1
|
Pu Y, Du Y, He J, He S, Chen Y, Cao A, Dang Y. The mediating role of steroid hormones in the relationship between bisphenol A and its alternatives bisphenol S and F exposure and preeclampsia. J Steroid Biochem Mol Biol 2024; 244:106591. [PMID: 39059562 DOI: 10.1016/j.jsbmb.2024.106591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Steroid hormone imbalance is believed to increase the odds of developing PE. Bisphenol A (BPA) and its substitutes (e.g., bisphenol S (BPS) and bisphenol F (BPF)) have estrogen-like effects, and its exposure may be related to the development of preeclampsia (PE). To explore the effects of bisphenol exposure on maternal serum steroid hormones and the potential mediating role of steroid hormones in the association between bisphenol exposure and developing PE, concentrations of bisphenols and steroid hormones in serum samples of 383 pregnant women were examined before delivery (including 160 PE cases and 223 control cases). Multivariable logistic and linear models were used to explore the associations of maternal serum bisphenols concentrations with both maternal steroid hormones and PE risk. Mediation modeling was employed to evaluate the mediating role of steroid hormones in the association between bisphenols and PE. Results showed that maternal serum BPS concentrations were positively associated with testosterone (T) concentrations. The mediation analyses suggested that approximately 10.17 % of the associations between BPS concentrations and the development of PE might be mediated by maternal T. In conclusion, maternal exposure to BPS during pregnancy is linked to higher maternal T concentrations, which might increase the odds of developing PE. T might mediate the association between BPS exposure and the development of PE.
Collapse
Affiliation(s)
- Yudong Pu
- Institute of The Songshan Lake Central Hospital of Dongguan City, Dongguan 523326, China.
| | - Yue Du
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| | - Jie He
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| | - Shuzhen He
- Institute of The Songshan Lake Central Hospital of Dongguan City, Dongguan 523326, China.
| | - Ya Chen
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| | - Aitong Cao
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| | - Yuhui Dang
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Zhao X, Xu A, Lu X, Chen B, Hua Y, Ma Y. Association of phthalates exposure and sex steroid hormones with late-onset preeclampsia: a case-control study. BMC Pregnancy Childbirth 2024; 24:577. [PMID: 39227873 PMCID: PMC11369995 DOI: 10.1186/s12884-024-06793-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND This study aimed to investigate the relationship between phthalates exposure and estrogen and progesterone levels, as well as their role in late-onset preeclampsia. METHODS A total of 60 pregnant women who met the inclusion and exclusion criteria were recruited. Based on the diagnosis of preeclampsia, participants were divided into two groups: normotensive pregnant women (n = 30) and pregnant women with late-onset preeclampsia (n = 30). The major metabolites of phthalates (MMP, MEP, MiBP, MBP, MEHP, MEOHP, MEHHP) and sex steroid hormones (estrogen and progesterone) were quantified in urine samples of the participants. RESULTS No significant differences were observed in the levels of MMP, MEP, MiBP, MBP, MEHP, MEOHP, and MEHHP between women with preeclampsia and normotensive pregnant women (P > 0.05). The urinary estrogen showed a negative correlation with systolic blood pressure (rs= -0.46, P < 0.001) and diastolic blood pressure (rs= -0.47, P < 0.001). Additionally, the urinary estrogen and progesterone levels were lower in women with preeclampsia compared to those in normotensive pregnant women (P < 0.05). After adjusting for confounding factors, we observed a significant association between reduced urinary estrogen levels and an increased risk of preeclampsia (aOR = 0.09, 95%CI = 0.02-0.46). Notably, in our decision tree model, urinary estrogen emerged as the most crucial variable for identifying pregnant women at a high risk of developing preeclampsia. A positive correlation was observed between urinary progesterone and MEHP (rs = 0.36, P < 0.05) in normotensive pregnant women. A negative correlation was observed between urinary estrogen and MEP in pregnant women with preeclampsia (rs= -0.42, P < 0.05). CONCLUSIONS Phthalates exposure was similar in normotensive pregnant women and those with late-onset preeclampsia within the same region. Pregnant women with preeclampsia had lower levels of estrogen and progesterone in their urine, while maternal urinary estrogen was negatively correlated with the risk of preeclampsia and phthalate metabolites (MEP). TRIAL REGISTRATION Registration ID in Clinical Trials: NCT04369313; registration date: 30/04/2020.
Collapse
Affiliation(s)
- Xiaomin Zhao
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Department of Obstetrics and Gynecology, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, China
| | - Anjian Xu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xinyue Lu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Baoyi Chen
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ying Hua
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Yanyan Ma
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
3
|
Mineiro R, Cardoso MR, Pinheiro JV, Cipolla-Neto J, do Amaral FG, Quintela T. Overlapping action of melatonin and female reproductive hormones-Understand the impact in pregnancy and menopause. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:163-190. [PMID: 39059985 DOI: 10.1016/bs.apcsb.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Melatonin is an indolamine secreted to circulation by the pineal gland according to a circadian rhythm. Melatonin levels are higher during nighttime, and the principal function of this hormone is to organize the temporal night and day distribution of physiological adaptive processes. Besides hormonal pineal production, melatonin is synthesized in various organs and tissues like the ovaries or the placenta for local utilization. In addition to its function as a circadian messenger, melatonin is also associated with many physiological functions. For example, melatonin has antioxidant properties and is involved in the regulation of energy and bone metabolism, and reproduction. Melatonin impacts several stages of reproduction and the action across the hypothalamus-pituitary-gonadal axis is well described. However, it is not well understood how those actions impact the female reproductive hormones secretion nor the consequent physiological outcomes. Thus, the first part of this chapter describes the regulation of female reproductive hormone synthesis by melatonin. Moreover, melatonin and female reproductive hormones have coincident physiological functions. Life stages like pregnancy or menopause are characterized by alterations in the reproductive hormones secretion that may be associated with certain physiological stages. Therefore, the second part discusses whether melatonin fluctuations could have an overlapping role with reproductive hormones in contributing to clinical outcomes associated with pregnancy and menopause.
Collapse
Affiliation(s)
- Rafael Mineiro
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | | | - João Vieira Pinheiro
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Telma Quintela
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal; Instituto Politécnico da Guarda, Guarda, Portugal.
| |
Collapse
|
4
|
KAN Y, PENG YL, ZHAO ZH, DONG ST, XU YX, MA XT, LIU XL, LIU YY, ZHOU YJ. The impact of female sex hormones on cardiovascular disease: from mechanisms to hormone therapy. J Geriatr Cardiol 2024; 21:669-681. [PMID: 38973823 PMCID: PMC11224657 DOI: 10.26599/1671-5411.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
Cardiovascular disease remains the leading cause of mortality in women, yet it has not raised the awareness from the public. The pathogenesis of cardiovascular disease differs significantly between females and males concerning the effect of sex hormones. Estrogen and progestogen impact cardiovascular system through genomic and non-genomic effects. Before menopause, cardiovascular protective effects of estrogens have been well described. Progestogens were often used in combination with estrogens in hormone therapy. Fluctuations in sex hormone levels, particularly estrogen deficiency, were considered the specific risk factor in women's cardiovascular disease. However, considerable heterogeneity in the impact of hormone therapy was observed in clinical trials. The heterogeneity is likely closely associated with factors such as the initial time, administration route, dosage, and formulation of hormone therapy. This review will delve into the pathogenesis and hormone therapy, summarizing the effect of female sex hormones on hypertension, pre-eclampsia, coronary heart disease, heart failure with preserved ejection fraction, and cardiovascular risk factors specific to women.
Collapse
Affiliation(s)
- Yi KAN
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Yu-Lu PENG
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Ze-Hao ZHAO
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Shu-Tong DONG
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Yin-Xiao XU
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Xiao-Teng MA
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Xiao-Li LIU
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Yu-Yang LIU
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Yu-Jie ZHOU
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Zhou L, Liu X, He G, Sun C. Causal effects of hypertensive disorders of pregnancy on future gynecologic tumors: A two-sample Mendelian randomization study. Cancer Med 2024; 13:e7300. [PMID: 38800978 PMCID: PMC11129165 DOI: 10.1002/cam4.7300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/02/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Numerous observational studies have investigated the potential link between hypertensive disorders of pregnancy (HDPs) and the subsequent risks of gynecologic tumors, yet the findings have been inconsistent. In this study, we utilized Mendelian randomization (MR) approach to assess the influence of HDPs on the future risks of ovarian, cervical, endometrial, and breast cancer and uterine fibroids, controlling for confounding factors. METHODS The genome-wide association studies (GWAS) summary data relevant to HDPs was obtained from the FinnGen databases (10,736 cases and 136,325 controls). Gynecologic tumor outcomes were extracted from the IEU Open GWAS project and UK Biobank (47,690 cases and 1, 092,073 controls). The inverse variance weighted (IVW) approach was selected as the principal method for MR analysis, supplemented by MR-Egger, weighted median, weighted model, simple model methods, MR pleiotropy residual sum and outlier (MR-PRESSO) test, and leave-one-out method. Multivariate MR (MVMR) analysis was conducted after adjusting systolic blood pressure (SBP), body mass index (BMI) and type 2 diabetes mellitus (T2DM). RESULTS Our univariate MR analysis (UVMR) results revealed no significant relationship between HDPs and the risks of ovarian cancer (odds ratio [OR] = 0.924, p = 0.360), cervical cancer (OR = 1.230, p = 0.738), endometrial cancer (OR = 1.006, p = 0.949), uterine fibroids (OR = 1.155, p = 0.158), and breast cancer (OR = 0.792, p = 0.241) by IVW test. Similar results were observed in gestational hypertension and preeclampsia/eclampsia. Additionally, our study detected neither heterogeneity nor pleiotropy. MVMR analysis also provided no evidence of a causal association between HDPs and common gynecologic tumors after adjusting SBP, BMI, and T2DM. CONCLUSION We discovered no causal relationship between HDPs and ovarian, cervical, endometrial, breast cancer, and uterine fibroids in European populations. However, present analysis did not explore the effect of HDPs on the subtypes of gynecologic tumors across varied ethnic populations, which may require additional research.
Collapse
Affiliation(s)
- Le Zhou
- Department of Gynecology and ObstetricsWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Obstetrics & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationWest China Second University Hospital, Sichuan UniversityChengduChina
| | - Xinghui Liu
- Department of Gynecology and ObstetricsWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Obstetrics & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationWest China Second University Hospital, Sichuan UniversityChengduChina
| | - Guolin He
- Department of Gynecology and ObstetricsWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Obstetrics & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationWest China Second University Hospital, Sichuan UniversityChengduChina
| | - Chuntang Sun
- Department of Gynecology and ObstetricsWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Obstetrics & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationWest China Second University Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
6
|
Gomes VCL, Gilbert BM, Bernal C, Crissman KR, Sones JL. Estrogen and Progesterone Receptors Are Dysregulated at the BPH/5 Mouse Preeclamptic-Like Maternal-Fetal Interface. BIOLOGY 2024; 13:192. [PMID: 38534461 DOI: 10.3390/biology13030192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024]
Abstract
The etiopathogenesis of preeclampsia, a leading hypertensive disorder of pregnancy, has been proposed to involve an abnormal circulating sex hormone profile and misexpression of placental estrogen and progesterone receptors (ER and PR, respectively). However, existing research is vastly confined to third trimester preeclamptic placentas. Consequently, the placental-uterine molecular crosstalk and the dynamic ER and PR expression pattern in the peri-conception period remain overlooked. Herein, our goal was to use the BPH/5 mouse to elucidate pre-pregnancy and early gestation Er and Pr dynamics in a preeclamptic-like uterus. BPH/5 females display low circulating estrogen concentration during proestrus, followed by early gestation hypoestrogenemia, hyperprogesteronemia, and a spontaneous preeclamptic-like phenotype. Preceding pregnancy, the gene encoding Er alpha (Erα, Esr1) is upregulated in the diestrual BPH/5 uterus. At the peak of decidualization, Esr1, Er beta (Erβ, Esr2), and Pr isoform B (Pr-B) were upregulated in the BPH/5 maternal-fetal interface. At the protein level, BPH/5 females display higher percentage of decidual cells with nuclear Erα expression, as well as Pr downregulation in the decidua, luminal and glandular epithelium. In conclusion, we provide evidence of disrupted sex hormone signaling in the peri-conception period of preeclamptic-like pregnancies, potentially shedding some light onto the intricate role of sex hormone signaling at unexplored timepoints of human preeclampsia.
