1
|
Zhao Y, Zhou C, Zhang S, Mishra JS, Li H, Lei W, Wang K, Kumar S, Zheng J. An endogenous aryl hydrocarbon receptor ligand induces preeclampsia-like phenotypes in rats. J Physiol 2025; 603:579-594. [PMID: 39652430 PMCID: PMC11737537 DOI: 10.1113/jp287503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Preeclampsia (PE) is a hypertensive disorder during human pregnancy. Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor. Exogenous and endogenous AhR ligands can induce hypertension in male rats and mice. Herein, using rats as a model, we tested the hypothesis that over-regulation of endogenous AhR ligands during pregnancy impairs vascular functions by disrupting the transcriptome in the placenta, contributing to the development of PE. Pregnant rats were injected daily with an endogenous AhR ligand, 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), from gestational day (GD) 10 to 19. Maternal mean blood pressure was measured on GD16-20. Proteinuria and uteroplacental blood flow were monitored on GD20. Placentas collected on GD20 were used to determine changes in vascular density and transcriptome. Compared with the vehicle control, ITE elevated maternal mean blood pressure by 22% and 16% on GD16 and 17, respectively. ITE increased proteinuria by 50% and decreased uteroplacental blood flow by 26%. ITE reduced the placental vascular density by 18%. RNA sequencing analysis revealed that ITE induced 1316 and 2020 differentially expressed genes (DEGs) in female and male placentas, respectively. These DEGs were enriched in pathways relevant to heart diseases, vascular functions and inflammation. Bioinformatics analysis also predicted that ITE altered immune cell infiltration in placentas depending on fetal sex. These data suggest that over-regulation of endogenous AhR ligands may lead to PE with impaired vascular functions and disrupted fetal sex-specific transcriptomes and immune cell infiltration in placentas. These AhR ligand-induced DEGs and pathways may represent promising therapeutic targets for PE-induced cardiovascular dysfunctions. KEY POINTS: An endogenous AhR ligand (ITE) elevated maternal blood pressure and proteinuria in pregnant rats, and decreased uteroplacental blood flow and fetal and placental growth, all of which are hallmarks of preeclampsia. ITE reduced vascular density and altered immune cell distribution in rat placentas. ITE dysregulated transcriptomes in rat placentas in a fetal sex-specific manner. These ITE-dysregulated genes and pathways are highly relevant to diseases of heart, vascular functions and inflammatory responses.
Collapse
Affiliation(s)
- Ying‐Jie Zhao
- Department of Obstetrics and GynecologyUniversity of Wisconsin‐MadisonMadisonWIUSA
- Department of Rheumatology, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Chi Zhou
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonAZUSA
| | - Si‐Yan Zhang
- Department of Obstetrics and GynecologyUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Jay S. Mishra
- Department of Comparative BiosciencesUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Hui‐Hui Li
- Department of Obstetrics and GynecologyUniversity of Wisconsin‐MadisonMadisonWIUSA
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Wei Lei
- Cardiovascular Medicine CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdongChina
| | - Kai Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Sathish Kumar
- Department of Obstetrics and GynecologyUniversity of Wisconsin‐MadisonMadisonWIUSA
- Department of Comparative BiosciencesUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Jing Zheng
- Department of Obstetrics and GynecologyUniversity of Wisconsin‐MadisonMadisonWIUSA
| |
Collapse
|
2
|
Huo Y, Wang W, Zhang J, Xu D, Bai F, Gui Y. Maternal androgen excess inhibits fetal cardiomyocytes proliferation through RB-mediated cell cycle arrest and induces cardiac hypertrophy in adulthood. J Endocrinol Invest 2024; 47:603-617. [PMID: 37642904 PMCID: PMC10904501 DOI: 10.1007/s40618-023-02178-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE Maternal hyperandrogenism during pregnancy is associated with adverse gestational outcomes and chronic non-communicable diseases in offspring. However, few studies are reported to demonstrate the association between maternal androgen excess and cardiac health in offspring. This study aimed to explore the relation between androgen exposure in utero and cardiac health of offspring in fetal and adult period. Its underlying mechanism is also illustrated in this research. METHODS Pregnant mice were injected with dihydrotestosterone (DHT) from gestational day (GD) 16.5 to GD18.5. On GD18.5, fetal heart tissue was collected for metabolite and morphological analysis. The hearts from adult offspring were also collected for morphological and qPCR analysis. H9c2 cells were treated with 75 μM androsterone. Immunofluorescence, flow cytometry, qPCR, and western blot were performed to observe cell proliferation and explore the underlying mechanism. RESULTS Intrauterine exposure to excessive androgen led to thinner ventricular wall, decreased number of cardiomyocytes in fetal offspring and caused cardiac hypertrophy, compromised cardiac function in adult offspring. The analysis of steroid hormone metabolites in fetal heart tissue by ultra performance liquid chromatography and tandem mass spectrometry showed that the content of androgen metabolite androsterone was significantly increased. Mechanistically, H9c2 cells treated with androsterone led to a significant decrease in phosphorylated retinoblastoma protein (pRB) and cell cycle-related protein including cyclin-dependent kinase 2 (CDK2), cyclin-dependent kinase 4 (CDK4), and cyclin D1 (CCND1) in cardiomyocytes. This resulted in cell cycle arrest at G1-S phase, which in turn inhibited cardiomyocyte proliferation. CONCLUSION Taken together, our results indicate that in utero exposure to DHT, its metabolite androsterone could directly decrease cardiomyocytes proliferation through cell cycle arrest, which has a life-long-lasting effect on cardiac health. Our study highlights the importance of monitoring sex hormones in women during pregnancy and the follow-up of cardiac function in offspring with high risk of intrauterine androgen exposure.
Collapse
Affiliation(s)
- Y Huo
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China
| | - W Wang
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510080, China
| | - J Zhang
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - D Xu
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China
| | - F Bai
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China
| | - Y Gui
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China.
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China.
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
3
|
Zhao YJ, Zhou C, Wei YY, Zhang SY, Mishra JS, Li HH, Lei W, Wang K, Kumar S, Zheng J. An Endogenous Aryl Hydrocarbon Receptor Ligand Induces Preeclampsia-like Phenotypes: Transcriptome, Phosphoproteome, and Cell Functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572271. [PMID: 38187714 PMCID: PMC10769228 DOI: 10.1101/2023.12.20.572271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Background Preeclampsia (PE) is one hypertensive disorder and a leading cause of maternal and fetal mortality and morbidity during human pregnancy. Aryl hydrocarbon receptor (AhR) is a transcription factor, which regulates vascular functions. Exogenous and endogenous AhR ligands can induce hypertension in animals. However, if dysregulation of endogenous AhR ligands contributes to the pathophysiology of PE remains elusive. Methods We measured AhR activities in human maternal and umbilical vein sera. We also applied physiological, cellular, and molecular approaches to dissect the role of endogenous AhR ligands in vascular functions during pregnancy using pregnant rats and primary human umbilical vein endothelial cells (HUVECs) as models. Results PE elevated AhR activities in human umbilical vein sera. Exposure of pregnant rats to an endogenous AhR ligand, 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) increased blood pressure and proteinuria, while decreased uteroplacental blood flow and reduced fetal and placental weights, all of which are hallmarks of PE. ITE dampened vascular growth and fetal sex-specifically altered immune cell infiltration in rat placentas. ITE also decreased cell proliferation and cell monolayer integrity in HUVECs in vitro . RNA sequencing analysis revealed that ITE dysregulated transcriptome in rat placentas and HUVECs in a fetal sex-specific manner. Bottom-up phosphoproteomics showed that ITE disrupted phosphoproteome in HUVECs. These ITE-dysregulated genes and phosphoproteins were enriched in biological functions and pathways which are highly relevant to diseases of heart, liver, and kidney, vascular functions, inflammation responses, cell death, and kinase inhibition. Conclusions Dysregulation of endogenous AhR ligands during pregnancy may lead to the development of PE with underlying impaired vascular functions, fetal sex-specific immune cell infiltration and transcriptome, and phosphoproteome. Thus, this study has provided a novel mechanism for the development of PE and potentially other forms of hypertensive pregnancies. These AhR ligand-activated genes and phosphoproteins might represent promising therapeutic and fetal sex-specific targets for PE-impaired vascular functions.
