterine growth restriction, preterm birth). Importantly, down-regulation or deficiency of placental 11beta-HSD2 is associated with significant restriction in fetal growth and low-birth weight, and unfavorable cardio-metabolic profile in adulthood. The potential association of 11beta-HSD1 tissue-specific dysregulation with gestational diabetes, as well as the plausible utility of 11beta-HSD2, as a biomarker of pregnancy adversity and later life morbidity, are emerging areas of intense scientific interest and future investigation.
Collapse
Affiliation(s)
- P Konstantakou
- a Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieio Hospital , Athens , Greece
| | - G Mastorakos
- a Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieio Hospital , Athens , Greece
| | - N Vrachnis
- b Department of Obstetrics and Gynecology , Aretaieio Hospital , Athens , Greece
| | - J W Tomlinson
- c Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital , Headington , UK
| | - G Valsamakis
- a Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieio Hospital , Athens , Greece
| |
Collapse
|
46
|
Kosicka K, Siemiątkowska A, Główka FK. 11β-Hydroxysteroid Dehydrogenase 2 in Preeclampsia. Int J Endocrinol 2016; 2016:5279462. [PMID: 27200090 PMCID: PMC4856917 DOI: 10.1155/2016/5279462] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/05/2016] [Indexed: 11/17/2022] Open
Abstract
Preeclampsia is a serious medical problem affecting the mother and her child and influences their health not only during the pregnancy, but also many years after. Although preeclampsia is a subject of many research projects, the etiology of the condition remains unclear. One of the hypotheses related to the etiology of preeclampsia is the deficiency in placental 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2), the enzyme which in normal pregnancy protects the fetus from the excess of maternal cortisol. The reduced activity of the enzyme was observed in placentas from pregnancies complicated with preeclampsia. That suggests the overexposure of the developing child to maternal cortisol, which in high levels exerts proapoptotic effects and reduces fetal growth. The fetal growth restriction due to the diminished placental 11β-HSD2 function may be supported by the fact that preeclampsia is often accompanied with fetal hypotrophy. The causes of the reduced function of 11β-HSD2 in placental tissue are still discussed. This paper summarizes the phenomena that may affect the activity of the enzyme at various steps on the way from the gene to the protein.
Collapse
Affiliation(s)
- Katarzyna Kosicka
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781 Poznań, Poland
- *Katarzyna Kosicka:
| | - Anna Siemiątkowska
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781 Poznań, Poland
| | - Franciszek K. Główka
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781 Poznań, Poland
| |
Collapse
|
47
|
St-Pierre J, Laurent L, King S, Vaillancourt C. Effects of prenatal maternal stress on serotonin and fetal development. Placenta 2015; 48 Suppl 1:S66-S71. [PMID: 26691753 DOI: 10.1016/j.placenta.2015.11.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 12/19/2022]
Abstract
Fetuses are exposed to many environmental perturbations that can influence their development. These factors can be easily identifiable such as drugs, chronic diseases or prenatal maternal stress. Recently, it has been demonstrated that the serotonin synthetized by the placenta was crucial for fetal brain development. Moreover, many studies show the involvement of serotonin system alteration in psychiatric disease during childhood and adulthood. This review summarizes existing studies showing that prenatal maternal stress, which induces alteration of serotonin systems (placenta and fetal brain) during a critical window of early development, could lead to alteration of fetal development and increase risks of psychiatric diseases later in life. This phenomenon, termed fetal programming, could be moderated by the sex of the fetus. This review highlights the need to better understand the modification of the maternal, placental and fetal serotonin systems induced by prenatal maternal stress in order to find early biomarkers of psychiatric disorders.
Collapse
Affiliation(s)
- Joey St-Pierre
- INRS-Institut Armand-Frappier and BioMed Research Center, Laval, QC, Canada
| | - Laetitia Laurent
- INRS-Institut Armand-Frappier and BioMed Research Center, Laval, QC, Canada
| | - Suzanne King
- Douglas Mental Health University Institute and McGill University, Montreal, QC, Canada
| | - Cathy Vaillancourt
- INRS-Institut Armand-Frappier and BioMed Research Center, Laval, QC, Canada.