Collapse
Affiliation(s)
- Viviane C L Gomes
- Department of Small Animal Clinical Sciences, Michigan State University College of Veterinary Medicine, East Lansing, MI 48824, USA
| | - Bryce M Gilbert
- Department of Small Animal Clinical Sciences, Michigan State University College of Veterinary Medicine, East Lansing, MI 48824, USA
| | - Carolina Bernal
- Department of Small Animal Clinical Sciences, Michigan State University College of Veterinary Medicine, East Lansing, MI 48824, USA
| | - Kassandra R Crissman
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Jenny L Sones
- Equine Reproduction Laboratory, Colorado State University, Fort Collins, CO 80521, USA
| |
Collapse
|
7
|
Sewnarain S, Singh S, Naicker T. Placental progesterone and its receptor in HIV-infected pre-eclamptic women. Histochem Cell Biol 2024; 161:255-267. [PMID: 37975897 PMCID: PMC10912128 DOI: 10.1007/s00418-023-02250-0] [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] [Accepted: 10/11/2023] [Indexed: 11/19/2023]
Abstract
Given the high prevalence of HIV infection and pre-eclampsia (PE) in South Africa, this study evaluated and compared the placental immunostaining of progesterone (P) and progesterone receptors (PR) in the synergy of HIV-infected PE compared to normotensive pregnant women using immunohistochemistry interfaced with morphometric image analysis. Progesterone immunostaining was expressed widely across exchange and conducting villi within mesenchymal, endothelial, and trophoblast cells. In contrast, PR was expressed within syncytiotrophoblasts and was absent within endothelial cells. In exchange villi, P and PR immuno-expression was significantly lower in PE compared to the normotensive group (p = < 0.0001 and p = < 0.0001, respectively) and within the early-onset pre-eclampsia (EOPE) compared to the late-onset pre-eclampsia (LOPE) group (p = < 0.0001 and p = < 0.0001, respectively). Progesterone immuno-expression was significantly lower in the HIV+ compared to the HIV- group (p = < 0.0001), whilst PR was non-significant. In conducting villi, P and PR immuno-expression was significantly lower in the EOPE compared to the LOPE group (p = < 0.0001 and p = < 0.0001, respectively) and in the HIV+ compared to the HIV- group (p = < 0.0001 and p = 0.0009, respectively). Progesterone immuno-expression was slightly higher in the PE compared to normotensive group, and PR immuno-expression was non-significant. There was a significant difference between P and PR within exchange versus conducting villi regardless of pregnancy type, with villi type accounting for 34.47% and 15.28% of total variance for P and PR, respectively. Placental P and PR immuno-expression is downregulated in the duality of PE and HIV+ infection. The use of combined antiretroviral therapy (cART) may result in defective P synthesis, which causes insufficient binding to its receptors. Consequently, PI3K/AKT, JAK-STAT, and MAPK signalling pathways are affected, impairing trophoblast invasion and leading to pre-eclampsia development. Notably, the decrease in P and PR immuno-expression in EOPE validates their effect on placentation.
Collapse
Affiliation(s)
- Serisha Sewnarain
- Optics and Imaging Centre, College of Health Sciences, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Private Bag X7, Congella, Durban, 4013, KwaZulu-Natal, South Africa.
| | - Shoohana Singh
- Optics and Imaging Centre, College of Health Sciences, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Private Bag X7, Congella, Durban, 4013, KwaZulu-Natal, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, College of Health Sciences, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Private Bag X7, Congella, Durban, 4013, KwaZulu-Natal, South Africa
| |
Collapse
|
8
|
Boyle P, Andralojc K, van der Velden S, Najmabadi S, de Groot T, Turczynski C, Stanford JB. Restoration of serum estradiol and reduced incidence of miscarriage in patients with low serum estradiol during pregnancy: a retrospective cohort study using a multifactorial protocol including DHEA. FRONTIERS IN REPRODUCTIVE HEALTH 2024; 5:1321284. [PMID: 38239818 PMCID: PMC10794495 DOI: 10.3389/frph.2023.1321284] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/08/2023] [Indexed: 01/22/2024] Open
Abstract
Background Low serum estradiol in early pregnancy is associated with an elevated risk of miscarriage. We sought to determine whether efforts to restore low blood estradiol via estradiol or dehydroepiandrosterone (DHEA) supplementation would reduce the risk of miscarriage as part of a multifactorial symptom-based treatment protocol. Methods This retrospective cohort study included women with low serum estradiol levels in early pregnancy, defined as ≤50% of reference levels by gestational age. Estradiol or DHEA were administered orally, and the primary outcome measure was serum estradiol level, in reference to gestational age. The secondary outcome measures included miscarriage, birth weight, and gestational age at birth. Results We found no significant effect of estradiol supplementation on serum estradiol levels referenced to gestational age, while DHEA supplementation strongly increased estradiol levels. For pregnancies with low estradiol, the miscarriage rate in the non-supplemented group was 45.5%, while miscarriage rate in the estradiol and DHEA supplemented groups were 21.2% (p = 0.067) and 17.5% (p = 0.038), respectively. Birth weight, size, gestational age, and preterm deliveries were not significantly different. No sexual abnormalities were reported in children (n = 29) of DHEA-supplemented patients after 5-7 years follow-up. Conclusions In conclusion, DHEA supplementation restored serum estradiol levels, and when included in the treatment protocol, there was a statistically significant reduction in miscarriage.
Collapse
Affiliation(s)
- Phil Boyle
- International Institute for Restorative Reproductive Medicine, London, United Kingdom
- NeoFertility Clinic, Dublin, Ireland
| | - Karolina Andralojc
- NeoFertility Clinic, Dublin, Ireland
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Shahpar Najmabadi
- Office of Cooperative Reproductive Health, Department of Family and Preventive Medicine, University of Utah, Salt Lake City, UT, United States
| | - Theun de Groot
- NeoFertility Clinic, Dublin, Ireland
- Department of Medical Microbiology and Infectious Diseases, Canisius Wilhelmina Hospital, Nijmegen, Netherlands
| | - Craig Turczynski
- NeoFertility Clinic, Dublin, Ireland
- Billings Ovulation Method Association-USA, Saint Cloud, MN, United States
| | - Joseph B. Stanford
- International Institute for Restorative Reproductive Medicine, London, United Kingdom
- Office of Cooperative Reproductive Health, Department of Family and Preventive Medicine, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
9
|
Oh Y, Quiroz E, Wang T, Medina-Laver Y, Redecke SM, Dominguez F, Lydon JP, DeMayo FJ, Wu SP. The NR2F2-HAND2 signaling axis regulates progesterone actions in the uterus at early pregnancy. Front Endocrinol (Lausanne) 2023; 14:1229033. [PMID: 37664846 PMCID: PMC10473531 DOI: 10.3389/fendo.2023.1229033] [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: 05/25/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
Endometrial function is dependent on a tight crosstalk between the epithelial and stromal cells of the endometrium. This communication is critical to ensure a fertile uterus and relies on progesterone and estrogen signaling to prepare a receptive uterus for embryo implantation in early pregnancy. One of the key mediators of this crosstalk is the orphan nuclear receptor NR2F2, which regulates uterine epithelial receptivity and stromal cell differentiation. In order to determine the molecular mechanism regulated by NR2F2, RNAseq analysis was conducted on the uterus of PgrCre;Nr2f2f/f mice at Day 3.5 of pregnancy. This transcriptomic analysis demonstrated Nr2f2 ablation in Pgr-expressing cells leads to a reduction of Hand2 expression, increased levels of Hand2 downstream effectors Fgf1 and Fgf18, and a transcriptome manifesting suppressed progesterone signaling with an altered immune baseline. ChIPseq analysis conducted on the Day 3.5 pregnant mouse uterus for NR2F2 demonstrated the majority of NR2F2 occupies genomic regions that have H3K27ac and H3K4me1 histone modifications, including the loci of major uterine transcription regulators Hand2, Egr1, and Zbtb16. Furthermore, functional analysis of an NR2F2 occupying site that is conserved between human and mouse was capable to enhance endogenous HAND2 mRNA expression with the CRISPR activator in human endometrial stroma cells. These data establish the NR2F2 dependent regulation of Hand2 in the stroma and identify a cis-acting element for this action. In summary, our findings reveal a role of the NR2F2-HAND2 regulatory axis that determines the uterine transcriptomic pattern in preparation for the endometrial receptivity.
Collapse
Affiliation(s)
- Yeongseok Oh
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
- Department of Life Science and Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Elvis Quiroz
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Tianyuan Wang
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Yassmin Medina-Laver
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Skylar Montague Redecke
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Francisco Dominguez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Francesco J. DeMayo
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - San-Pin Wu
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| |
Collapse
|
10
|
Lopian M, Kashani-Ligumsky L, Many A. A Balancing Act: Navigating Hypertensive Disorders of Pregnancy at Very Advanced Maternal Age, from Preconception to Postpartum. J Clin Med 2023; 12:4701. [PMID: 37510816 PMCID: PMC10380965 DOI: 10.3390/jcm12144701] [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: 04/21/2023] [Revised: 05/31/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The decision to postpone parenting has gained momentum in recent years, a shift driven by evolving social dynamics and improved access to fertility treatments. Despite their increasing prevalence, pregnancies at advanced maternal ages are associated with increased risks of adverse maternal and neonatal outcomes. This article addresses the association between advanced maternal age and hypertensive disorders of pregnancies (HDPs), which are more prevalent and a significant cause of maternal morbidity and mortality in this population. This review explores the biological mechanisms and age-related risk factors that underpin this increased susceptibility and offers practical management strategies that can be implemented prior to, as well as during, each stage of pregnancy to mitigate the incidence and severity of HDPs in this group. Lastly, this review acknowledges both the short-term and long-term postpartum implications of HDPs in women of advanced maternal age.