Collapse
|
4
|
Guo F, Mao S, Long Y, Zhou B, Gao L, Huang H. The Influences of Perinatal Androgenic Exposure on Cardiovascular and Metabolic Disease of Offspring of PCOS. Reprod Sci 2023; 30:3179-3189. [PMID: 37380913 DOI: 10.1007/s43032-023-01286-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
Hyperandrogenism is an endocrine disorder affecting a large population of reproductive-aged women, thus proportionally high number of fetuses are subjected to prenatal androgenic exposure (PNA). The short-term stimulations at critical ontogenetic stages can wield lasting influences on the health. The most commonly diagnosed conditions in reproductive age women is polycystic ovary syndrome (PCOS). PNA may affect the growth and development of many systems in the whole body and disrupts the normal metabolic trajectory in the offspring of PCOS, contributing to the prevalence of cardiovascular and metabolic diseases (CVMD), including myocardial hypertrophy, hypertension, hyperinsulinemia, insulin resistance, hyperglycemia, obesity, and dyslipidemia, which are the leading causes of hospitalizations in young PCOS offspring. In this review, we focus on the effects of prenatal androgenic exposure on the cardiovascular and metabolic diseases in offspring, discuss the possible pathogenesis respectively, and summarize potential management strategies to improve metabolic health of PCOS offspring. It is expected that the incidence of CVMD and the medical burden will be reduced in the future.
Collapse
Affiliation(s)
- Fei Guo
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Suqing Mao
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Yuhang Long
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Bokang Zhou
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ling Gao
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Hefeng Huang
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| |
Collapse
|
5
|
Ramamoorthi Elangovan V, Saadat N, Ghnenis A, Padmanabhan V, Vyas AK. Developmental programming: adverse sexually dimorphic transcriptional programming of gestational testosterone excess in cardiac left ventricle of fetal sheep. Sci Rep 2023; 13:2682. [PMID: 36792653 PMCID: PMC9932081 DOI: 10.1038/s41598-023-29212-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Adverse in-utero insults during fetal life alters offspring's developmental trajectory, including that of the cardiovascular system. Gestational hyperandrogenism is once such adverse in-utero insult. Gestational testosterone (T)-treatment, an environment of gestational hyperandrogenism, manifests as hypertension and pathological left ventricular (LV) remodeling in adult ovine offspring. Furthermore, sexual dimorphism is noted in cardiomyocyte number and morphology in fetal life and at birth. This study investigated transcriptional changes and potential biomarkers of prenatal T excess-induced adverse cardiac programming. Genome-wide coding and non-coding (nc) RNA expression were compared between prenatal T-treated (T propionate 100 mg intramuscular twice weekly from days 30 to 90 of gestation; Term: 147 days) and control ovine LV at day 90 fetus in both sexes. Prenatal T induced differential expression of mRNAs in the LV of female (2 down, 5 up) and male (3 down, 1 up) (FDR < 0.05, absolute log2 fold change > 0.5); pathways analysis demonstrated 205 pathways unique to the female, 382 unique to the male and 23 common pathways. In the male, analysis of ncRNA showed differential regulation of 15 lncRNAs (14 down, 1 up) and 27 snoRNAs (26 down and 1 up). These findings suggest sexual dimorphic modulation of cardiac coding and ncRNA with gestational T excess.
Collapse
Affiliation(s)
| | - Nadia Saadat
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Arpita K Vyas
- College of Medicine, California Northstate University, Elk Grove, CA, USA.
- Department of Pediatrics, Division of Pediatric Endocrinology, School of Medicine, Washington University, St Louis, MO, USA.
| |
Collapse
|
6
|
Feng Y, Lian X, Guo K, Zhang G, Huang X. A comprehensive analysis of metabolomics and transcriptomics to reveal major metabolic pathways and potential biomarkers of human preeclampsia placenta. Front Genet 2022; 13:1010657. [PMID: 36263435 PMCID: PMC9574103 DOI: 10.3389/fgene.2022.1010657] [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: 08/03/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The etiology of preeclampsia (PE) remains unclear. With the utilization of metabolomics, dysregulated production of several metabolic components in human plasma, such as lipids, amino acids, androgens and estrogens, was found to be important in the pathogenesis of PE. Transcriptomics adds more in-depth information, and the integration of transcriptomics and metabolomics may yield further insight into PE pathogenesis than either one alone.Objectives: We investigated the placental metabolomics and transcriptomics of PE patients to identify affected metabolic pathways and potential biological targets for exploring the disease pathogenesis.Methods: Integrated transcriptomics and metabolomics were used to analyze five paired human placentas from patients with severe PE and normal pregnancies. This was followed by further validation of our findings in a publicly available dataset of 173 PE vs. 157 control placentas. In addition, weighted gene coexpression network construction was performed to assess the correlation between genetic alterations and diseases.Results: We identified 66 and 41 differentially altered metabolites in negative and positive ion modes, respectively, in the PE group compared to the control group, and found 2,560 differentially expressed genes. Several pathways were aberrantly altered in the PE placenta at both the metabolic and transcriptional levels, including steroid hormone biosynthesis, the cAMP signaling pathway, neuroactive ligand–receptor interactions, taste transduction and prion diseases. Additionally, we found 11 differential metabolites and 11 differentially expressed genes involved in the steroid hormone biosynthesis pathway, indicating impaired metabolism of steroid hormones in the PE placenta. Furthermore, we found that CYP11A1, HSD3B2, and HSD17B6 are highly correlated with diseases.Conclusion: Our findings provide a profile of the dysregulated steroid hormone biosynthesis in PE placenta, we observed a dysregulated cortisol-to-cortisone ratio, testosterone accumulation, decreased testosterone downstream metabolites, impaired production of estrone and estriol, and aberrant hydroxylation and methylation of estradiol. Disorders of placental steroid hormone metabolism might be a consequence or a compensatory change in pathological placentation in PE, which underscores the need to investigate the physiology of steroid hormone metabolites in the etiology of PE.
Collapse
Affiliation(s)
- Yan Feng
- Fetal Care Center, Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xinlei Lian
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, China
| | - Kaimin Guo
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Guanglan Zhang
- Fetal Care Center, Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xuan Huang
- Fetal Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xuan Huang,
| |
Collapse
|
7
|
Ghnenis A, Padmanabhan V, Vyas A. Sexual dimorphism in testosterone programming of cardiomyocyte development in sheep. Am J Physiol Heart Circ Physiol 2022; 322:H607-H621. [PMID: 35119334 PMCID: PMC8957338 DOI: 10.1152/ajpheart.00691.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 11/22/2022]
Abstract
Perturbed in utero hormone milieu leads to intrauterine growth retardation (IUGR), a known risk factor for left ventricular (LV) dysfunction later in life. Gestational testosterone (T) excess predisposes offspring to IUGR and leads to LV myocardial disarray and hypertension in adult females. However, the early impact of T excess on LV programming and if it is female specific is unknown. LV tissues were obtained at day 90 gestation from days 30-90 T-treated or control fetuses (n = 6/group/sex) and morphometric and molecular analyses were conducted. Gestational T treatment increased cardiomyocyte number only in female fetuses. T excess upregulated receptor expression of insulin and insulin-like growth factor. Furthermore, in a sex-specific manner, T increased expression of phosphatidylinositol 3-kinase (PI3K) while downregulating phosphorylated mammalian target of rapamycin (pmTOR)-to-mTOR ratio suggestive of compensatory response. T excess 1) upregulated atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), markers of stress and cardiac hypertrophy and 2) upregulated estrogen receptors1 (ESR1) and 2 (ESR2), but not in androgen receptor (AR). Thus, gestational T excess upregulated markers of cardiac stress and hypertrophy in both sexes while inducing cardiomyocyte hyperplasia only in females, likely mediated via insulin and estrogenic programming.NEW & NOTEWORTHY The present study demonstrates sex-specific effects of gestational T excess between days 30 and 90 of gestation on the cardiac phenotype. Furthermore, the sex-specific programming is likely secondary to perturbation in both estrogen and insulin signaling pathways collectively. These findings are supportive of the role of androgen excess to serve as early biomarkers of CVD and could be critical in identifying therapeutic targets for LV hypertrophy and predict long-term CVD.
