1
|
Lockington C, Favetta LA. How Per- and Poly-Fluoroalkyl Substances Affect Gamete Viability and Fertilization Capability: Insights from the Literature. J Xenobiot 2024; 14:651-678. [PMID: 38804291 PMCID: PMC11130945 DOI: 10.3390/jox14020038] [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: 03/31/2024] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
There has been emerging research linking per- and poly-fluoroalkyl substances (PFAS) to gamete viability and fertility. PFAS, prevalent in the environment and water supplies, undergo slow degradation due to their C-F bond and a long half-life (2.3-8.5 years). In females, PFAS inhibit the hypothalamic-pituitary-gonadal (HPG) axis, reducing follicle-stimulating hormone (FSH) and luteinizing hormone (LH) levels, leading to the inhibition of androgen and estradiol production. PFAS have been found to cause detrimental effects on egg quality through impairing folliculogenesis. In males, PFAS can impair sperm motility and morphology: two fundamental qualities of successful fertilization. PFAS exposure has been proven to inhibit testosterone production, sperm capacitation, and acrosomal reaction. After fertilization, the results of PFAS exposure to embryos have also been investigated, showing reduced development to the blastocyst stage. The aim of this review is to report the main findings in the literature on the impact of PFAS exposure to gamete competency and fertilization capability by highlighting key studies on both male and female fertility. We report that there is significant evidence demonstrating the negative impacts on fertility after PFAS exposure. At high doses, these environmentally abundant and widespread compounds can significantly affect human fertility.
Collapse
Affiliation(s)
| | - Laura A. Favetta
- Reproductive Health and Biotechnology Lab, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| |
Collapse
|
2
|
Rawan AF, Langar H, Munetomo M, Yamamoto Y, Kawano K, Kimura K. Effects of insulin-like growth factor-1 on the mRNA expression of estradiol receptors, steroidogenic enzymes, and steroid production in bovine follicles. J Reprod Dev 2023; 69:337-346. [PMID: 37940556 PMCID: PMC10721850 DOI: 10.1262/jrd.2023-047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/05/2023] [Indexed: 11/10/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) plays a crucial role in follicular growth and stimulates steroid hormone production in bovine follicles. Steroid hormones are synthesized through the actions of steroidogenic enzymes, specifically STAR, CYP11A1, HSD3B, and CYP19A1 in both theca cells (TCs) and granulosa cells (GCs), under the influence of gonadotropins. Particularly, estradiol 17β (E2) assumes a central role in follicular development and selection by activating estrogen receptors β (ESR2) in GCs. We assessed ESR2 mRNA expression in GCs of developing follicles and investigated the impact of IGF-1 on the mRNA expression of ESR2, CYP19A1, FSHR, and LHCGR, STAR, CYP11A1, and HSD17B in cultured GCs and TCs, respectively. Additionally, we assessed the influence of IGF-1 on androstenedione (A4), progesterone (P4), and testosterone (T) production in TCs. Small-sized follicles (< 6 mm) exhibited the highest levels of ESR2 mRNA expression, whereas medium-sized follicles (7-8 mm) displayed higher levels than large-sized follicles (≥ 9 mm) (P < 0.05). IGF-1 increased the mRNA expression of ESR2, CYP19A1, and FSHR in GCs of follicles of both sizes, except for FSHR mRNA in medium-sized follicles (P < 0.05). IGF-1 significantly elevated mRNA expression of LHCGR, STAR, CYP11A1, and CYP17B in TCs of small- and medium-sized follicles (P < 0.05). Moreover, IGF-1 augmented the production of A4 and P4 but had no impact on T production in TCs of small- and medium-sized follicles. Taken together, our findings indicate that IGF-1 upregulates steroidogenic enzymes and steroid hormone production, underscoring the crucial role of IGF-1 in follicle development and selection.