Collapse
Affiliation(s)
- Miriam Lopian
- Department of Obstetrics and Gynecology, Mayanei Hayeshua Medical Center, Bnei Brak 51544, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lior Kashani-Ligumsky
- Department of Obstetrics and Gynecology, Mayanei Hayeshua Medical Center, Bnei Brak 51544, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ariel Many
- Department of Obstetrics and Gynecology, Mayanei Hayeshua Medical Center, Bnei Brak 51544, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
11
|
Jordao H, Herink K, Ka E, McVicker L, Kearns C, McMenamin ÚC. Pre-eclampsia during pregnancy and risk of endometrial cancer: a systematic review and meta-analysis. BMC Womens Health 2023; 23:259. [PMID: 37173714 PMCID: PMC10182685 DOI: 10.1186/s12905-023-02408-x] [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: 01/16/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Pre-eclampsia may be associated with the development of endometrial cancer; however, previous findings have been conflicting. OBJECTIVES To investigate if pre-eclampsia is associated with an increased risk of endometrial cancer. METHOD Two independent reviewers screened titles and abstracts of studies identified in MEDLINE, Embase, and Web of Science databases from inception until March 2022. Studies were included if they investigated pre-eclampsia and subsequent risk of endometrial cancer (or precursor lesions). Random-effects meta-analysis was used to calculate pooled hazard ratios (HRs) and 95% confidence intervals (CIs) for the association between pre-eclampsia during pregnancy and endometrial cancer risk. MAIN RESULTS There were seven articles identified which investigated endometrial cancer, of which one also investigated endometrial cancer precursors. Overall, the studies include 11,724 endometrial cancer cases. No association was observed between pre-eclampsia and risk of endometrial cancer with moderate heterogeneity observed (pooled HR 1.07, 95% CI 0.79-1.46, I2 = 34.1%). In sensitivity analysis investigating risk of endometrial neoplasia (atypical hyperplasia, carcinoma in situ, or cancer), there was some evidence that pre-eclampsia was associated with an increased risk (HR 1.34, 95% CI 1.15-1.57, I2 = 29.6%). CONCLUSIONS Pre-eclampsia was not associated with an increased risk of endometrial cancer. Additional large studies with information on pre-eclampsia sub-type aiming to investigate endometrial cancer precursor conditions are merited.
Collapse
Affiliation(s)
- H Jordao
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences-B Building, Royal Victoria Hospital site, Grosvenor Rd, Belfast, Northern Ireland, BT12 6BJ, UK.
| | - K Herink
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences-B Building, Royal Victoria Hospital site, Grosvenor Rd, Belfast, Northern Ireland, BT12 6BJ, UK
| | - Eastwood Ka
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences-B Building, Royal Victoria Hospital site, Grosvenor Rd, Belfast, Northern Ireland, BT12 6BJ, UK
- Department of St. Michael's Hospital, Bristol, UK
| | - L McVicker
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences-B Building, Royal Victoria Hospital site, Grosvenor Rd, Belfast, Northern Ireland, BT12 6BJ, UK
| | - C Kearns
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences-B Building, Royal Victoria Hospital site, Grosvenor Rd, Belfast, Northern Ireland, BT12 6BJ, UK
| | - Ú C McMenamin
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences-B Building, Royal Victoria Hospital site, Grosvenor Rd, Belfast, Northern Ireland, BT12 6BJ, UK
| |
Collapse
|
12
|
Main C, Chen X, Zhao M, Chamley LW, Chen Q. Understanding How Pregnancy Protects Against Ovarian and Endometrial Cancer Development: Fetal Antigens May Be Involved. Endocrinology 2022; 163:6675223. [PMID: 36004540 PMCID: PMC9574549 DOI: 10.1210/endocr/bqac141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Indexed: 11/19/2022]
Abstract
It is well known that many factors, including infertility, obesity, type 2 diabetes, and family history of cancer, increase the risk of developing endometrial and ovarian cancer. However, multiparous women are known to have a lower risk of developing either ovarian or endometrial cancer than nonparous women. The lack of ovulation and shifting of sex hormonal balance, with decreased estrogen levels and increased progesterone levels during pregnancy, has traditionally been thought to be the major contributor to this decreased risk. However, in reality, the mechanisms underlying this phenomenon are relatively unknown. Increasing evidence suggests that endocrine factors are unlikely to completely explain the protective effect of pregnancies, and that multiple other nonendocrine mechanisms including fetal antigens and the newly proposed dormant cells hypothesis may also be involved. In this review, we summarize recent evidence and describe the potential underlying mechanisms that may explain how pregnancy protects against the development of ovarian and endometrial cancers in women's later life.
Collapse
Affiliation(s)
- Claudia Main
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Science, The University of Auckland, Auckland 1141, New Zealand
| | - Xinyue Chen
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Science, The University of Auckland, Auckland 1141, New Zealand
| | - Min Zhao
- Department of Gynecological Cancer, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing 214002, China
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Science, The University of Auckland, Auckland 1141, New Zealand
| | - Qi Chen
- Correspondence: Qi Chen, MD, PhD, Department of Obstetrics and Gynaecology, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand. ; or Min Zhao, MD, PhD, Department of Gynecological Cancer, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, China.
| |
Collapse
|
13
|
Zhang X, Chen K, Meng Z, Jia R, Lian F, Lin F. Cadmium-induced preeclampsia-like phenotype in the rat is related to decreased progesterone synthesis in the placenta. Xenobiotica 2022; 52:625-632. [DOI: 10.1080/00498254.2022.2124204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Xiaojie Zhang
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Kai Chen
- Wenzhou Medical University, Wenzhou 325000, China
| | - Zhu Meng
- Wenzhou Medical University, Wenzhou 325000, China
| | - Ru Jia
- Wenzhou Medical University, Wenzhou 325000, China
| | - Feifei Lian
- Wenzhou Medical University, Wenzhou 325000, China
| | - Feng Lin
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
14
|
Cheng W, Zhang L, Sa P, Luo J, Li M. Transcriptomic analysis reveals the effects of maternal selenium deficiency on placental transport, hormone synthesis, and immune response in mice. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6674774. [PMID: 36002020 DOI: 10.1093/mtomcs/mfac062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/19/2022] [Indexed: 11/14/2022]
Abstract
Selenium deficiency has been considered to increase the risk of gestational complications. Our previous work showed that maternal selenium deficiency suppressed proliferation, induced autophagy dysfunction and apoptosis in the placenta of mice. However, other effects of maternal selenium deficiency on the placenta and the underlying mechanisms remain unclear. In the present study, dietary selenium deficiency in dams significantly suppressed glutathione peroxidase (GSH-Px) activity, total antioxidant capacity (T-AOC), and increased malondialdehyde (MDA) content in the placentae, confirming the oxidative stress in the placenta. By transcriptome sequencing analysis, the DEGs were involved in many biological processes, including ion transport, lipid metabolic process, immune response, transmembrane transport, and others. According to the KEGG analysis, the DEGs were primarily enriched in metabolic pathways, PI3K-Akt signaling pathway, and others. Among these, the steroid hormone biosynthesis pathway enriched the most DEGs. Hsd3b1, an ER enzyme involved in progesterone synthesis, was validated downregulated. Consistently, the progesterone content in the serum of the selenium-deficient group was decreased. Ion transporters and transmembrane transporters, such as Heph, Trf, Slc39a8, Slc23a1, Atp7b, and Kcnc1, were reduced in the selenium-deficient placentae. Immune response-related genes, including Ccl3, Ccl8, Cxcl10, and Cxcl14, were increased in the selenium-deficient placentae, along with an increase in macrophage number. These results suggested that maternal selenium deficiency may impair progesterone biosynthesis, reduce nutrient transporters expression, and promote immune response by increasing the oxidative stress of the placentae. This present study provides a novel insight into the possible cause of placenta disorder during pregnancy.
Collapse
Affiliation(s)
- Wanpeng Cheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lantian Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Peiyue Sa
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jing Luo
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Mengdi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
15
|
Flavonoids exert potential in the management of hypertensive disorders in pregnancy. Pregnancy Hypertens 2022; 29:72-85. [DOI: 10.1016/j.preghy.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/29/2022] [Indexed: 11/19/2022]
|
16
|
Arleevskaya M, Takha E, Petrov S, Kazarian G, Renaudineau Y, Brooks W, Larionova R, Korovina M, Valeeva A, Shuralev E, Mukminov M, Kravtsova O, Novikov A. Interplay of Environmental, Individual and Genetic Factors in Rheumatoid Arthritis Provocation. Int J Mol Sci 2022; 23:ijms23158140. [PMID: 35897715 PMCID: PMC9329780 DOI: 10.3390/ijms23158140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023] Open
Abstract
In this review, we explore systemization of knowledge about the triggering effects of non-genetic factors in pathogenic mechanisms that contribute to the development of rheumatoid arthritis (RA). Possible mechanisms involving environmental and individual factors in RA pathogenesis were analyzed, namely, infections, mental stress, sleep deprivation ecology, age, perinatal and gender factors, eating habits, obesity and smoking. The non-genetic factors modulate basic processes in the body with the impact of these factors being non-specific, but these common challenges may be decisive for advancement of the disease in the predisposed body at risk for RA. The provocation of this particular disease is associated with the presence of congenital loci minoris resistentia. The more frequent non-genetic factors form tangles of interdependent relationships and, thereby, several interdependent external factors hit one vulnerable basic process at once, either provoking or reinforcing each other. Understanding the specific mechanisms by which environmental and individual factors impact an individual under RA risk in the preclinical stages can contribute to early disease diagnosis and, if the factor is modifiable, might be useful for the prevention or delay of its development.
Collapse
Affiliation(s)
- Marina Arleevskaya
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia;
- Correspondence: ; Tel.: +7-89172-886-679; Fax: +7-843-238-5413
| | - Elena Takha
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
| | - Sergey Petrov
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Gevorg Kazarian
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
| | - Yves Renaudineau
- Department of Immunology, CHU Toulouse, INSERM U1291, CNRS U5051, University Toulouse IIII, 31000 Toulouse, France;
| | - Wesley Brooks
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA;
| | - Regina Larionova
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
| | - Marina Korovina
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia;
| | - Anna Valeeva
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
| | - Eduard Shuralev
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Malik Mukminov
- Central Research Laboratory, Kazan State Medical Academy, 420012 Kazan, Russia; (E.T.); (S.P.); (G.K.); (R.L.); (M.K.); (A.V.); (E.S.); (M.M.)
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
| | - Olga Kravtsova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia;
| | - Andrey Novikov
- Mathematical Center, Sobolev Instiute of Mathematics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| |
Collapse
|
17
|
Bigham Z, Robles Y, Freund KM, Palmer JR, Bertrand KA. Hypertensive diseases of pregnancy and risk of breast cancer in the Black Women's Health Study. Breast Cancer Res Treat 2022; 194:127-135. [PMID: 35478297 DOI: 10.1007/s10549-022-06606-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/06/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Compared to white women, Black women have increased risk of developing hypertensive diseases of pregnancy (HDOP) and have a higher incidence of aggressive breast cancer subtypes. Few studies of HDOP and breast cancer risk have included large numbers of Black women. This study examined the relation of HDOP to incidence of breast cancer overall and by estrogen receptor (ER) status in Black women. METHODS We followed 42,982 parous women in the Black Women's Health Study, a nationwide prospective study of Black women. Multivariable Cox proportional hazards regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) to assess associations of self-reported HDOP, including preeclampsia and gestational hypertension, with breast cancer incidence overall and by ER subtype, adjusted for age and established breast cancer risk factors. RESULTS Over 20 years of follow-up, we identified 2376 incident breast cancer cases. History of HDOP (11.7%) was not associated with breast cancer risk overall (HR 0.98; 95% CI 0.87, 1.11). HRs for invasive ER+ and ER- breast cancer were 1.11 (95% CI 0.93, 1.34) and 0.81 (95% CI 0.61, 1.07), respectively. CONCLUSIONS HDOP was not associated with risk of overall breast cancer in Black women. A suggestive inverse association with ER- breast cancer may reflect an anti-tumorigenic hormone profile in HDOP, but those results require confirmation in other studies.