Collapse
Affiliation(s)
- Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | | | - Arpita Vyas
- College of Human Medicine, California Northstate University, Elk Grove, California
| |
Collapse
|
8
|
Gopalakrishnan K, Mishra JS, Ross JR, Abbott DH, Kumar S. Hyperandrogenism diminishes maternal-fetal fatty acid transport by increasing FABP 4-mediated placental lipid accumulation. Biol Reprod 2022; 107:514-528. [PMID: 35357467 DOI: 10.1093/biolre/ioac059] [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: 01/18/2022] [Revised: 02/22/2022] [Accepted: 03/17/2022] [Indexed: 11/12/2022] Open
Abstract
Long-chain polyunsaturated fatty acids (LCPUFAs) are critical for fetal brain development. Infants born to preeclamptic mothers or those born growth restricted due to placental insufficiency have reduced LCPUFA, and are at higher risk for developing neurodevelopmental disorders. Since plasma levels of testosterone (T) and fatty acid-binding protein 4 (FABP4) are elevated in preeclampsia, we hypothesized that elevated T induces the expression of FABP4 in the placenta leading to compromised transplacental transport of LCPUFAs. Increased maternal T in pregnant rats significantly decreased n-3 and n-6 LCPUFA levels in maternal and fetal circulation, but increased their placental accumulation. Dietary LCPUFAs supplementation in T dams increased LCPUFA levels in the maternal circulation and further augmented placental storage, while failing to increase fetal levels. The placenta in T dams exhibited increased FABP4 mRNA and protein levels. In vitro, T dose-dependently upregulated FABP4 transcription in trophoblasts. T stimulated androgen receptor (AR) recruitment to the androgen response element and trans-activated FABP4 promoter activity, both of which were abolished by AR antagonist. T in pregnant rats and cultured trophoblasts significantly reduced transplacental transport of C14-docosahexaenoic acid (DHA) and increased C14-DHA accumulation in the placenta. Importantly, FABP4-overexpression by itself in pregnant rats and trophoblasts increased transplacental transport of C14-DHA with no significant placental accumulation. T exposure, in contrast, inhibited this FABP4-mediated effect by promoting C14-DHA placental accumulation. In summary, our studies show that maternal hyperandrogenism increases placental FABP4 expression via transcriptional upregulation and preferentially routes LCPUFAs toward cellular storage in the placenta leading to offspring lipid deficiency.
Collapse
Affiliation(s)
- Kathirvel Gopalakrishnan
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Jay S Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Jordan R Ross
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - David H Abbott
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA.,Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA.,Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA.,Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA.,Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, WI 53715, USA
| |
Collapse
|
9
|
Neven ACH, Mousa A, Boyle JA, Teede HJ. Endocrine and metabolic interactions in healthy pregnancies and hyperinsulinemic pregnancies affected by polycystic ovary syndrome, diabetes and obesity. Front Endocrinol (Lausanne) 2022; 13:993619. [PMID: 36733795 PMCID: PMC9886898 DOI: 10.3389/fendo.2022.993619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/06/2022] [Indexed: 01/18/2023] Open
Abstract
During pregnancy, the fetoplacental unit is key in the pronounced physiological endocrine changes which support pregnancy, fetal development and survival, birth and lactation. In healthy women, pregnancy is characterized by changes in insulin sensitivity and increased maternal androgen levels. These are accompanied by a suite of mechanisms that support fetal growth, maintain glucose homeostasis and protect both mother and fetus from adverse effects of pregnancy induced insulin and androgen excess. In pregnancies affected by endocrine, metabolic disorders such as polycystic ovary syndrome (PCOS), diabetes and obesity, there is an imbalance of beneficial and adverse impacts of pregnancy induced endocrine changes. These inter-related conditions are characterized by an interplay of hyperinsulinemia and hyperandrogenism which influence fetoplacental function and are associated with adverse pregnancy outcomes including hypertensive disorders of pregnancy, macrosomia, preterm delivery and caesarean section. However, the exact underlying mechanisms and relationships of the endocrine and metabolic milieu in these disorders and the impact they have on the prenatal endocrine environment and developing fetus remain poorly understood. Here we aim to review the complex endocrine and metabolic interactions in healthy women during normal pregnancies and those in pregnancies complicated by hyperinsulinemic disorders (PCOS, diabetes and obesity). We also explore the relationships between these endocrine and metabolic differences and the fetoplacental unit, pregnancy outcomes and the developing fetus.
Collapse
Affiliation(s)
- Adriana C. H. Neven
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia
| | - Jacqueline A. Boyle
- Monash Department of Obstetrics and Gynaecology, Monash Health, Clayton, VIC, Australia
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- *Correspondence: Jacqueline A. Boyle,
| | - Helena J. Teede
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia
| |
Collapse
|
10
|
Placental Development and Pregnancy-Associated Diseases. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
11
|
Meakin AS, Cuffe JSM, Darby JRT, Morrison JL, Clifton VL. Let's Talk about Placental Sex, Baby: Understanding Mechanisms That Drive Female- and Male-Specific Fetal Growth and Developmental Outcomes. Int J Mol Sci 2021; 22:6386. [PMID: 34203717 PMCID: PMC8232290 DOI: 10.3390/ijms22126386] [Citation(s) in RCA: 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
|
12
|
Zhang FF, Zhang Q, Wang YL, Wang FF, Hardiman PJ, Qu F. Intergenerational Influences between Maternal Polycystic Ovary Syndrome and Offspring: An Updated Overview. J Pediatr 2021; 232:272-281. [PMID: 33482217 DOI: 10.1016/j.jpeds.2021.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Fang-Fang Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan-Lin Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fang-Fang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
13
|
Le-Ha C, Beilin LJ, Burrows S, Keelan JA, Hickey M, Mori TA. Prenatal Testosterone Associates With Blood Pressure in Young Adults: A Prospective Cohort Study. Hypertension 2021; 77:1756-1764. [PMID: 33775121 DOI: 10.1161/hypertensionaha.120.16256] [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] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Chi Le-Ha
- From the Medical School, Royal Perth Hospital Campus (C.L-H, L.J.B., S.B., T.A.M.), the University of Western Australia, Perth
| | - Lawrence J Beilin
- From the Medical School, Royal Perth Hospital Campus (C.L-H, L.J.B., S.B., T.A.M.), the University of Western Australia, Perth
| | - Sally Burrows
- From the Medical School, Royal Perth Hospital Campus (C.L-H, L.J.B., S.B., T.A.M.), the University of Western Australia, Perth
| | - Jeffrey A Keelan
- the School of Biomedical Sciences (J.A.K.), the University of Western Australia, Perth
| | - Martha Hickey
- the Department of Obstetrics and Gynaecology and Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia (M.H.)
| | - Trevor A Mori
- From the Medical School, Royal Perth Hospital Campus (C.L-H, L.J.B., S.B., T.A.M.), the University of Western Australia, Perth
| |
Collapse
|
14
|
Jósvai A, Török M, Mátrai M, Hetthéssy J, Monori-Kiss A, Makk J, Székács B, Nádasy GL, Várbíró S. Effects of Testosterone Deficiency and Angiotensin II-Induced Hypertension on the Biomechanics of Intramural Coronary Arteries. J Sex Med 2020; 17:2322-2330. [PMID: 33067160 DOI: 10.1016/j.jsxm.2020.09.003] [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: 06/01/2020] [Revised: 08/14/2020] [Accepted: 09/12/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Andropause and hypertension also increase the risk of coronary artery damage. AIM To investigate the effect of testosterone deficiency and hypertension on intramural coronary vessels. METHODS 4 groups of 8-week-old Sprague-Dawley rats were studied: control male (Co, n=10), orchidectomized male (OCT, n=13), angiotensin (AII) hypertensive male (AII, n=10), and AII hypertensive and OCT (AII + OCT, n=8). Surgical orchidectomy was performed, and an osmotic minipump was inserted for chronic angiotensin II infusion (100 ng/min/kg). After 4 weeks, spontaneous tone and biomechanical properties of the intramural coronary resistance artery were investigated in vitro, by pressure microarteriography. OUTCOMES Morphology and biomechanics of the intramural coronaries were evaluated: the outer diameter, wall thickness-to-lumen diameter ratio, and tangential wall stress in the contracted and relaxed states. RESULTS The outer diameter was reduced in OCT and AII + OCT groups (on 50 mmHg 315 ± 20 Co; 237 ± 21 OCT; 291 ± 16 AII, and 166 ± 12 μm AII + OCT). The increased wall thickness-to-lumen diameter ratio resulted in lower tangential wall stress in AII + OCT rats (on 50 mmHg 19 ± 2 Co; 24 ± OCT; 26 ± 5 AII, and 9 ± 1 kPa AII + OCT). Spontaneous tone was increased in the hypertensive rats (AII and AII + OCT groups) (on 50 mmHg 7.7 ± 1.8 Co; 6.1 ± 1.4 OCT; 14.5 ± 3.0 AII, and 17.4 ± 4.1 % AII + OCT). CLINICAL IMPLICATIONS Andropause alone can be considered as a cardiovascular risk factor that will further exacerbate vascular damage in hypertension. STRENGTHS & LIMITATIONS A limitation of our study is that it was performed on relatively young rats, and the conclusions might not apply to coronary remodelling in older animals with slower adaptation processes. CONCLUSIONS Testosterone deficiency and hypertension damage the mechanical adaptation of the vessel wall additively: double noxa caused inward eutrophic remodeling and increased tone. Jósvai A, Török M, Mátrai M, et al. Effects of Testosterone Deficiency and Angiotensin II-Induced Hypertension on the Biomechanics of Intramural Coronary Arteries. J Sex Med 2020;17:2322-2330.