Collapse
Affiliation(s)
- Ahmad Farid Rawan
- Laboratory of Reproductive Physiology, Faculty of Environmental, Life, Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
- Pre-Clinic Department, Veterinary Science Faculty, Nangarhar University, 2603, Afghanistan
| | - Hikmatullah Langar
- Laboratory of Reproductive Physiology, Faculty of Environmental, Life, Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Maho Munetomo
- Laboratory of Reproductive Physiology, Faculty of Environmental, Life, Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Yuki Yamamoto
- Laboratory of Reproductive Physiology, Faculty of Environmental, Life, Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
- Laboratory of Veterinary Physiology, Tokyo University of Agriculture and Technology, Tokyo 183-0054, Japan
| | - Kohei Kawano
- Laboratory of Reproductive Physiology, Faculty of Environmental, Life, Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Koji Kimura
- Laboratory of Reproductive Physiology, Faculty of Environmental, Life, Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
3
|
Bharati J, Kumar S, Kumar S, Mohan NH, Islam R, Pegu SR, Banik S, Das BC, Borah S, Sarkar M. Androgen receptor gene deficiency results in the reduction of steroidogenic potential in porcine luteal cells. Anim Biotechnol 2023; 34:2183-2196. [PMID: 35678291 DOI: 10.1080/10495398.2022.2079517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Luteal steroidogenesis is critical to implantation and pregnancy maintenance in mammals. The role of androgen receptors (AR) in the progesterone (P4) producing luteal cells of porcine corpus luteum (CL) remains unexplored. The aim of the present study was to establish AR gene knock out (KO) porcine luteal cell culture system model by CRISPR/Cas9 genome editing technology and to study the downstream effects of AR gene deficiency on steroidogenic potential and viability of luteal cells. For this purpose, genomic cleavage detection assay, microscopy, RT-qPCR, ELISA, annexin, MTT, and viability assay complemented by bioinformatics analysis were employed. There was significant downregulation (p < 0.05) in the relative mRNA expression of steroidogenic marker genes STAR, CYP11A1, HSD3B1 in AR KO luteal cells as compared to the control group, which was further validated by the significant (p < 0.05) decrease in the P4 production. Significant decrease (p < 0.05) in relative viability on third passage were also observed. The relative mRNA expression of hypoxia related gene HIF1A was significantly (p < 0.05) downregulated in AR KO luteal cells. Protein-protein interaction analysis mapped AR to signaling pathways associated with luteal cell functionality. These findings suggests that AR gene functionality is critical to luteal cell steroidogenesis in porcine.
Collapse
Affiliation(s)
- Jaya Bharati
- Animal Physiology, ICAR-National Research Centre on Pig, Guwahati, India
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Satish Kumar
- Animal Genetics and Breeding, ICAR-National Research Centre on Pig, Guwahati, India
| | - Sunil Kumar
- Animal Reproduction, ICAR-National Research Centre on Pig, Guwahati, India
| | - N H Mohan
- Animal Physiology, ICAR-National Research Centre on Pig, Guwahati, India
| | - Rafiqul Islam
- Animal Reproduction, ICAR-National Research Centre on Pig, Guwahati, India
| | - Seema Rani Pegu
- Animal Health, ICAR-National Research Centre on Pig, Guwahati, India
| | - Santanu Banik
- Animal Genetics and Breeding, ICAR-National Research Centre on Pig, Guwahati, India
| | - Bikash Chandra Das
- Animal Physiology, ICAR-National Research Centre on Pig, Guwahati, India
| | - Sanjib Borah
- Lakhimpur College of Veterinary Science, Assam Agricultural University, North Lakhimpur, India
| | - Mihir Sarkar
- Director, ICAR-National Research Centre on Yak, Dirang, India
| |
Collapse
|
4
|