Collapse
Affiliation(s)
- Zahna Bigham
- Tufts University School of Medicine and Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Yvonne Robles
- Slone Epidemiology Center at Boston University, 72 East Concord Street, L-7, Boston, MA, USA
| | - Karen M Freund
- Tufts University School of Medicine and Graduate School of Biomedical Sciences, Boston, MA, USA.,Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Julie R Palmer
- Slone Epidemiology Center at Boston University, 72 East Concord Street, L-7, Boston, MA, USA
| | - Kimberly A Bertrand
- Slone Epidemiology Center at Boston University, 72 East Concord Street, L-7, Boston, MA, USA.
| |
Collapse
|
18
|
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
|
19
|
Bert S, Ward EJ, Nadkarni S. Neutrophils in pregnancy: New insights into innate and adaptive immune regulation. Immunology 2021; 164:665-676. [PMID: 34287859 PMCID: PMC8561097 DOI: 10.1111/imm.13392] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
The immunology of pregnancy has been the focus of many studies to better understand how the mother is able to tolerate the presence of a semi-allogeneic fetus. Far from the initial view of pregnancy as a state of immunosuppression, successful fetal development from implantation to birth is now known to be under the control of an intricate balance of immune cells. The balance between pro-inflammatory functions used to promote embryo implantation and placental development and immunosuppressive activity to maintain maternal tolerance of the fetus is an immunological phenotype unique to pregnancy, which is dependent on the time of gestation. Neutrophils are one of a host of innate immune cells detected at the maternal-fetal interface, but very little is known of their function. In this review, we explore the emerging functions of neutrophils during pregnancy and their interactions with and regulation of T cells, a key adaptive immune cell population essential for the establishment of fetal-maternal tolerance.
Collapse
Affiliation(s)
- Serena Bert
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| | - Eleanor J. Ward
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| | - Suchita Nadkarni
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| |
Collapse
|
20
|
Du Y, Wang B, Cai Z, Zhang H, Wang B, Liang W, Zhou G, Ouyang F, Wang W. The triclosan-induced shift from aerobic to anaerobic metabolism link to increased steroidogenesis in human ovarian granulosa cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 220:112389. [PMID: 34082246 DOI: 10.1016/j.ecoenv.2021.112389] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 06/12/2023]
Abstract
Triclosan (TCS) is an endocrine-disrupting chemical (EDC), which is used ubiquitously as an antimicrobial ingredient in healthcare products and causes contamination in the environment such as air, water, and biosolid-amended soil. Exposure to TCS may increase the risk of reproduction diseases and health issues. Several groups, including ours, have proved that TCS increased the biosynthesis of steroid hormones in different types of steroidogenic cells. However, the precise mechanism of toxic action of TCS on increased steroidogenesis at a molecular level remains to be elucidated. In this study, we try to address the mode of action that TCS affects energy metabolism with increased steroidogenesis. We evaluated the adverse effects of TCS on energy metabolism and steroidogenesis in human ovarian granulosa cells. The goal is to elucidate how increased steroidogenesis can occur with a shortage of adenosine triphosphate (ATP) whereas mitochondria-based energy metabolism is impaired. Our results demonstrated TCS increased estradiol and progesterone levels with upregulated steroidogenesis gene expression at concentrations ranging from 0 to 10 µM. Besides, glucose consumption, lactate level, and pyruvate kinase transcription were increased. Interestingly, the lactate level was attenuated with increased steroidogenesis, suggesting that pyruvate fate was shifted away from the formation of lactate towards steroidogenesis. Our study is gathering evidence suggesting a mode of action that TCS changes energy metabolism by predominating glucose flow towards the biosynthesis of steroid hormones. To the best of our knowledge, this is the first report that TCS presents such toxic action in disrupting hormone homeostasis.
Collapse
Affiliation(s)
- Yatao Du
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200292, China
| | - Bin Wang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200292, China
| | - Zhenzhen Cai
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200292, China
| | - Huihui Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bo Wang
- Department of Reproductive Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200292, China
| | - Wei Liang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200292, China
| | - Guangdi Zhou
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200292, China
| | - Fengxiu Ouyang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200292, China
| | - Weiye Wang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200292, China.
| |
Collapse
|
21
|
Hu X, Zhang L. Uteroplacental Circulation in Normal Pregnancy and Preeclampsia: Functional Adaptation and Maladaptation. Int J Mol Sci 2021; 22:8622. [PMID: 34445328 PMCID: PMC8395300 DOI: 10.3390/ijms22168622] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023] Open
Abstract
Uteroplacental blood flow increases as pregnancy advances. Adequate supply of nutrients and oxygen carried by uteroplacental blood flow is essential for the well-being of the mother and growth/development of the fetus. The uteroplacental hemodynamic change is accomplished primarily through uterine vascular adaptation, involving hormonal regulation of myogenic tone, vasoreactivity, release of vasoactive factors and others, in addition to the remodeling of spiral arteries. In preeclampsia, hormonal and angiogenic imbalance, proinflammatory cytokines and autoantibodies cause dysfunction of both endothelium and vascular smooth muscle cells of the uteroplacental vasculature. Consequently, the vascular dysfunction leads to increased vascular resistance and reduced blood flow in the uteroplacental circulation. In this article, the (mal)adaptation of uteroplacental vascular function in normal pregnancy and preeclampsia and underlying mechanisms are reviewed.
Collapse
Affiliation(s)
- Xiangqun Hu
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
22
|
Pereira MM, Mainigi M, Strauss JF. Secretory products of the corpus luteum and preeclampsia. Hum Reprod Update 2021; 27:651-672. [PMID: 33748839 PMCID: PMC8222764 DOI: 10.1093/humupd/dmab003] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite significant advances in our understanding of the pathophysiology of preeclampsia (PE), there are still many unknowns and controversies in the field. Women undergoing frozen-thawed embryo transfer (FET) to a hormonally prepared endometrium have been found to have an unexpected increased risk of PE compared to women who receive embryos in a natural FET cycle. The differences in risk have been hypothesized to be related to the absence or presence of a functioning corpus luteum (CL). OBJECTIVE AND RATIONALE To evaluate the literature on secretory products of the CL that could be essential for a healthy pregnancy and could reduce the risk of PE in the setting of FET. SEARCH METHODS For this review, pertinent studies were searched in PubMed/Medline (updated June 2020) using common keywords applied in the field of assisted reproductive technologies, CL physiology and preeclampsia. We also screened the complete list of references in recent publications in English (both animal and human studies) on the topics investigated. Given the design of this work as a narrative review, no formal criteria for study selection or appraisal were utilized. OUTCOMES The CL is a major source of multiple factors regulating reproduction. Progesterone, estradiol, relaxin and vasoactive and angiogenic substances produced by the CL have important roles in regulating its functional lifespan and are also secreted into the circulation to act remotely during early stages of pregnancy. Beyond the known actions of progesterone and estradiol on the uterus in early pregnancy, their metabolites have angiogenic properties that may optimize implantation and placentation. Serum levels of relaxin are almost undetectable in pregnant women without a CL, which precludes some maternal cardiovascular and renal adaptations to early pregnancy. We suggest that an imbalance in steroid hormones and their metabolites and polypeptides influencing early physiologic processes such as decidualization, implantation, angiogenesis and maternal haemodynamics could contribute to the increased PE risk among women undergoing programmed FET cycles. WIDER IMPLICATIONS A better understanding of the critical roles of the secretory products of the CL during early pregnancy holds the promise of improving the efficacy and safety of ART based on programmed FET cycles.
Collapse
Affiliation(s)
- María M Pereira
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Centre for Research on Reproduction and Women’s Health, University of Pennsylvania, Philadelphia, PA,19104 USA
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Centre for Research on Reproduction and Women’s Health, University of Pennsylvania, Philadelphia, PA,19104 USA
| |
Collapse
|
23
|
Shu C, Han S, Xu P, Wang Y, Cheng T, Hu C. Estrogen and Preeclampsia: Potential of Estrogens as Therapeutic Agents in Preeclampsia. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2543-2550. [PMID: 34163140 PMCID: PMC8214522 DOI: 10.2147/dddt.s304316] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/17/2021] [Indexed: 12/20/2022]
Abstract
There is a significant decline in the estrogen levels in preeclampsia, and exogenous administration of estradiol normalizes blood pressure and other associated symptoms of preeclampsia. The decrease in estrogen levels may be due to changes in enzyme activities of hydroxysteroid (17-β) dehydrogenase 1, aromatase, and COMT. There is also a decrease in the novel, estrogenic G-protein-coupled receptor 30 (GPR30) in the placental trophoblast cells in preeclampsia. The activation of GPR30 protects the placenta from hypoxia-reoxygenation injury, decreases apoptosis and increases proliferation through eNOS and PI3K-Akt signaling pathways. Estrogens may also increase Ca2+-activated K+ channel function, decrease the release of inflammatory cytokines, and oxidative stress to improve placental perfusion. Both preclinical and clinical studies show the decrease in the 2-methoxyestradiol levels in preeclampsia, which may be due to a decrease in estradiol itself along with a decrease in the enzymatic actions of the COMT enzyme. 2-Methoxyestradiol activates HIF1α and vascular endothelial growth factor receptors (VEGFR-2) to maintain placental perfusion by increasing angiogenesis. The present review discusses the preclinical and clinical studies describing the role of estrogen in preeclampsia along with possible mechanisms.
Collapse
Affiliation(s)
- Chang Shu
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130061, People's Republic of China
| | - Shumei Han
- Department of Medical Administration, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Peng Xu
- Department of Sports Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Ying Wang
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130061, People's Republic of China
| | - Tingting Cheng
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130061, People's Republic of China
| | - Cong Hu
- Reproductive Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, People's Republic of China
| |
Collapse
|
24
|
Hirschi KM, Tsai KYF, Davis T, Clark JC, Knowlton MN, Bikman BT, Reynolds PR, Arroyo JA. Growth arrest-specific protein-6/AXL signaling induces preeclampsia in rats†. Biol Reprod 2021; 102:199-210. [PMID: 31347670 DOI: 10.1093/biolre/ioz140] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 07/06/2019] [Accepted: 07/21/2019] [Indexed: 12/19/2022] Open
Abstract
Preeclampsia (PE) is a complicated obstetric complication characterized by increased blood pressure, decreased trophoblast invasion, and inflammation. The growth arrest-specific 6 (Gas6) protein is known to induce dynamic cellular responses and is elevated in PE. Gas6 binds to the AXL tyrosine kinase receptor and AXL-mediated signaling is implicated in proliferation and migration observed in several tissues. Our laboratory utilized Gas6 to induce preeclamptic-like conditions in pregnant rats. Our objective was to determine the role of Gas6/AXL signaling as a possible model of PE. Briefly, pregnant rats were divided into three groups that received daily intraperitoneal injections (from gestational day 7.5 to 17.5) of phosphate buffered saline (PBS), Gas6, or Gas6 + R428 (an AXL inhibitor administered from gestational day 13.5 to 17.5). Animals dispensed Gas6 experienced elevated blood pressure, increased proteinuria, augmented caspase-3-mediated placental apoptosis, and diminished trophoblast invasion. Gas6 also enhanced expression of several PE-related genes and a number of inflammatory mediators. Gas6 further enhanced placental oxidative stress and impaired mitochondrial respiration. Each of these PE-related characteristics was ameliorated in dams and/or their placentae when AXL inhibition by R428 occurred in tandem with Gas6 treatment. We conclude that Gas6 signaling is capable of inducing PE and that inhibition of AXL prevents disease progression in pregnant rats. These results provide insight into pathways associated with PE that could be useful in the clarification of potential therapeutic approaches.