Collapse
Affiliation(s)
- Attila Jósvai
- Department of Neurosurgery, Hungarian Defence Forces Medical Centre, Budapest, Hungary.
| | - Marianna Török
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Máté Mátrai
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Judit Hetthéssy
- Department of Orthopedics, Semmelweis University, Budapest, Hungary
| | - Anna Monori-Kiss
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Jennifer Makk
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Béla Székács
- 2nd Department Internal Medicine, Department Section of Geriatrics, Szt Imre Teaching Hospital, Budapest, Hungary
| | - György L Nádasy
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Szabolcs Várbíró
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
15
|
Li S, Liu Q, Wu D, He T, Yuan J, Qiu H, Tickner J, Zheng SG, Li X, Xu J, Rong L. PKC-δ deficiency in B cells displays osteopenia accompanied with upregulation of RANKL expression and osteoclast-osteoblast uncoupling. Cell Death Dis 2020; 11:762. [PMID: 32938907 PMCID: PMC7494897 DOI: 10.1038/s41419-020-02947-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/27/2022]
Abstract
PKC-δ is an important molecule for B-cell proliferation and tolerance. B cells have long been recognized to play a part in osteoimmunology and pathological bone loss. However, the role of B cells with PKC-δ deficiency in bone homeostasis and the underlying mechanisms are unknown. We generated mice with PKC-δ deletion selectively in B cells by crossing PKC-δ-loxP mice with CD19-Cre mice. We studied their bone phenotype using micro-CT and histology. Next, immune organs were obtained and analyzed. Western blotting was used to determine the RANKL/OPG ratio in vitro in B-cell cultures, ELISA assay and immunohistochemistry were used to analyze in vivo RANKL/OPG balance in serum and bone sections respectively. Finally, we utilized osteoclastogenesis to study osteoclast function via hydroxyapatite resorption assay, and isolated primary calvaria osteoblasts to investigate osteoblast proliferation and differentiation. We also investigated osteoclast and osteoblast biology in co-culture with B-cell supernatants. We found that mice with PKC-δ deficiency in B cells displayed an osteopenia phenotype in the trabecular and cortical compartment of long bones. In addition, PKC-δ deletion resulted in changes of trabecular bone structure in association with activation of osteoclast bone resorption and decrease in osteoblast parameters. As expected, inactivation of PKC-δ in B cells resulted in changes in spleen B-cell number, function, and distribution. Consistently, the RANKL/OPG ratio was elevated remarkably in B-cell culture, in the serum and in bone specimens after loss of PKC-δ in B cells. Finally, in vitro analysis revealed that PKC-δ ablation suppressed osteoclast differentiation and function but co-culture with B-cell supernatant reversed the suppression effect, as well as impaired osteoblast proliferation and function, indicative of osteoclast–osteoblast uncoupling. In conclusion, PKC-δ plays an important role in the interplay between B cells in the immune system and bone cells in the pathogenesis of bone lytic diseases.
Collapse
Affiliation(s)
- Shangfu Li
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China.
| | - Qiuli Liu
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, China
| | - Depeng Wu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| | - Tianwei He
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| | - Jinbo Yuan
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Heng Qiu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jennifer Tickner
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Song Guo Zheng
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xiaojuan Li
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Southern Medical University, Guangzhou Guangdong, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China.
| |
Collapse
|
16
|
Li S, He T, Wu D, Zhang L, Chen R, Liu B, Yuan J, Tickner J, Qin A, Xu J, Rong L. Conditional Knockout of PKC-δ in Osteoclasts Favors Bone Mass Accrual in Males Due to Decreased Osteoclast Function. Front Cell Dev Biol 2020; 8:450. [PMID: 32582715 PMCID: PMC7295979 DOI: 10.3389/fcell.2020.00450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
Protein kinase C delta (PKC-δ) functions as an important regulator in bone metabolism. However, the precise involvement of PKC-δ in the regulation of osteoclasts remains elusive. We generated an osteoclast specific PKC-δ knockout mouse strain to investigate the function of PKC-δ in osteoclast biology. Bone phenotype was investigated using microcomputed tomography. Osteoclast and osteoblast parameters were assessed using bone histomorphometry, and analysis of osteoclast formation and function with osteoclastogensis and hydroxyapatite resorption assays. The molecular mechanisms by which PKC-δ regulated osteoclast function were dissected by Western Blotting, TUNEL assay, transfection and transcriptome sequencing. We found that ablation of PKC-δ in osteoclasts resulted in an increase in trabecular and cortical bone volume in male mice, however, the bone mass phenotype was not observed in female mice. This was accompanied by decreased osteoclast number and surface, and Cathepsin-K protein levels in vivo, as well as decreased osteoclast formation and resorption in vitro in a male-specific manner. PKC-δ regulated androgen receptor transcription by binding to its promoter, moreover, PKC-δ conditional knockout did not increase osteoclast apoptosis but increased MAPK signaling and enhanced androgen receptor transcription and expression, finally leding to significant alterations in gene expression and signaling changes related to extracellular matrix proteins specifically in male mice. In conclusion, PKC-δ plays an important role in osteoclast formation and function in a male-specific manner. Our work reveals a previously unknown target for treatment of gender-related bone diseases.
Collapse
Affiliation(s)
- Shangfu Li
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| | - Tianwei He
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| | - Depeng Wu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| | - Ruiqiang Chen
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| | - Jinbo Yuan
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jennifer Tickner
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, China
| |
Collapse
|
17
|
Zhao C, Zhou Y, Shen X, Gong M, Lu Y, Fang C, Chen J, Ju R. Circular RNA expression profiling in the fetal side of placenta from maternal polycystic ovary syndrome and circ_0023942 inhibits the proliferation of human ovarian granulosa cell. Arch Gynecol Obstet 2020; 301:963-971. [PMID: 32193602 DOI: 10.1007/s00404-020-05495-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 03/05/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Circular RNAs (circRNAs) are widely expressed noncoding RNAs which play important roles in various processes. The present study aimed to explore the effect of maternal PCOS on the expression of circRNAs in fetus and assessed the potential role of circRNA in human ovarian granulosa cell proliferation. METHODS Total RNA was extracted from the fetal side of placental tissues from maternal PCOS (n = 3) and healthy puerpera (n = 3) for circRNA microarray. Real-time reverse transcriptase quantitative PCR (RT-qPCR) was used to validate the microarray data in fetal side of placental tissues from puerpera with (n = 18) and without (n = 30) PCOS. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were applied to predict the functions and pathways of circ_0023942 host genes. The circRNA-miRNA-mRNA network was constructed through bioinformatics prediction. Circ_0023942 overexpression vector was transiently transfected into human ovarian granulosa cell lines KGN and COV434. Cell proliferation was detected by cell counting kit-8. The protein expression level was determined by western blot. RESULTS Compared with healthy puerpera, 14 circRNAs were significantly upregulated and 101 circRNAs were significantly downregulated in the fetal side of placenta from maternal PCOS according to the microarray data. Six differentially expressed circRNAs were selected for validation by RT-qPCR, and the expression patterns of circ_0023942, circ_0002151, circ_0001274, and circ_0008514 were consistent with the microarray data. Circ_0023942 was chosen for further investigation. GO and KEGG analysis predicted that circ_0023942 participated in the regulation of developmental process and the MAPK signaling pathway. Seven miRNAs were predicted to be the targets of circ_0023942. Overexpression of circ_0023942 inhibited human ovarian granulosa cell proliferation and suppressed the expression of CDK-4. CONCLUSION Maternal PCOS impairs circ_0023942 expression in fetus. Overexpression of circ_0023942 inhibits human ovarian granulosa cell proliferation possibly via regulating CDK-4.