| |
Collapse
|
48
|
Placental 11β-Hydroxysteroid dehydrogenase type 2 expression: Correlations with birth weight and placental metal concentrations. Placenta 2015; 36:1212-7. [DOI: 10.1016/j.placenta.2015.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/07/2015] [Accepted: 09/24/2015] [Indexed: 01/22/2023]
|
49
|
Penailillo R, Guajardo A, Llanos M, Hirsch S, Ronco AM. Folic acid supplementation during pregnancy induces sex-specific changes in methylation and expression of placental 11β-hydroxysteroid dehydrogenase 2 in rats. PLoS One 2015; 10:e0121098. [PMID: 25793274 PMCID: PMC4368185 DOI: 10.1371/journal.pone.0121098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/09/2015] [Indexed: 02/03/2023] Open
Abstract
In the placenta, 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) limits fetal glucocorticoid exposure and its inhibition has been associated to low birth weight. Its expression, encoded by the HSD11B2 gene is regulated by DNA methylation. We hypothesized that maternal diets supplemented with folic acid (FA) during pregnancy modify the expression of placental HSD11B2 through gene methylation. Wistar rats were fed with high (8 mg/kg) or normal low (1mg/kg, control) levels of FA during pregnancy. Concentrations of mRNA and protein in placentas were determined by qRT-PCR and Western blot respectively. Methylation in five CpG sites of the placental HSD11B2 promoter (-378 to -275) was analyzed by bacterial cloning and subsequent sequencing. In the FA-supplemented group, mRNA and protein levels of 11β-HSD2 decreased by 58% and increased by 89%, respectively, only in placentas attached to males. In controls, most CpG sites were not methylated except for the CpG2 site which was 80% methylated. CpG2 methylation level increased under the FA treatment; however, only in placentas attached to females was this increase significant (113%). This change was not related to HSD11B2 expression. Fetal weight of females from FA- supplemented mothers was 6% higher than females from control mothers. In conclusion, this is the first study reporting that FA over supplementation during pregnancy modifies the placental HSD11B2 gene expression and methylation in a sex-dependent manner, suggesting that maternal diets with high content of FA can induce early sex-specific responses, which may lead to long-term consequences for the offspring.
Collapse
Affiliation(s)
- Reyna Penailillo
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Angelica Guajardo
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Miguel Llanos
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Sandra Hirsch
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Ana Maria Ronco
- Laboratory of Nutrition and Metabolic Regulation, Human Nutrition Unit, Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| |
Collapse
|
50
|
Green BB, Armstrong DA, Lesseur C, Paquette AG, Guerin DJ, Kwan LE, Marsit CJ. The Role of Placental 11-Beta Hydroxysteroid Dehydrogenase Type 1 and Type 2 Methylation on Gene Expression and Infant Birth Weight. Biol Reprod 2015; 92:149. [PMID: 25788665 DOI: 10.1095/biolreprod.115.128066] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 03/17/2015] [Indexed: 12/16/2022] Open
Abstract
Maternal stress has been linked to infant birth weight outcomes, which itself may be associated with health later in life. The placenta acts as a master regulator for the fetal environment, mediating intrauterine exposures to stress through the activity of genes regulating glucocorticoids, including the 11beta-hydroxysteroid dehydrogenase (HSD11B) type 1 and 2 genes, and so we hypothesized that variation in these genes will be associated with infant birth weight. We investigated DNA methylation levels at six sites across the two genes, as well as mRNA expression for each, and the relationship to infant birth weight. Logistic regressions correcting for potential confounding factors revealed a significant association between methylation at a single CpG site within HSD11B1 and being born large for gestational age. In addition, our analysis identified correlations between methylation and gene expression, including sex-specific transcriptional regulation of HSD11B2. Our work is one of the first comprehensive views of DNA methylation and expression in the placenta for both HSD11B types 1 and 2, linking epigenetic alterations with the regulation of fetal stress and birth weight outcomes.
Collapse
Affiliation(s)
- Benjamin B Green
- Departments of Pharmacology and Toxicology and of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| | - David A Armstrong
- Departments of Pharmacology and Toxicology and of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| | - Corina Lesseur
- Departments of Pharmacology and Toxicology and of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| | - Alison G Paquette
- Departments of Pharmacology and Toxicology and of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| | - Dylan J Guerin
- Departments of Pharmacology and Toxicology and of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| | - Lauren E Kwan
- Departments of Pharmacology and Toxicology and of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| | - Carmen J Marsit
- Departments of Pharmacology and Toxicology and of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire
| |
Collapse
|