Collapse
Affiliation(s)
- Kelsey M Hirschi
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Kary Y F Tsai
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Taylor Davis
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - J Christian Clark
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - M Nekel Knowlton
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Benjamin T Bikman
- Laboratory of Obesity and Metabolism, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Paul R Reynolds
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| | - Juan A Arroyo
- Lung and Placenta Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, USA
| |
Collapse
|
25
|
Gunaratne MDSK, Thorsteinsdottir B, Garovic VD. Combined Oral Contraceptive Pill-Induced Hypertension and Hypertensive Disorders of Pregnancy: Shared Mechanisms and Clinical Similarities. Curr Hypertens Rep 2021; 23:29. [PMID: 33982185 DOI: 10.1007/s11906-021-01147-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Oral contraceptive pill-induced hypertension (OCPIH) and hypertensive disorders in pregnancy (HDP) share common risk factors and pathophysiological mechanisms, yet the bidirectional relationship between these two conditions is not well-established. We review and describe OCPIH and HDP to better understand how hormonal and metabolic imbalances affect hypertension. RECENT FINDINGS Oral contraceptive pills continue to be a popular method of contraception, with an incidence of OCPIH ranging from 1-8.5% among OCP users. HDP have an incidence of 5-10% of all pregnancies in the USA and have been shown to be a powerful predictor of lifetime adverse cardiovascular outcomes, including future hypertension. OCPIH and HDP share common risk factors such as age, BMI, past personal and family history of hypertension, as well as pathogenic mechanisms, including alterations in hormonal metabolism and the renin angiotensin aldosterone system; imbalance of vasodilator-vasoconstrictor compounds; and changes in the cardiovascular system. Future research should address additional potential mechanisms that underlie hypertension in these two conditions where endocrine changes, either physiological (pregnancy) or iatrogenic (use of OCP), play a role. This may lead to novel, targeted treatment options to improve hypertension management and overall cardiovascular risk profile management in this subset of young female patients.
Collapse
Affiliation(s)
- Madugodaralalage D S K Gunaratne
- Division of Nephrology and Hypertension, Department of Internal Medicine, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Mayo Clinic, 200 First Street, SW, Rochester, MN, USA
| | - Bjorg Thorsteinsdottir
- Mayo Clinic KERN Center for the Science of Health Care Delivery and the Knowledge Evaluation and Research Unit, Division of Community Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Department of Internal Medicine, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Mayo Clinic, 200 First Street, SW, Rochester, MN, USA.
| |
Collapse
|
26
|
Cañumil VA, Bogetti E, de la Cruz Borthiry FL, Ribeiro ML, Beltrame JS. Steroid hormones and first trimester vascular remodeling. VITAMINS AND HORMONES 2021; 116:363-387. [PMID: 33752825 DOI: 10.1016/bs.vh.2021.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Successful implantation and placentation require neoangiogenesis and the remodeling of the uterine spiral arteries. Progesterone and estradiol control various of the placental functions, but their role in vascular remodeling remains controversial. Therefore, this chapter aims to summarize the current knowledge regarding the role of steroid hormones in the uteroplacental vascular remodeling during the first trimester of gestation.
Collapse
Affiliation(s)
- V A Cañumil
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina
| | - E Bogetti
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina
| | - F L de la Cruz Borthiry
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina
| | - M L Ribeiro
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina
| | - J S Beltrame
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
27
|
Hu XQ, Zhang L. Hypoxia and Mitochondrial Dysfunction in Pregnancy Complications. Antioxidants (Basel) 2021; 10:antiox10030405. [PMID: 33800426 PMCID: PMC7999178 DOI: 10.3390/antiox10030405] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia is a common and severe stress to an organism's homeostatic mechanisms, and hypoxia during gestation is associated with significantly increased incidence of maternal complications of preeclampsia, adversely impacting on the fetal development and subsequent risk for cardiovascular and metabolic disease. Human and animal studies have revealed a causative role of increased uterine vascular resistance and placental hypoxia in preeclampsia and fetal/intrauterine growth restriction (FGR/IUGR) associated with gestational hypoxia. Gestational hypoxia has a major effect on mitochondria of uteroplacental cells to overproduce reactive oxygen species (ROS), leading to oxidative stress. Excess mitochondrial ROS in turn cause uteroplacental dysfunction by damaging cellular macromolecules, which underlies the pathogenesis of preeclampsia and FGR. In this article, we review the current understanding of hypoxia-induced mitochondrial ROS and their role in placental dysfunction and the pathogenesis of pregnancy complications. In addition, therapeutic approaches selectively targeting mitochondrial ROS in the placental cells are discussed.
Collapse
|
28
|
Vishnyakova P, Poltavets A, Nikitina M, Midiber K, Mikhaleva L, Muminova K, Potapova A, Khodzhaeva Z, Pyregov A, Elchaninov A, Fatkhudinov T, Sukhikh G. Expression of Estrogen Receptor α by Decidual Macrophages in Preeclampsia. Biomedicines 2021; 9:biomedicines9020191. [PMID: 33672970 PMCID: PMC7917975 DOI: 10.3390/biomedicines9020191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia is a gestation-associated hypertensive syndrome that threatens the life and health of the mother and the child. The condition is presumably caused by systemic failure with a strong involvement of innate immunity. In particular, it has been associated with flexible phenotypes of macrophages, which depend on the molecules circulating in the blood and tissue fluid, such as cytokines and hormones. This study aimed at a comparative evaluation of pro-inflammatory (TNFα) and anti-inflammatory (CD206, MMP9, HGF) markers, as well as the levels of estrogen receptor α, expressed by decidual macrophages in normal pregnancy and in patients with early- and late-onset preeclampsia. The tissue samples of decidua basalis were examined by immunohistochemistry and Western blotting. Isolation of decidual macrophages and their characterization were performed using cultural methods, flow cytometry and real-time PCR. Over 50% of the isolated decidual macrophages were positive for the pan-macrophage marker CD68. In the early-onset preeclampsia group, the levels of estrogen receptor α in decidua were significantly decreased. Furthermore, significantly decreased levels of HGF and CD206 were observed in both preeclampsia groups compared with the control group. The observed downregulation of estrogen receptor α, HGF and CD206 may contribute to the balance of pro- and anti-inflammatory macrophages and thereby to pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Polina Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia; (A.P.); (K.M.); (A.P.); (Z.K.); (A.P.); (A.E.); (G.S.)
- Histology Department, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
- Correspondence: ; Tel.: +7-9150658577
| | - Anastasiya Poltavets
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia; (A.P.); (K.M.); (A.P.); (Z.K.); (A.P.); (A.E.); (G.S.)
| | - Maria Nikitina
- Scientific Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (K.M.); (L.M.)
| | - Konstantin Midiber
- Scientific Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (K.M.); (L.M.)
| | - Liudmila Mikhaleva
- Scientific Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (K.M.); (L.M.)
| | - Kamilla Muminova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia; (A.P.); (K.M.); (A.P.); (Z.K.); (A.P.); (A.E.); (G.S.)
| | - Alena Potapova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia; (A.P.); (K.M.); (A.P.); (Z.K.); (A.P.); (A.E.); (G.S.)
| | - Zulfiya Khodzhaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia; (A.P.); (K.M.); (A.P.); (Z.K.); (A.P.); (A.E.); (G.S.)
| | - Alexey Pyregov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia; (A.P.); (K.M.); (A.P.); (Z.K.); (A.P.); (A.E.); (G.S.)
| | - Andrey Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia; (A.P.); (K.M.); (A.P.); (Z.K.); (A.P.); (A.E.); (G.S.)
- Pirogov Russian National Research Medical University (RNRMU), 117997 Moscow, Russia
| | - Timur Fatkhudinov
- Histology Department, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
- Scientific Research Institute of Human Morphology, 117418 Moscow, Russia; (M.N.); (K.M.); (L.M.)
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia; (A.P.); (K.M.); (A.P.); (Z.K.); (A.P.); (A.E.); (G.S.)
| |
Collapse
|
29
|
Plant M, Armstrong C, Ruggiero A, Sherrill C, Uberseder B, Jeffries R, Nevarez J, Jorgensen MJ, Kavanagh K, Quinn MA. Advanced maternal age impacts physiologic adaptations to pregnancy in vervet monkeys. GeroScience 2020; 42:1649-1661. [PMID: 32588342 PMCID: PMC7732933 DOI: 10.1007/s11357-020-00219-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
The trend to delay pregnancy in the USA has resulted in the number of advanced maternal age (AMA) pregnancies to also increase. In humans, AMA is associated with a variety of pregnancy-related pathologies such as preeclampsia (PE). While AMA is known to be a factor which contributes to the development of pregnancy-induced diseases, the molecular and cellular mechanisms giving rise to this phenomenon are still very limited. This is due in part to lack of a preclinical model which has physiologic relevance to human pregnancy while also allowing control of environmental and genetic variability inherent in human studies. To determine potential physiologic relevance of the vervet/African green monkey (Chlorocebus aethiops sabaeus) as a preclinical model to study the effects of AMA on adaptations to pregnancy, thirteen age-diverse pregnant vervet monkeys (3-16 years old) were utilized to measure third trimester blood pressure (BP), complete blood count, iron measurements, and hormone levels. Significant associations were observed between third trimester diastolic BP and maternal age. Furthermore, the presence of leukocytosis with enhanced circulating neutrophils was observed in AMA mothers compared to younger mothers. Moreover, we observed a negative relationship between maternal age and estradiol, progesterone, and cortisol levels. Finally, offspring born to AMA mothers displayed a postnatal growth retardation phenotype. These studies demonstrate physiologic impairment in the adaptation to pregnancy in AMA vervet/African green monkeys. Our data indicate that the vervet/African green monkey may serve as a useful preclinical model and tool for deciphering pathological mediators of maternal disease in AMA pregnancy.
Collapse
Affiliation(s)
- Maren Plant
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Cecilia Armstrong
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Alistaire Ruggiero
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Chrissy Sherrill
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Beth Uberseder
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Rachel Jeffries
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Justin Nevarez
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Kylie Kavanagh
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Matthew A Quinn
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
30
|
Higher gestational weight gain and lower serum estradiol levels are associated with increased risk of preeclampsia after in vitro fertilization. Pregnancy Hypertens 2020; 22:126-131. [PMID: 32889248 DOI: 10.1016/j.preghy.2020.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 08/05/2020] [Accepted: 08/22/2020] [Indexed: 01/23/2023]
Abstract
PURPOSE To assess the association of preeclampsia with serum estradiol (E2) and progesterone (P4) levels on the day of human chorionic gonadotropin (hCG) administration during controlled ovarian hyperstimulation (COH) for in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI). METHODS This was a hospital-based cohort study using clinical data from the Kaohsiung Chang Gung Memorial Hospital Obstetric and Neonatal Database (KCGMHOND) from Jan 1, 2001 to December 1, 2018. RESULTS A total of 622 women who had live births after fresh IVF/ICSI-ET during the study period met our inclusion criteria. Twenty-eight women (4.5%) met the diagnostic criteria for preeclampsia. However, women in the preeclampsia group had a significantly higher body weight at delivery (80.5 vs. 70.0 kg, p < 0.001) and gestational weight gain (19.6 vs. 13.0 kg, p = 0.002) and had lower use of ICSI (10.7% vs. 29.9%, p = 0.021). We performed logistic regression analysis of the relationship of patient and treatment characteristics with preeclampsia. The crude ORs indicated that young female age ≤ 34, not using ICSI, E2 on hCG day < 1200 pg/mL and gestational weight gain > 20 kg were associated with preeclampsia. After adjustment for confounding, the only factors that remained significant were E2 on hCG day < 1200 pg/mL (aOR = 4.634, 95% CI = 1.061-20.222), and gestational weight gain > 20 kg (aOR: 13.601, 95% CI: 3.784, 48.880). CONCLUSIONS For women receiving IVF/ICSI, lower estradiol hormone levels on the day of hCG administration and higher pregnancy weight gain are related with subsequent preeclampsia.