Collapse
Affiliation(s)
- Chengcheng Zhao
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Yu Zhou
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China.,Department of Gynecology and Obstetrics, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Xia Shen
- Department of Gynecology and Obstetrics, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Min Gong
- Department of Gynecology and Obstetrics, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Yingfei Lu
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Chao Fang
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China.,Department of Gynecology and Obstetrics, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Rong Ju
- Department of Gynecology and Obstetrics, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China.
| |
Collapse
|
18
|
Review: Understanding the role of androgens and placental AR variants: Insight into steroid-dependent fetal-placental growth and development. Placenta 2019; 84:63-68. [DOI: 10.1016/j.placenta.2019.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/30/2022]
|
19
|
Wainstock T, Yoles I, Sergienko R, Walfisch A. The association between anogenital distance, reproductive and general health in adult females- a prospective cohort of 17 years. Reprod Toxicol 2019; 90:77-81. [PMID: 31421229 DOI: 10.1016/j.reprotox.2019.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/16/2019] [Accepted: 08/12/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Studies have suggested an association between prenatal hormonal environment and anogenital distance (AGD). We aimed to study the association between AGD and health characteristics in adult women. METHODS In this prospective cohort AGD was measured in 300 parturients during the years 2000-2001. In December 2017, study participants` hospital records were abstracted and medical diagnoses grouped into major categories, including gynecological, cardiovascular, and other. Associations between AGD and the morbidities categories were evaluated using multivariable survival models, which adjusted for confounding variables. RESULTS Participants were 44.3 ± 5.4 years old with mean AGD 40.3 mm (±10.7 mm). Women with below vs. above mean AGD were more likely to develop gynecological morbidities (39.9% vs. 27.1%, adjusted Hazard ratio 1.82; 95%CI 1.08-3.06). CONCLUSIONS Women with short AGD are at increased risk for gynecological morbidities. AGD should be studied further, and may possibly be used for screening of women at risk for these conditions.
Collapse
Affiliation(s)
- Tamar Wainstock
- Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Israel Yoles
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Clalit Health Services, Central District, Israel
| | - Ruslan Sergienko
- Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Asnat Walfisch
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
20
|
Prenatal exposure to testosterone induces cardiac hypertrophy in adult female rats through enhanced Pkcδ expression in cardiac myocytes. J Mol Cell Cardiol 2019; 128:1-10. [PMID: 30641088 DOI: 10.1016/j.yjmcc.2019.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 10/28/2018] [Accepted: 01/10/2019] [Indexed: 11/23/2022]
Abstract
High circulating androgen in women with polycystic ovary syndrome (PCOS) may increase the risk of cardiovascular disease in offspring. The aim of the present study is to investigate whether maternal androgen excess in the rat PCOS model would lead to cardiac hypertrophy in offspring. Maternal testosterone propionate (maternal-TP)-treated adult female offspring displayed cardiac hypertrophy associated with local high cardiac dihydrotestosterone (DHT). The molecular markers of cardiac hypertrophy along with androgen receptor (AR) and PKCδ, were increased in the Maternal-TP group. Treatment of primary neonatal rat ventricular cardiomyocytes (NRCMs) and H9c2 cells with DHT significantly increased cell size and upregulated PKCδ expression, which could be attenuated by AR antagonist. Treatment with phorbol 12-myristate 13-acetate (PMA), a PKC activator, significantly increased cell size and upregulated myh7 level. Rottlerin, that may inhibit PKCδ, significantly reduced the hypertrophic effect of DHT and PMA on NRCMs and H9c2 cells. Chromatin immunoprecipitation revealed that AR could bind to Pkcδ promoter. Our results indicate that prenatal exposure to testosterone may induce cardiac hypertrophy in adult female rats through enhanced Pkcδ expression in cardiac myocytes.
Collapse
|
21
|
Mishra JS, More AS, Gopalakrishnan K, Kumar S. Testosterone plays a permissive role in angiotensin II-induced hypertension and cardiac hypertrophy in male rats. Biol Reprod 2019; 100:139-148. [PMID: 30102356 PMCID: PMC6335213 DOI: 10.1093/biolre/ioy179] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 12/21/2022] Open
Abstract
Sex hormones contribute to sex differences in blood pressure. Inappropriate activation of the renin-angiotensin system is involved in vascular dysfunction and hypertension. This study evaluated the role of androgens (testosterone) in angiotensin II (Ang II)-induced increase in blood pressure, vascular reactivity, and cardiac hypertrophy. Eight-week-old male Wistar rats underwent sham operation, castration, or castration with testosterone replacement. After 12 weeks of chronic changes in androgen status, Ang II (120 ng/kg per minute) or saline was infused for 28 days via subcutaneous miniosmotic pump, and changes in blood pressure was measured. Vascular reactivity and Ang II receptor levels were examined in mesenteric arteries. Heart weight, cardiac ANP mRNA levels, and fibrosis were also assessed. Ang II infusion increased arterial pressure in intact males. The Ang II-induced increase in hypertensive response was prevented in castrated males. Testosterone replacement in castrated males restored Ang II-induced hypertensive responses. Castration reduced vascular AT1R/AT2R ratio, an effect that was reversed by testosterone replacement. Ang II-induced hypertension was associated with increased contractile response of mesenteric arteries to Ang II and phenylephrine in intact and testosterone-replaced castrated males; these increases were prevented in castrated males. Ang II infusion induced increased left ventricle-to-body weight ratio and ANP mRNA expression, indicators of left ventricular hypertrophy, and fibrosis in intact and testosterone-replaced castrated males, and castration prevented the increase in these parameters caused by Ang II. This study demonstrates that testosterone plays a permissive role in development and maintenance of Ang II-induced vascular dysfunction, hypertension, and cardiac hypertrophy.
Collapse
Affiliation(s)
- Jay S Mishra
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amar S More
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Sathish Kumar
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
22
|
Tamadon A, Hu W, Cui P, Ma T, Tong X, Zhang F, Li X, Shao LR, Feng Y. How to choose the suitable animal model of polycystic ovary syndrome? TRADITIONAL MEDICINE AND MODERN MEDICINE 2018. [DOI: 10.1142/s2575900018300047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a gynecological metabolic and endocrine disorder with uncertain etiology. To understand the etiology of PCOS or the evaluation of various therapeutic agents, different animal models have been introduced. Considering this fact that is difficult to develop an animal model that mimics all aspects of this syndrome, but, similarity of biological, anatomical, and/or biochemical features of animal model to the human PCOS phenotypes can increase its application. This review paper evaluates the recently researched animal models and introduced the best models for different research purposes in PCOS studies. During January 2013 to January 2017, 162 studies were identified which applied various kinds of animal models of PCOS including rodent, primate, ruminant and fish. Between these models, prenatal and pre-pubertal androgen rat models and then prenatal androgen mouse model have been studied in detail than others. The comparison of main features of these models with women PCOS demonstrates higher similarity of these three models to human conditions. Thereafter, letrozole models can be recommended for the investigation of various aspects of PCOS. Interestingly, similarity of PCOS features of post-pubertal insulin and human chorionic gonadotropin rat models with women PCOS were considerable which can make it as a good choice for future investigations.
Collapse
Affiliation(s)
- Amin Tamadon
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Peng Cui
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Tong Ma
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Feifei Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P. R. China
| | - Xin Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P. R. China
| | - Linus R. Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
23
|
Sherman SB, Sarsour N, Salehi M, Schroering A, Mell B, Joe B, Hill JW. Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes 2018; 9:400-421. [PMID: 29469650 PMCID: PMC6219642 DOI: 10.1080/19490976.2018.1441664] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/24/2017] [Accepted: 02/08/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Conditions of excess androgen in women, such as polycystic ovary syndrome (PCOS), often exhibit intergenerational transmission. One way in which the risk for PCOS may be increased in daughters of affected women is through exposure to elevated androgens in utero. Hyperandrogenemic conditions have serious health consequences, including increased risk for hypertension and cardiovascular disease. Recently, gut dysbiosis has been found to induce hypertension in rats, such that blood pressure can be normalized through fecal microbial transplant. Therefore, we hypothesized that the hypertension seen in PCOS has early origins in gut dysbiosis caused by in utero exposure to excess androgen. We investigated this hypothesis with a model of prenatal androgen (PNA) exposure and maternal hyperandrogenemia by single-injection of testosterone cypionate or sesame oil vehicle (VEH) to pregnant dams in late gestation. We then completed a gut microbiota and cardiometabolic profile of the adult female offspring. RESULTS The metabolic assessment revealed that adult PNA rats had increased body weight and increased mRNA expression of adipokines: adipocyte binding protein 2, adiponectin, and leptin in inguinal white adipose tissue. Radiotelemetry analysis revealed hypertension with decreased heart rate in PNA animals. The fecal microbiota profile of PNA animals contained higher relative abundance of bacteria associated with steroid hormone synthesis, Nocardiaceae and Clostridiaceae, and lower abundance of Akkermansia, Bacteroides, Lactobacillus, Clostridium. The PNA animals also had an increased relative abundance of bacteria associated with biosynthesis and elongation of unsaturated short chain fatty acids (SCFAs). CONCLUSIONS We found that prenatal exposure to excess androgen negatively impacted cardiovascular function by increasing systolic and diastolic blood pressure and decreasing heart rate. Prenatal androgen was also associated with gut microbial dysbiosis and altered abundance of bacteria involved in metabolite production of short chain fatty acids. These results suggest that early-life exposure to hyperandrogenemia in daughters of women with PCOS may lead to long-term alterations in gut microbiota and cardiometabolic function.
Collapse
Affiliation(s)
- Shermel B. Sherman
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Nadeen Sarsour
- Department of Biological Sciences, University of Toledo, Toledo, OH
| | - Marziyeh Salehi
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Allen Schroering
- Department of Neurosciences and Neurological Disorders, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Blair Mell
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Hypertension and Personalized Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Bina Joe
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Hypertension and Personalized Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Diabetes and Endocrine Research, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| |
Collapse
|
24
|
Mishra JS, More AS, Hankins GDV, Kumar S. Hyperandrogenemia reduces endothelium-derived hyperpolarizing factor-mediated relaxation in mesenteric artery of female rats. Biol Reprod 2018; 96:1221-1230. [PMID: 28486649 DOI: 10.1093/biolre/iox043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/08/2017] [Indexed: 01/05/2023] Open
Abstract
Women with polycystic ovary syndrome (PCOS) are often presented with hyperandrogenemia along with vascular dysfunction and elevated blood pressure. In animal models of PCOS, anti-androgen treatment decreased blood pressure, indicating a key role for androgens in the development of hypertension. However, the underlying androgen-mediated mechanism that contributes to increased blood pressure is not known. This study determined whether elevated androgens affect endothelium-derived hyperpolarizing factor (EDHF)-mediated vascular relaxation responses through alteration in function of gap junctional proteins. Female rats were implanted with placebo or dihydrotestosterone (DHT) pellets (7.5 mg, 90-day release). After 12 weeks of DHT exposure, blood pressure was assessed through carotid arterial catheter and endothelium-dependent mesenteric arterial EDHF relaxation using wire myograph. Connexin expression in mesenteric arteries was also examined. Elevated DHT significantly increased mean arterial pressure and decreased endothelium-dependent EDHF-mediated acetylcholine relaxation. Inhibition of Cx40 did not have any effect, while inhibition of Cx37 decreased EDHF relaxation to a similar magnitude in both controls and DHT females. On the other hand, inhibition of Cx43 significantly attenuated EDHF relaxation in mesenteric arteries of controls but not DHT females. Elevated DHT did not alter Cx37 or Cx40, but decreased Cx43 mRNA and protein levels in mesenteric arteries. In vitro exposure of DHT to cultured mesenteric artery smooth muscle cells dose-dependently downregulated Cx43 expression. In conclusion, increased blood pressure in hyperandrogenic females is due, at least in part, to decreased EDHF-mediated vascular relaxation responses. Decreased Cx43 expression and activity may play a role in contributing to androgen-induced decrease in EDHF function.
Collapse
Affiliation(s)
- Jay S Mishra
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Amar S More
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Gary D V Hankins
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Sathish Kumar
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Texas, USA
| |
Collapse
|
25
|
Xie W, Ren M, Li L, Zhu Y, Chu Z, Zhu Z, Ruan Q, Lou W, Zhang H, Han Z, Huang X, Xiang W, Wang T, Yao P. Perinatal testosterone exposure potentiates vascular dysfunction by ERβ suppression in endothelial progenitor cells. PLoS One 2017; 12:e0182945. [PMID: 28809938 PMCID: PMC5557363 DOI: 10.1371/journal.pone.0182945] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/27/2017] [Indexed: 12/04/2022] Open
Abstract
Recent clinical cohort study shows that testosterone therapy increases cardiovascular diseases in men with low testosterone levels, excessive circulating androgen levels may play a detrimental role in the vascular system, while the potential mechanism and effect of testosterone exposure on the vascular function in offspring is still unknown. Our preliminary results showed that perinatal testosterone exposure in mice induces estrogen receptor β (ERβ) suppression in endothelial progenitor cells (EPCs) in offspring but not mothers, while estradiol (E2) had no effect. Further investigation showed that ERβ suppression is due to perinatal testosterone exposure-induced epigenetic changes with altered DNA methylation on the ERβ promoter. During aging, EPCs with ERβ suppression mobilize to the vascular wall, differentiate into ERβ-suppressed mouse endothelial cells (MECs) with downregulated expression of SOD2 (mitochondrial superoxide dismutase) and ERRα (estrogen-related receptor α). This results in reactive oxygen species (ROS) generation and DNA damage, and the dysfunction of mitochondria and fatty acid metabolism, subsequently potentiating vascular dysfunction. Bone marrow transplantation of EPCs that overexpressed with either ERβ or a SIRT1 single mutant SIRT1-C152(D) that could modulate SIRT1 phosphorylation significantly ameliorated vascular dysfunction, while ERβ knockdown worsened the problem. We conclude that perinatal testosterone exposure potentiates vascular dysfunction through ERβ suppression in EPCs.
Collapse
Affiliation(s)
- Weiguo Xie
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan, P.R.China
| | - Mingming Ren
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, P.R.China
| | - Ling Li
- Department of Pediatrics, Maternal and Child Health Care Hospital of Hainan Province, Haikou, P.R.China
| | - Yin Zhu
- Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Zhigang Chu
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan, P.R.China
| | - Zhigang Zhu
- Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Qiongfang Ruan
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan, P.R.China
| | - Wenting Lou
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan, P.R.China
| | - Haimou Zhang
- School of Life Sciences, Hubei University, Wuhan, P.R.China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, P.R.China
| | - Xiaodong Huang
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan, P.R.China
| | - Wei Xiang
- Department of Pediatrics, Maternal and Child Health Care Hospital of Hainan Province, Haikou, P.R.China
| | - Tao Wang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, P.R.China
| | - Paul Yao
- Institute of Burns, Tongren Hospital of Wuhan University, Wuhan, P.R.China
- Department of Pediatrics, Maternal and Child Health Care Hospital of Hainan Province, Haikou, P.R.China
| |
Collapse
|
26
|
Shao X, Liu Y, Liu M, Wang Y, Yan L, Wang H, Ma L, Li YX, Zhao Y, Wang YL. Testosterone Represses Estrogen Signaling by Upregulating miR-22. Hypertension 2017; 69:721-730. [DOI: 10.1161/hypertensionaha.116.08468] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/10/2016] [Accepted: 12/26/2016] [Indexed: 12/11/2022]
Abstract
Preeclampsia, a multisystem syndrome occurring during mid- to late gestation in humans, is a leading cause of maternal and perinatal morbidity and mortality. Patients usually present with high circulating testosterone and reduced estradiol production, but the mechanisms remain unclear. Revealing the mechanism that modulating the imbalance of testosterone and estradiol in preeclampsia is of great value in understanding the cause of the disease. The placenta is the predominant source of steroid hormone production during gestation, and we observed markedly increased 17β-HSD3 (17β-hydroxysteroid dehydrogenase 3) levels and downregulated aromatase expression, the key enzymes responsible for synthesis of testosterone and estradiol, respectively, in preeclamptic placentas compared with controls. Furthermore, we found a significant upregulation of microRNA (miR)-22 in preeclamptic placentas. In a trophoblast cell line, JEG-3 cells, testosterone repressed the expression of aromatase and estrogen receptor α and the production of estradiol while promoting miR-22 expression. miR-22 directly targeted and inhibited estrogen receptor α expression while indirectly decreasing aromatase expression and estradiol production by interfering with estrogen receptor α signaling. Furthermore, inhibition of miR-22 expression significantly reversed the inhibitory effect of testosterone on de novo estradiol synthesis in human trophoblastic cells. The findings reveal a mechanism underlying the balanced production of androgen and estrogen modulated by miR-22 in the human placenta and provide new insights into the pathogenesis of preeclampsia from the aspect of endocrine regulation.