Collapse
|
31
|
Wiciński M, Malinowski B, Puk O, Socha M, Słupski M. Methyldopa as an inductor of postpartum depression and maternal blues: A review. Biomed Pharmacother 2020; 127:110196. [DOI: 10.1016/j.biopha.2020.110196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/18/2022] Open
|
32
|
Zhao G, Li X, Miao H, Chen S, Hou Y. Estrogen Promotes cAMP Production in Mesenchymal Stem Cells by Regulating ADCY2. Int J Stem Cells 2020; 13:55-64. [PMID: 32114743 PMCID: PMC7119214 DOI: 10.15283/ijsc19139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/25/2020] [Accepted: 01/30/2020] [Indexed: 11/29/2022] Open
Abstract
Background and Objectives The maternal-fetal interface is an important source of mesenchymal stem cells (MSCs), and it is influenced by high levels of estradiol (E2) during pregnancy. It is highly important to study the role of E2 in MSCs for both clinical application and understanding of the mechanisms underlying pregnancy related diseases. Methods and Results In this study, differently expressed genes (DEGs) were found in the MSCs after exposure to E2. Then, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs was performed and the integrated regulatory network of DEGs-miRNA was constructed. A total of 390 DEGs were found in the MSCs exposed to E2, including 164 upregulated DEGs (e.g. ADCY2, VEGFA and PPY) and 226 downregulated DEGs (e.g. KNG1, AGT and NPY). Additionally, 10 miRNAs (such as miR-148A/B, miR-152, miR-182) identified the integrated regulatory network of DEGs-miRNAs. Among them, the expression of ADCY2 was significantly upregulated, and this was associated with multiple changed genes. We confirmed that the expression of ADCY2 is significantly promoted by E2 and subsequently promoted the production of cAMP in MSCs. We also found that E2 promoted ADCY2 expression by inhibiting miR-152 and miR-148a. Conclusions E2 promotes the expression of cAMP through miR-148a/152-ADCY2 in MSCs. It is suggested that E2 plays a key role in the growth and function of MSCs.
Collapse
Affiliation(s)
- Guangfeng Zhao
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xiujun Li
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Huishuang Miao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Shiwen Chen
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
33
|
Jeong JS, Lee DH, Lee JE, An SM, Yi PI, Lee GS, Hwang DY, Yang SY, Kim SC, An BS. The Expression and Contribution of SRCs with Preeclampsia Placenta. Reprod Sci 2020; 27:1513-1521. [PMID: 31997259 DOI: 10.1007/s43032-020-00142-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/26/2019] [Indexed: 11/28/2022]
Abstract
The steroid hormones act by binding to their receptors and subsequently interacting with coactivators. Several classes of coactivators have been identified and shown to be essential in estradiol (E2) responsiveness. The major coregulators are the p160 steroid receptor coactivator (SRC) family. Although the function of SRCs in other organs has been well studied, it has not been thoroughly studied in the placenta. In addition, the correlation between preeclampsia (PE) and SRCs has not been examined previously. Therefore, we compared the expression patterns of SRCs in normal and PE placentas. In human PE placental tissues, SRC-1 mRNA, and protein levels were downregulated in the PE group. In addition, to assess the expression of SRCs in a PE environment, we used Reduced Uterine Perfusion Pressure (RUPP) model and placental cells were cultured in hypoxia condition. SRC-1 proteins were reduced in the placenta of PE-like rat RUPP model. Furthermore, SRCs proteins were significantly downregulated in hypoxia-grown placental cells. To examine the interaction between estrogen receptors (ERs) and SRC-1 protein, we performed co-immunoprecipitation. The interaction of SRC-1 with ERα was significantly stronger than that with ERβ. In PE placenta, the interaction of both ERα and ERβ with SRC-1 was stronger than that in normal placenta. In summary, our results demonstrate that expression levels of SRC-1, not SRC-2 and SRC-3, were decreased in hypoxia-induced PE placenta, which may further reduce the signaling of sex steroid hormones such as E2. The dysregulated signaling of E2 by SRC-1 expression could be associated with the PE placental symptoms of patients.
Collapse
Affiliation(s)
- Jea Sic Jeong
- Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, 50 Cheonghak-ri, Samrangjin-eup, Miryang, Gyeongsangnam-do, 627-706, South Korea
| | - Dong Hyung Lee
- Department of Obstetrics and Gynecology, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University College of Medicine, Yangsan, Gyeongnam, 50612, South Korea
| | - Jae-Eon Lee
- Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, 50 Cheonghak-ri, Samrangjin-eup, Miryang, Gyeongsangnam-do, 627-706, South Korea
| | - Sung-Min An
- Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, 50 Cheonghak-ri, Samrangjin-eup, Miryang, Gyeongsangnam-do, 627-706, South Korea
| | - Pyong In Yi
- Department of Bioenvironmental Energy, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Busan, South Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, South Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, 50 Cheonghak-ri, Samrangjin-eup, Miryang, Gyeongsangnam-do, 627-706, South Korea
| | - Seung Yun Yang
- Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, 50 Cheonghak-ri, Samrangjin-eup, Miryang, Gyeongsangnam-do, 627-706, South Korea
| | - Seung Chul Kim
- Department of Obstetrics and Gynecology, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University College of Medicine, Yangsan, Gyeongnam, 50612, South Korea.
| | - Beum-Soo An
- Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, 50 Cheonghak-ri, Samrangjin-eup, Miryang, Gyeongsangnam-do, 627-706, South Korea.
| |
Collapse
|
34
|
Carlson NS, Frediani JK, Corwin EJ, Dunlop A, Jones D. Metabolomic Pathways Predicting Labor Dystocia by Maternal Body Mass Index. AJP Rep 2020; 10:e68-e77. [PMID: 32140295 PMCID: PMC7056397 DOI: 10.1055/s-0040-1702928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 12/20/2019] [Indexed: 11/25/2022] Open
Abstract
Objectives The purpose of this study was to evaluate the metabolic pathways activated in the serum of African-American women during late pregnancy that predicted term labor dystocia. Study Design Matched case-control study ( n = 97; 48 cases of term labor dystocia and 49 normal labor progression controls) with selection based on body mass index (BMI) at hospital admission and maternal age. Late pregnancy serum samples were analyzed using ultra-high-resolution metabolomics. Differentially expressed metabolic features and pathways between cases experiencing term labor dystocia and normal labor controls were evaluated in the total sample, among women who were obese at the time of labor (BMI ≥ 30 kg/m2), and among women who were not obese. Results Labor dystocia was predicted by different metabolic pathways in late pregnancy serum among obese (androgen/estrogen biosynthesis) versus nonobese African-American women (fatty acid activation, steroid hormone biosynthesis, bile acid biosynthesis, glycosphingolipid metabolism). After adjusting for maternal BMI and age in the total sample, labor dystocia was predicted by tryptophan metabolic pathways in addition to C21 steroid hormone, glycosphingolipid, and androgen/estrogen metabolism. Conclusion Metabolic pathways consistent with lipotoxicity, steroid hormone production, and tryptophan metabolism in late pregnancy serum were significantly associated with term labor dystocia in African-American women.
Collapse
Affiliation(s)
- Nicole S. Carlson
- Emory University Nell Hodgson Woodruff School of Nursing, Atlanta, Georgia
| | | | - Elizabeth J. Corwin
- Department of Physiology, Columbia University School of Nursing, New York, New York
| | - Anne Dunlop
- Departments of Family and Preventive Medicine, Epidemiology, and Nursing, Emory University, Atlanta, Georgia
| | - Dean Jones
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
35
|
Kale K, Vishwekar P, Balsarkar G, Jassawalla MJ, Sawant G, Madan T. Differential levels of surfactant protein A, surfactant protein D, and progesterone to estradiol ratio in maternal serum before and after the onset of severe early‐onset preeclampsia. Am J Reprod Immunol 2019; 83:e13208. [DOI: 10.1111/aji.13208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/23/2019] [Accepted: 11/01/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Kavita Kale
- Department of Innate Immunity ICMR‐National Institute for Research in Reproductive Health Mumbai India
| | - Pallavi Vishwekar
- Department of Obstetrics and Gynaecology Dr. DY Patil Medical College, Hospital and Research Centre Navi Mumbai Mumbai India
| | - Geetha Balsarkar
- Department of Obstetrics and Gynaecology Nowrosjee Wadia Maternity Hospital Mumbai India
| | | | - Ganpat Sawant
- Department of Obstetrics and Gynaecology Dr. DY Patil Medical College, Hospital and Research Centre Navi Mumbai Mumbai India
| | - Taruna Madan
- Department of Innate Immunity ICMR‐National Institute for Research in Reproductive Health Mumbai India
| |
Collapse
|
36
|
Zheng J, Xu J, Zhang Y, Zhou N. Effects of insulin combined with metformin on serum cystatin C, homocysteine and maternal and neonatal outcomes in pregnant women with gestational diabetes mellitus. Exp Ther Med 2019; 19:467-472. [PMID: 31853319 PMCID: PMC6909788 DOI: 10.3892/etm.2019.8224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/02/2019] [Indexed: 12/30/2022] Open
Abstract
Effects of insulin combined with metformin on serum cystatin C (Cys C), homocysteine (Hcy) and maternal and neonatal outcomes in pregnant women with gestational diabetes mellitus (GDM) were investigated. In total, 80 cases of pregnant women diagnosed with GDM in the Department of Obstetrics and Gynecology of Liaocheng Third People's Hospital from July 2015 to July 2017 were selected and divided into a study group (42 cases) and a control group (38 cases). The study group was treated with insulin combined with metformin, and the control group was treated with insulin. Fasting blood glucose (FBG) and postprandial blood glucose after 2 h (2hPG) of the two groups were compared before and after treatment. Levels of serum Cys C, Hcy, urinary protein (UmAlb), postpartum maternal outcomes and adverse reactions during pregnancy were compared in the two groups before and after treatment. After treatment, the level of FBG and 2hPG in the control group was higher than that in the treatment group (P<0.05). After treatment, the level of serum Cys C and Hcy in both groups were lower than that before the treatment, and the level in the study group was lower than that in the control group (P<0.05). The total incidence of neonatal adverse outcomes and the number of adverse pregnancies in GDM patients in the study group were significantly lower than those in the control group (P<0.05). There were no significant differences in adverse reactions during pregnancy between the two groups (P>0.05). In conclusion, insulin combined with metformin is more effective than insulin alone in reducing serum Cys C and Hcy levels, with significant effect on the improvement of maternal and neonatal outcomes.