Collapse
Affiliation(s)
- Xuan Shao
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yanlei Liu
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Ming Liu
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yongqing Wang
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Liying Yan
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Hao Wang
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Liyang Ma
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yu-xia Li
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yangyu Zhao
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yan-ling Wang
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| |
Collapse
|
27
|
Metzler VM, de Brot S, Robinson RS, Jeyapalan JN, Rakha E, Walton T, Gardner DS, Lund EF, Whitchurch J, Haigh D, Lochray JM, Robinson BD, Allegrucci C, Fray RG, Persson JL, Ødum N, Miftakhova RR, Rizvanov AA, Hughes IA, Tadokoro-Cuccaro R, Heery DM, Rutland CS, Mongan NP. Androgen dependent mechanisms of pro-angiogenic networks in placental and tumor development. Placenta 2017; 56:79-85. [PMID: 28238455 DOI: 10.1016/j.placenta.2017.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 11/25/2022]
Abstract
The placenta and tumors share important characteristics, including a requirement to establish effective angiogenesis. In the case of the placenta, optimal angiogenesis is required to sustain the blood flow required to maintain a successful pregnancy, whereas in tumors establishing new blood supplies is considered a key step in supporting metastases. Therefore the development of novel angiogenesis inhibitors has been an area of active research in oncology. A subset of the molecular processes regulating angiogenesis are well understood in the context of both early placentation and tumorigenesis. In this review we focus on the well-established role of androgen regulation of angiogenesis in cancer and relate these mechanisms to placental angiogenesis. The physiological actions of androgens are mediated by the androgen receptor (AR), a ligand dependent transcription factor. Androgens and the AR are essential for normal male embryonic development, puberty and lifelong health. Defects in androgen signalling are associated with a diverse range of clinical disorders in men and women including disorders of sex development (DSD), polycystic ovary syndrome in women and many cancers. We summarize the diverse molecular mechanisms of androgen regulation of angiogenesis and infer the potential significance of these pathways to normal and pathogenic placental function. Finally, we offer potential research applications of androgen-targeting molecules developed to treat cancer as investigative tools to help further delineate the role of androgen signalling in placental function and maternal and offspring health in animal models.
Collapse
Affiliation(s)
- Veronika M Metzler
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Simone de Brot
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Robert S Robinson
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Jennie N Jeyapalan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Emad Rakha
- School of Medicine and Sciences, University of Nottingham, Nottingham City Hospital, NG5 1PB, UK
| | - Thomas Walton
- Department of Urology, Nottingham University Hospitals NHS Trust, NG5 1PB, UK
| | - David S Gardner
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Emma F Lund
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | | | - Daisy Haigh
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Jack M Lochray
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Brian D Robinson
- Department of Pathology, Weill Cornell Medicine, New York 10065, USA
| | - Cinzia Allegrucci
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK
| | - Rupert G Fray
- School of Biosciences, University of Nottingham, LE12 5RD, UK
| | - Jenny L Persson
- Department of Translational Medicine, Lund University, Malmö, Sweden; Department of Molecular Biology, Umeå University, Sweden
| | - Niels Ødum
- Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Regina R Miftakhova
- Department of Molecular Biology, Umeå University, Sweden; Kazan Federal University, Kazan, Republic of Tatarstan 420008, Russian Federation
| | - Albert A Rizvanov
- Kazan Federal University, Kazan, Republic of Tatarstan 420008, Russian Federation
| | - Ieuan A Hughes
- Department of Paediatrics, University of Cambridge, Hills Rd, Cambridge CB2 0QQ, UK
| | | | - David M Heery
- School of Pharmacy, University of Nottingham, NG7 2TQ, UK
| | - Catrin S Rutland
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK.
| | - Nigel P Mongan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Health Sciences, University of Nottingham, LE12 5RD, UK; Department of Pharmacology, Weill Cornell Medicine, New York 10065, USA.
| |
Collapse
|
28
|
Hakim C, Padmanabhan V, Vyas AK. Gestational Hyperandrogenism in Developmental Programming. Endocrinology 2017; 158:199-212. [PMID: 27967205 PMCID: PMC5413081 DOI: 10.1210/en.2016-1801] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/07/2016] [Indexed: 12/16/2022]
Abstract
Androgen excess (hyperandrogenism) is a common endocrine disorder affecting women of reproductive age. The potential causes of androgen excess in women include polycystic ovary syndrome, congenital adrenal hyperplasia (CAH), adrenal tumors, and racial disparity among many others. During pregnancy, luteoma, placental aromatase deficiency, and fetal CAH are additional causes of gestational hyperandrogenism. The present report reviews the various phenotypes of hyperandrogenism during pregnancy and its origin, pathophysiology, and the effect of hyperandrogenism on the fetal developmental trajectory and offspring consequences.
Collapse
Affiliation(s)
- Christopher Hakim
- College of Human Medicine, Michigan State University, East Lansing, Michigan 48824
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48109; and
| | - Arpita K. Vyas
- College of Human Medicine, Michigan State University, East Lansing, Michigan 48824
- Department of Pediatrics, Texas Tech University Health Sciences Center, Permian Basin Campus, Odessa, Texas 79763
| |
Collapse
|
29
|
Mishra JS, Hankins GD, Kumar S. Testosterone downregulates angiotensin II type-2 receptor via androgen receptor-mediated ERK1/2 MAP kinase pathway in rat aorta. J Renin Angiotensin Aldosterone Syst 2016; 17:17/4/1470320316674875. [PMID: 27765882 PMCID: PMC5465964 DOI: 10.1177/1470320316674875] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/16/2016] [Indexed: 11/15/2022] Open
Abstract
Introduction: Blood pressure is lower in females than males. Angiotensin II type-2 receptor (AT2R) induces vasodilation. This study determined whether sex differences in vascular AT2R expression occur and if androgens exert control on AT2R expression in the vasculature. Methods: AT2Rs in the aorta of male and female Sprague-Dawley rats were examined following alteration in androgen levels by gonadectomy or hormone supplementation. Results: AT2R mRNA and protein expression levels were lower in the aortas of males than females. In males, testosterone withdrawal by castration significantly elevated AT2R mRNA and protein levels and testosterone replacement restored them. In females, increasing androgen levels decreased AT2R mRNA and protein expression and this was attenuated by androgen receptor blocker flutamide. Ex vivo, dihydrotestosterone downregulated AT2R in endothelium-intact but not endothelium-denuded aorta. Dihydrotestosterone-induced AT2R downregulation in isolated aorta was blocked by an androgen receptor antagonist. Furthermore, blockade of ERK1/2 but not p38 MAP kinase or TGFβ signaling with specific inhibitors abolished dihydrotestosterone-induced AT2R downregulation. Conclusion: Androgens downregulate AT2R expression levels in aorta, in vivo and ex vivo. The androgen receptor-mediated ERK1/2 MAP kinase-signaling pathway may be a key mechanism by which testosterone downregulates AT2R expression, implicating androgens’ contributing role to gender differences in vascular AT2R expression.
Collapse
Affiliation(s)
- Jay S Mishra
- Division of Reproductive Endocrinology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Gary D Hankins
- Division of Reproductive Endocrinology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Sathish Kumar
- Division of Reproductive Endocrinology, University of Texas Medical Branch at Galveston, Texas, USA
| |
Collapse
|
30
|
Dasinger JH, Davis GK, Newsome AD, Alexander BT. Developmental Programming of Hypertension: Physiological Mechanisms. Hypertension 2016; 68:826-31. [PMID: 27550912 DOI: 10.1161/hypertensionaha.116.06603] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- John Henry Dasinger
- From the Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson
| | - Gwendolyn K Davis
- From the Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson
| | - Ashley D Newsome
- From the Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson
| | - Barbara T Alexander
- From the Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson.
| |
Collapse
|
31
|
More AS, Mishra JS, Hankins GD, Kumar S. Prenatal Testosterone Exposure Decreases Aldosterone Production but Maintains Normal Plasma Volume and Increases Blood Pressure in Adult Female Rats. Biol Reprod 2016; 95:42. [PMID: 27385784 PMCID: PMC5029475 DOI: 10.1095/biolreprod.116.141705] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/29/2016] [Indexed: 12/14/2022] Open
Abstract
Plasma testosterone levels are elevated in pregnant women with preeclampsia and polycystic ovaries; their offspring are at increased risk for hypertension during adult life. We tested the hypothesis that prenatal testosterone exposure induces dysregulation of the renin-angiotensin-aldosterone system, which is known to play an important role in water and electrolyte balance and blood pressure regulation. Female rats (6 mo old) prenatally exposed to testosterone were examined for adrenal expression of steroidogenic genes, telemetric blood pressure, blood volume and Na+ and K+ levels, plasma aldosterone, angiotensin II and vasopressin levels, and vascular responses to angiotensin II and arg8-vasopressin. The levels of Cyp11b2 (aldosterone synthase), but not the other adrenal steroidogenic genes, were decreased in testosterone females. Accordingly, plasma aldosterone levels were lower in testosterone females. Plasma volume and serum and urine Na+ and K+ levels were not significantly different between control and testosterone females; however, prenatal testosterone exposure significantly increased plasma vasopressin and angiotensin II levels and arterial pressure in adult females. In testosterone females, mesenteric artery contractile responses to angiotensin II were significantly greater, while contractile responses to vasopressin were unaffected. Angiotensin II type-1 receptor expression was increased, while angiotensin II type-2 receptor was decreased in testosterone arteries. These results suggest that prenatal testosterone exposure downregulates adrenal Cyp11b2 expression, leading to decreased plasma aldosterone levels. Elevated angiotensin II and vasopressin levels along with enhanced vascular responsiveness to angiotensin II may serve as an underlying mechanism to maintain plasma volume and Na+ and K+ levels and mediate hypertension in adult testosterone females.
Collapse
Affiliation(s)
- Amar S More
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Jay S Mishra
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Gary D Hankins
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Sathish Kumar
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| |
Collapse
|
32
|
Li W, Lv J, Wu J, Zhou X, Jiang L, Zhu X, Tu Q, Tang J, Liu Y, He A, Zhong Y, Xu Z. Maternal high-salt diet altered PKC/MLC20 pathway and increased ANG II receptor-mediated vasoconstriction in adult male rat offspring. Mol Nutr Food Res 2016; 60:1684-94. [PMID: 26991838 DOI: 10.1002/mnfr.201500998] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/06/2016] [Accepted: 02/09/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Weisheng Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Juanxiu Lv
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jue Wu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiuwen Zhou
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Lin Jiang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiaolin Zhu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Qing Tu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jiaqi Tang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yanping Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Axin He
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yuan Zhong
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
- Center for Prenatal Biology, Loma Linda University, CA, USA
| |
Collapse
|
33
|
Gopalakrishnan K, Mishra JS, Chinnathambi V, Vincent KL, Patrikeev I, Motamedi M, Saade GR, Hankins GD, Sathishkumar K. Elevated Testosterone Reduces Uterine Blood Flow, Spiral Artery Elongation, and Placental Oxygenation in Pregnant Rats. Hypertension 2016; 67:630-9. [PMID: 26781277 DOI: 10.1161/hypertensionaha.115.06946] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
Elevated maternal testosterone levels are shown to cause fetal growth restriction, eventually culminating in sex-specific adult-onset hypertension that is more pronounced in males than in females. In this study, we tested whether uteroplacental and fetoplacental disturbances underlie fetal growth restriction and if these changes vary in male and female placentas. Pregnant Sprague-Dawley rats were injected with vehicle (n=16) or testosterone propionate (0.5 mg/kg per day from gestation day 15-19; n=16). On gestation day 20, we quantified uterine artery blood flow using microultrasound, visualized placental arterial network using x-ray microcomputed tomography, determined fetoplacental hypoxia using pimonidazole and hypoxia-inducible factor-1α, and used Affymetrix array to determine changes in placental expression of genes involved in vascular development. Plasma testosterone levels increased 2-fold in testosterone-injected rats. Placental and fetal weights were lower in rats with elevated testosterone. Uterine artery blood flow was lower, and resistance index was higher in the testosterone group. Radial and spiral artery diameter and length, the number of fetoplacental arterial branches, and umbilical artery diameter were reduced in the testosterone group. In addition, markers of hypoxia in the placentas and fetuses were elevated in the testosterone group. The magnitude of changes in placental vasculature and hypoxia was greater in males than in females and was associated with sex-specific alteration of unique sets of genes involved in angiogenesis and blood vessel morphogenesis. The results demonstrate that elevated testosterone during gestation induces a decrease in uterine arterial blood flow and fetal sex-related uteroplacental vascular changes, which may set the stage for subsequent sex differences in adult-onset diseases.
Collapse
Affiliation(s)
- Kathirvel Gopalakrishnan
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Jay S Mishra
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Vijayakumar Chinnathambi
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Kathleen L Vincent
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Igor Patrikeev
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Massoud Motamedi
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - George R Saade
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Gary D Hankins
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Kunju Sathishkumar
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston.
| |
Collapse
|
34
|
More AS, Mishra JS, Gopalakrishnan K, Blesson CS, Hankins GD, Sathishkumar K. Prenatal Testosterone Exposure Leads to Gonadal Hormone-Dependent Hyperinsulinemia and Gonadal Hormone-Independent Glucose Intolerance in Adult Male Rat Offspring. Biol Reprod 2015; 94:5. [PMID: 26586841 PMCID: PMC4809560 DOI: 10.1095/biolreprod.115.133157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/06/2015] [Indexed: 12/14/2022] Open
Abstract
Elevated testosterone levels during prenatal life lead to hyperandrogenism and insulin resistance in adult females. This study evaluated whether prenatal testosterone exposure leads to the development of insulin resistance in adult male rats in order to assess the influence of gonadal hormones on glucose homeostasis in these animals. Male offspring of pregnant rats treated with testosterone propionate or its vehicle (control) were examined. A subset of male offspring was orchiectomized at 7 wk of age and reared to adulthood. At 24 wk of age, fat weights, plasma testosterone, glucose homeostasis, pancreas morphology, and gastrocnemius insulin receptor (IR) beta levels were examined. The pups born to testosterone-treated mothers were smaller at birth and remained smaller through adult life, with levels of fat deposition relatively similar to those in controls. Testosterone exposure during prenatal life induced hyperinsulinemia paralleled by an increased HOMA-IR index in a fasting state and glucose intolerance and exaggerated insulin responses following a glucose tolerance test. Prenatal androgen-exposed males had more circulating testosterone during adult life. Gonadectomy prevented hyperandrogenism, reversed hyperinsulinemia, and attenuated glucose-induced insulin responses but did not alter glucose intolerance in these rats. Prenatal androgen-exposed males had decreased pancreatic islet numbers, size, and beta-cell area along with decreased expression of IR in gastrocnemius muscles. Gonadectomy restored pancreatic islet numbers, size, and beta-cell area but did not normalize IRbeta expression. This study shows that prenatal testosterone exposure leads to a defective pancreas and skeletal muscle function in male offspring. Hyperinsulinemia during adult life is gonad-dependent, but glucose intolerance appears to be independent of postnatal testosterone levels.
Collapse
Affiliation(s)
- Amar S More
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas
| | - Jay S Mishra
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas
| | - Kathirvel Gopalakrishnan
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas
| | - Chellakkan S Blesson
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| | - Gary D Hankins
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas
| | - Kunju Sathishkumar
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|