Collapse
Affiliation(s)
- Jizeng Zheng
- Department of Obstetrics and Gynecology, Liaocheng Third People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Juan Xu
- Department of Obstetrics, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Yin Zhang
- Department of Obstetrics and Gynecology, Liaocheng Third People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Nan Zhou
- Department of Health Care, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
37
|
Chasalow F, John CM, Bochner R. Spiral steroids as potential markers for pre-eclampsia: A pilot study. Steroids 2019; 151:108466. [PMID: 31351941 DOI: 10.1016/j.steroids.2019.108466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/14/2019] [Accepted: 07/19/2019] [Indexed: 11/18/2022]
Abstract
Pre-eclampsia is a condition that complicates 3-6% of pregnancies. At present, there is no FDA approved diagnostic method to evaluate risk or progression. We would like to report our observation of elevated levels of the spiral steroid phosphoester precursor in patients with pre-eclampsia. Samples from 20 normotensive pregnant women and from 20 women with pre-eclampsia were purchased from Global Alliance for the Prevention of Prematurity and Stillbirth (GAPPS). After addition of miltefosine (hexadecyl phosphocholine) as an internal control, each sample was extracted and evaluated by tandem MS. The method detected 2 spiral steroid phosphoesters and their common precursor. For each mass ion, the ion counts obtained were compared to the ion counts of miltefosine. The samples from the normotensive women were used to establish the mean and standard deviation. Then, Z-scores were determined for each of the serum components. Of the samples from the women with pre-eclampsia, 12 of 19 (63%) samples had Z-scores over 2.0 for at least one of the steroid phosphoesters. In contrast, current markers under development for risk of pre-eclampsia have prediction scores ranging from 8 to 33%. As the spiral steroids are lactones, similar to spironolactone and related compounds, they could function as endogenous potassium sparing hormones. However, as only about half of the affected patients had elevated levels of the spiral steroids, it may not be the only underlying pathology. Parturition would end transfer of placental spiral steroids and account for the termination of the symptoms of pre-eclampsia/eclampsia. Previous investigators have proposed inadequate placental function as the critical pathology of pre-eclampsia, but it is hard to imagine how inadequate placental function leads to the maternal pathology without invoking an endogenous potassium sparing hormone originating in the placenta.
Collapse
Affiliation(s)
- Fred Chasalow
- IOMA LLC, Belmont, CA, United States; Department of Laboratory Medicine, VAMC, San Francisco, CA, United States.
| | - Constance M John
- Center for Immunochemistry, VAMC, San Francisco, CA, United States
| | - Ron Bochner
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
38
|
Abstract
Pregnancy is associated with physiological adjustments in order to allow adequate growth and fetal development. In particular, steroids are necessary to maintain in balance numerous functions during gestation. Steroidogenesis in the maternal, placental and fetal compartments and the biological effects of progestins and estrogens that play a pivotal role before and during pregnancy are described. Although it is well-known that androgens are considered as substrate for estrogens biosynthesis, their biosynthesis and functionality in placental and other tissues have been questioned. As compared with healthy pregnancy, steroid hormones levels have been found altered in complicated pregnancies and hormonal treatments have been used is some pathologies. Therefore, the aim of this work was to review the biosynthesis, function and regulation of progestins, androgens and estrogens during gestation. Furthermore, steroid hormones concentrations during healthy and complicated pregnancy as well hormonal therapies for the prevention of miscarriages and preterm deliveries are discussed in the present review.
Collapse
Affiliation(s)
- Nancy Noyola-Martínez
- a Departamento de Biología de la Reproducción , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , D.F. México , México
| | - Ali Halhali
- a Departamento de Biología de la Reproducción , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , D.F. México , México
| | - David Barrera
- a Departamento de Biología de la Reproducción , Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , D.F. México , México
| |
Collapse
|
39
|
Hahn S, Hasler P, Vokalova L, van Breda SV, Lapaire O, Than NG, Hoesli I, Rossi SW. The role of neutrophil activation in determining the outcome of pregnancy and modulation by hormones and/or cytokines. Clin Exp Immunol 2019; 198:24-36. [PMID: 30768780 DOI: 10.1111/cei.13278] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2018] [Indexed: 12/16/2022] Open
Abstract
Neutrophils are often exclusively considered as a first-line innate immune defence, able to rapidly kill or trap pathogens and causing in case of over-activation tissue damage. In the female reproductive tract, however, the presence and activity of neutrophils seems to be tightly regulated. Major players in orchestrating this regulation are cyclical steroid sex hormones present during the menstrual cycle and pregnancy. This review describes the role of sex hormones in regulating directly or indirectly the functionality of neutrophils, the role of neutrophils during fertilization and pregnancy and in controlling viral, fungal and bacterial infection. This review also discusses the consequence of overt neutrophil activation in pregnancy pathologies.
Collapse
Affiliation(s)
- S Hahn
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - P Hasler
- Department of Rheumatology, Kantonsspital Aarau, Aarau, Switzerland
| | - L Vokalova
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - S V van Breda
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland.,Department of Rheumatology, Kantonsspital Aarau, Aarau, Switzerland
| | - O Lapaire
- Department of Obstetrics, University Women's Hospital Basel, Basel, Switzerland
| | - N G Than
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - I Hoesli
- Department of Obstetrics, University Women's Hospital Basel, Basel, Switzerland
| | - S W Rossi
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
40
|
Narvaez-Sanchez R, Calderón JC, Vega G, Trillos MC, Ospina S. Skeletal muscle as a protagonist in the pregnancy metabolic syndrome. Med Hypotheses 2019; 126:26-37. [PMID: 31010495 DOI: 10.1016/j.mehy.2019.02.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/12/2019] [Accepted: 02/27/2019] [Indexed: 12/13/2022]
Abstract
The pregnant woman normally shows clinical manifestations similar to a metabolic syndrome (MS), due to her metabolic and hemodynamic adaptations in order to share nutrients with the child. If those adjustments are surpassed, a kind of pregnancy MS (PregMS) could appear, characterized by excessive insulin resistance and vascular maladaptation. Skeletal muscle (SKM) must be a protagonist in the PregMS: SKM strength and mass have been associated inversely with MS incidence in non-pregnant patients, and in pregnant women muscular activity modulates metabolic and vascular adaptations that favor better outcomes. Of note, a sedentary lifestyle affects exactly in the other way. Those effects may be explained not only by the old paradigm of SKM being a great energy consumer and store, but because it is an endocrine organ whose chronic activity or deconditioning correspondingly releases myokines modulating insulin sensitivity and cardiovascular adaptation, by direct or indirect mechanisms not well understood. In this document, we present evidence to support the concept of a PregMS and hypothesize on the role of the SKM mass, fiber types composition and myokines in its pathophysiology. Also, we discuss some exercise interventions in pregnancy as a way to test our hypotheses.
Collapse
Affiliation(s)
- Raul Narvaez-Sanchez
- Physiology and Biochemistry Research Group PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia; Red iberoamericana de trastornos vasculares y del embarazo, RIVATREM, Colombia.
| | - Juan C Calderón
- Physiology and Biochemistry Research Group PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia. http://www.udea.edu.co/physis
| | - Gloria Vega
- Physiology and Biochemistry Research Group PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia. http://www.udea.edu.co/physis
| | - Maria Camila Trillos
- Physiology and Biochemistry Research Group PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia. http://www.udea.edu.co/physis
| | - Sara Ospina
- Physiology and Biochemistry Research Group PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia. http://www.udea.edu.co/physis
| |
Collapse
|
41
|
Effect of Oxidative Stress on the Estrogen-NOS-NO-K Ca Channel Pathway in Uteroplacental Dysfunction: Its Implication in Pregnancy Complications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9194269. [PMID: 30881600 PMCID: PMC6387699 DOI: 10.1155/2019/9194269] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/19/2018] [Accepted: 01/14/2019] [Indexed: 12/27/2022]
Abstract
During pregnancy, the adaptive changes in uterine circulation and the formation of the placenta are essential for the growth of the fetus and the well-being of the mother. The steroid hormone estrogen plays a pivotal role in this adaptive process. An insufficient blood supply to the placenta due to uteroplacental dysfunction has been associated with pregnancy complications including preeclampsia and intrauterine fetal growth restriction (IUGR). Oxidative stress is caused by an imbalance between free radical formation and antioxidant defense. Pregnancy itself presents a mild oxidative stress, which is exaggerated in pregnancy complications. Increasing evidence indicates that oxidative stress plays an important role in the maladaptation of uteroplacental circulation partly by impairing estrogen signaling pathways. This review is aimed at providing both an overview of our current understanding of regulation of the estrogen-NOS-NO-KCa pathway by reactive oxygen species (ROS) in uteroplacental tissues and a link between oxidative stress and uteroplacental dysfunction in pregnancy complications. A better understanding of the mechanisms will facilitate the development of novel and effective therapeutic interventions.
Collapse
|
42
|
Placental Expression of NEMO Protein in Normal Pregnancy and Preeclampsia. DISEASE MARKERS 2019; 2019:8418379. [PMID: 30723530 PMCID: PMC6339720 DOI: 10.1155/2019/8418379] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/24/2018] [Accepted: 11/11/2018] [Indexed: 12/13/2022]
Abstract
Background Preeclamptic pregnancies often present an intensified inflammatory state associated with the nuclear activity of NFκB. NEMO is an essential regulator of nuclear factor kappa B (NFκB) in cytoplasmic and nuclear cellular compartments. The aim of the present study is to examine the level and localization of the NEMO protein in preeclamptic and nonpreeclamptic placentas. Methods The study includes 97 preeclamptic cases and 88 controls. NEMO distribution was analyzed immunohistochemically. Its localization in the nuclear and cytoplasmic fractions, as well as in total homogenates of placental samples, was studied by western blot and ELISA. Results The western blot and ELISA results indicate a significant difference in NEMO concentration in the total and nuclear fractions between preeclamptic and control samples (p < 0.01 and p < 0.001, respectively). In the cytoplasmic complement, similar levels of NEMO were found in preeclamptic and control placentas. In addition, immunohistochemical staining revealed that the NEMO protein is mainly localized in the syncytiotrophoblast layer, with controls demonstrating a stronger reaction with NEMO antibodies. This study also shows that the placental level of NEMO depends on the sex of the fetus. Conclusions The depletion of the NEMO protein in the cellular compartments of placental samples may activate one of the molecular pathways influencing the development of preeclampsia, especially in pregnancies with a female fetus. A reduction of the NEMO protein in the nuclear fraction of preeclamptic placentas may intensify the inflammatory state characteristic for preeclampsia and increase the level of apoptosis and necrosis within preeclamptic placentas.
Collapse
|
43
|
Anelli GM, Mandò C, Letizia T, Mazzocco MI, Novielli C, Lisso F, Personeni C, Vago T, Cetin I. Placental ESRRG-CYP19A1 Expressions and Circulating 17-Beta Estradiol in IUGR Pregnancies. Front Pediatr 2019; 7:154. [PMID: 31069202 PMCID: PMC6491753 DOI: 10.3389/fped.2019.00154] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/01/2019] [Indexed: 12/04/2022] Open
Abstract
Introduction: Sex steroids are regulating factors for intrauterine growth. 17-β Estradiol (E2) is particularly critical to a physiological pregnancy, as increased maternal E2 was correlated to lower fetal weight at delivery. The placenta itself is a primary source of estrogens, synthetized from cholesterol precursors. Cytochrome P450 aromatase (encoded by CYP19A1 gene) is a rate-limiting enzyme for E2 biosynthesis. CYP19A1 transcription is supported by Estrogen Related-Receptor Gamma (ERRγ- ESRRG gene), which thus has an indirect role in placental steroidogenesis. Here we investigated maternal E2 levels and placental CYP19A1 and ESRRG expressions in pregnancies with IntraUterine Growth Restriction (IUGR). Methods: Singleton pregnancies were studied. E2 was measured in maternal plasma by electrochemiluminescence in 16 term controls and 11 IUGR (classified by umbilical artery doppler pulsatility index) at elective cesarean section, and also in 13 controls during pregnancy at a gestational age comparable to IUGR. CYP19A1 and ESRRG expressions were analyzed in placental tissue. Maternal/fetal characteristics, placental and molecular data were compared among study groups and tested for correlations. Results: Maternal E2 plasma concentrations were significantly decreased in IUGR compared to controls at delivery. When analyzing normal pregnancies at a gestational age similar to IUGR, E2 levels were not different to pathological cases. However, E2 levels at delivery positively correlated with placental efficiency. Placental CYP19A1 levels were significantly higher in IUGR placental tissue vs. controls, and specifically increased in female IUGR placentas. ESRRG expression was not different among groups. Discussion: We report a positive correlation between 17-β Estradiol levels and placental efficiency, that might indicate a disrupted steroidogenesis in IUGR pregnancies. Moreover, we show alterations of CYP19A1 expression in IUGR placentas, possibly indicating a compensatory effect to the adverse IUGR intrauterine environment, also depending on fetal sex. Further studies are needed to deeper investigate IUGR alterations in the complex interaction among molecules involved in placental steroidogenesis.
Collapse
Affiliation(s)
- Gaia Maria Anelli
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Chiara Mandò
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Teresa Letizia
- Endocrinology Laboratory, Sacco University Hospital, Milan, Italy
| | - Martina Ilaria Mazzocco
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Chiara Novielli
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Fabrizia Lisso
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Carlo Personeni
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Tarcisio Vago
- Endocrinology Laboratory, Sacco University Hospital, Milan, Italy
| | - Irene Cetin
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy.,Unit of Obstetrics and Gynecology, Buzzi University Hospital, Milan, Italy
| |
Collapse
|
44
|
Kumar S, Gordon GH, Abbott DH, Mishra JS. Androgens in maternal vascular and placental function: implications for preeclampsia pathogenesis. Reproduction 2018; 156:R155-R167. [PMID: 30325182 PMCID: PMC6198264 DOI: 10.1530/rep-18-0278] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adequate maternal vascular adaptations and blood supply to the uterus and placenta are crucial for optimal oxygen and nutrient transport to growing fetuses of eutherian mammals, including humans. Multiple factors contribute to hemodynamics and structuring of placental vasculature essential for term pregnancy with minimal complications. In women, failure to achieve or sustain favorable pregnancy progression is, not surprisingly, associated with high incidence of antenatal complications, including preeclampsia, a hypertensive disorder of pregnancy. While the pathogenesis of preeclampsia in women remains unknown, a role for androgens is emerging. The relationship between androgens and maternal cardiovascular and placental function deserves particular consideration because testosterone levels in the circulation of preeclamptic women are elevated approximately two- to three-fold and are positively correlated with vascular dysfunction. Preeclampsia is also associated with elevated placental androgen receptor (AR) gene expression. Studies in animal models mimicking the pattern and level of increase of adult female testosterone levels to those found in preeclamptic pregnancies, replicate key features of preeclampsia, including gestational hypertension, endothelial dysfunction, exaggerated vasoconstriction to angiotensin II, reduced spiral artery remodeling, placental hypoxia, decreased nutrient transport and fetal growth restriction. Taken together, these data strongly implicate AR-mediated testosterone action as an important pathway contributing to clinical manifestation of preeclampsia. This review critically addresses this hypothesis, taking into consideration both clinical and preclinical data.
Collapse
Affiliation(s)
- Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA.,Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.,Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Geoffrey H Gordon
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - David H Abbott
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.,Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, Wisconsin, USA.,Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Jay S Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
45
|
Chatuphonprasert W, Jarukamjorn K, Ellinger I. Physiology and Pathophysiology of Steroid Biosynthesis, Transport and Metabolism in the Human Placenta. Front Pharmacol 2018; 9:1027. [PMID: 30258364 PMCID: PMC6144938 DOI: 10.3389/fphar.2018.01027] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/24/2018] [Indexed: 12/11/2022] Open
Abstract
The steroid hormones progestagens, estrogens, androgens, and glucocorticoids as well as their precursor cholesterol are required for successful establishment and maintenance of pregnancy and proper development of the fetus. The human placenta forms at the interface of maternal and fetal circulation. It participates in biosynthesis and metabolism of steroids as well as their regulated exchange between maternal and fetal compartment. This review outlines the mechanisms of human placental handling of steroid compounds. Cholesterol is transported from mother to offspring involving lipoprotein receptors such as low-density lipoprotein receptor (LDLR) and scavenger receptor class B type I (SRB1) as well as ATP-binding cassette (ABC)-transporters, ABCA1 and ABCG1. Additionally, cholesterol is also a precursor for placental progesterone and estrogen synthesis. Hormone synthesis is predominantly performed by members of the cytochrome P-450 (CYP) enzyme family including CYP11A1 or CYP19A1 and hydroxysteroid dehydrogenases (HSDs) such as 3β-HSD and 17β-HSD. Placental estrogen synthesis requires delivery of sulfate-conjugated precursor molecules from fetal and maternal serum. Placental uptake of these precursors is mediated by members of the solute carrier (SLC) family including sodium-dependent organic anion transporter (SOAT), organic anion transporter 4 (OAT4), and organic anion transporting polypeptide 2B1 (OATP2B1). Maternal-fetal glucocorticoid transport has to be tightly regulated in order to ensure healthy fetal growth and development. For that purpose, the placenta expresses the enzymes 11β-HSD 1 and 2 as well as the transporter ABCB1. This article also summarizes the impact of diverse compounds and diseases on the expression level and activity of the involved transporters, receptors, and metabolizing enzymes and concludes that the regulatory mechanisms changing the physiological to a pathophysiological state are barely explored. The structure and the cellular composition of the human placental barrier are introduced. While steroid production, metabolism and transport in the placental syncytiotrophoblast have been explored for decades, few information is available for the role of placental-fetal endothelial cells in these processes. With regard to placental structure and function, significant differences exist between species. To further decipher physiologic pathways and their pathologic alterations in placental steroid handling, proper model systems are mandatory.
Collapse
Affiliation(s)
- Waranya Chatuphonprasert
- Pathophysiology of the Placenta, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Faculty of Medicine, Mahasarakham University, Maha Sarakham, Thailand
| | - Kanokwan Jarukamjorn
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Isabella Ellinger
- Pathophysiology of the Placenta, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
46
|
Li J, Ding Z, Yang Y, Mao B, Wang Y, Xu X. Lycium barbarum polysaccharides protect human trophoblast HTR8/SVneo cells from hydrogen peroxide‑induced oxidative stress and apoptosis. Mol Med Rep 2018; 18:2581-2588. [PMID: 30015960 PMCID: PMC6102627 DOI: 10.3892/mmr.2018.9274] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/13/2018] [Indexed: 02/03/2023] Open
Abstract
Pregnancy complications are associated with abnormal cytotrophoblast differentiation and invasion. Hydrogen peroxide (H2O2) is an important mediator of oxidative ischemia/reperfusion stress in the placenta. Lycium barbarum polysaccharides (LBP) have been demonstrated to counteract oxidative free radicals. The effects of LBP in trophoblast HTR8/SVneo cells injured with H2O2 were examined. A cell counting kit-8 assay was performed to detect the effect of LBP at different concentrations on the proliferative ability of H2O2 injured trophoblast cells. Flow cytometry was used to determine the levels of reactive oxygen species (ROS), mitochondria membrane potential (MMP) disruption and apoptosis. Superoxide dismutase (SOD) activity and lactate dehydrogenase (LDH) leakage into the supernatant was detected by ultraviolet spectrophotometry. Reverse transcription-quantitative polymerase chain reaction and western blot analysis were performed to detect the expression of apoptosis-associated factors, including survivin, hypoxia inducible factor 1-α (HIF1-α), Bcl-2 apoptosis regulator (Bcl-2), Bcl-2 associated X apoptosis regulator (Bax). The results revealed that LBP protected the proliferative ability of trophoblast cells injured with H2O2 in a dose-dependent manner. LBP inhibited the oxidative stress induced by H2O2, by reducing ROS and LDH levels and increasing SOD activity. Additionally, LBP decreased MMP disruption and cell apoptosis induced by H2O2, by increasing the mRNA and protein expression of survivin, HIF1-α and Bcl-2 and decreasing Bax expression. Therefore, it was concluded that LBP protected human trophoblast cells from H2O2-induced oxidative stress and cell apoptosis via regulation of apoptosis-associated factor expression. It will provide a novel strategy for the treatment of pregnancy complications.
Collapse
Affiliation(s)
- Jing Li
- Department of Women and Children's Medical Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, Gansu 730050, P.R. China
| | - Zhongjun Ding
- Reproduction Medicine Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, Gansu 730050, P.R. China
| | - Yue Yang
- Discipline of Physiology, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Baohong Mao
- Department of Women and Children's Medical Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, Gansu 730050, P.R. China
| | - Yanxia Wang
- Department of Women and Children's Medical Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, Gansu 730050, P.R. China
| | - Xiaoying Xu
- Perinatal Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, Gansu 730050, P.R. China
| |
Collapse
|
47
|
Vermillion MS, Nelson A, Vom Steeg L, Loube J, Mitzner W, Klein SL. Pregnancy preserves pulmonary function following influenza virus infection in C57BL/6 mice. Am J Physiol Lung Cell Mol Physiol 2018; 315:L517-L525. [PMID: 29847990 DOI: 10.1152/ajplung.00066.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Pregnancy is associated with significant anatomic and functional changes to the cardiopulmonary system. Using pregnant C57BL/6 mice, we characterized changes in pulmonary structure and function during pregnancy in healthy animals and following infection with influenza A virus (IAV). We hypothesized that pregnancy-associated alterations in pulmonary physiology would contribute to the more severe outcome of IAV infection. Nonpregnant and pregnant females (at embryonic day 10.5) were either mock-infected or infected with 2009 H1N1 IAV for assessment of pulmonary function, structure, and inflammation at 8 days postinoculation. There were baseline differences in pulmonary function, with pregnant females having greater lung compliance, total lung capacity, and fixed lung volume than nonpregnant females. Following IAV infection, both pregnant and nonpregnant females exhibited reduced circulating progesterone, which in nonpregnant females was associated with increased pulmonary resistance and decreased lung compliance, minute ventilation, and oxygen diffusing capacity compared with uninfected nonpregnant females. In pregnant females, reduced concentrations of progesterone were associated with adverse pregnancy outcomes, but measures of pulmonary function were preserved following IAV infection and were not significantly different from uninfected pregnant mice. Following IAV infection, infectious virus titers and total numbers of pulmonary leukocytes were similar between pregnant and nonpregnant females, but the histological density of pulmonary inflammation was reduced in pregnant animals. These data suggest that pregnancy in mice is associated with significant alterations in pulmonary physiology but that these changes served to preserve lung function during IAV infection. Pregnancy-associated alterations in pulmonary physiology may serve to protect females during severe influenza.
Collapse
Affiliation(s)
- Meghan S Vermillion
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland.,Department of Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine , Baltimore, Maryland
| | - Andrew Nelson
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Landon Vom Steeg
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Jeffery Loube
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland.,Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| |
Collapse
|