de Jesus Nascimento AE, Santos LC, Santos BR, Santos EO, Cunha MCDSG, Snoeck PPDN, de Lavor MSL, Silva JF. Spatial and temporal expression profile of sex steroid receptors and antioxidant enzymes in the maternal-fetal interface of domestic cats. Theriogenology 2023; 210:234-243. [PMID: 37542738 DOI: 10.1016/j.theriogenology.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Sex steroids and antioxidant enzymes modulate uterine and placental physiology. Failures in the expression, signaling, and/or secretion of these mediators are associated with female infertility and gestational problems. However, there is no data on the expression profile of receptors for sex steroids and antioxidant enzymes in the maternal-fetal interface of domestic cats. Uterus and placenta samples from non-pregnant diestrus cats and cats in mid- and late pregnancy were used to analyze the protein and gene expression of the receptors for estrogen alpha (ERα), progesterone (PR), and androgen (AR) and the antioxidant enzymes superoxide dismutase 1 (SOD1), catalase, and glutathione peroxidase 1 (GPX1) by immunohistochemistry and qPCR. Higher uterine expression of ERα, Pr, and Sod1 was observed in the pregnant cats, especially in mid-pregnancy, compared to non-pregnant diestrus cats, as well as reduced endometrial catalase immunostaining. In the placenta, the mRNA expression of Erα, Pr, Ar, and Gpx1 was higher in late pregnancy in relation to mid-pregnancy. Moreover, weak or no placental expression was observed for catalase in mid- and late pregnancy, while strong immunostaining was observed for AR in trophoblasts and decidual cells in mid-pregnancy. The findings of this study demonstrated that pregnancy in female cats increases the uterine expression of sex steroid receptors and antioxidant enzymes, and that the placental expression of these mediators varies according to gestational age.
Collapse
Affiliation(s)
- Acácia Eduarda de Jesus Nascimento
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Luciano Cardoso Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Bianca Reis Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Emilly Oliveira Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Maria Clara da Silva Galrão Cunha
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Paola Pereira das Neves Snoeck
- Hospital Veterinário, Departamento de Ciências Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Mário Sérgio Lima de Lavor
- Hospital Veterinário, Departamento de Ciências Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil.
| |
Collapse
|
5
|
Falahati-Pour SK, Pourmasumi S, Mirhashemi ES. Ovarian Endocrine Status and ART 0utcomes in Women within PCOS Based on Different Testosterone Levels. Indian J Endocrinol Metab 2023; 27:440-444. [PMID: 38107733 PMCID: PMC10723604 DOI: 10.4103/ijem.ijem_360_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/22/2023] [Accepted: 05/05/2023] [Indexed: 12/19/2023] Open
Abstract
Background It is estimated that in women at reproductive age, the risk of polycystic ovary syndrome (PCOS) is about 5-21%. In PCOS cases with ovulation dysfunction, assisted reproductive techniques (ART) are useful for infertility treatment. Objective This study aimed to evaluate the ART outcome in infertile PCOS women based on different testosterone levels. Finally, the relationships between testosterone in different levels and reproductive parameters including endocrine status, the response of ovaries, and pregnancy outcomes were assessed. Methods In this retrospective study, 352 infertile PCOS women were examined. The women were categorised into five groups according to their testosterone levels: A = T < 0.4, B = 0.4 < T > 0.6, C = 0.6 < T > 0.8, D = 0.8 < T > 1.0 and E = T > 1.0 ng/dL. All study cases were in similar hyper-stimulation protocol and finally, hormonal profile and ART outcomes were compared between testosterone levels. P value ≤ 0.05 was statistically significant. Results In testosterone levels >1.0, the levels of anti-mullerian hormone (AMH) and luteinising hormone (LH) were higher than in other testosterone level groups. AMH (P = 0.05) and LH (P = 0.001) levels showed significant differences. No correlation was present between testosterone levels and ART outcomes, including stimulation duration, endometrial thickness, oocyte numbers, numbers of matured oocytes, number of obtained embryos, fertilisation rate, implantation rate clinical pregnancy and abortion rate. Conclusions Serum testosterone levels did not show any correlation with pregnancy outcomes in ART cycles of PCOS. However, basal testosterone levels are a good predictor for ovarian reserve and ovarian response. Consequently, we suggest that some prospective studies must be designed to approve the role of testosterone in the prediction of the outcome of pregnancy in ART cycles.
Collapse
Affiliation(s)
| | - Soheila Pourmasumi
- Social Determinants of Health Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Clinical Research Development Unit (CRDU), Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Elham Sadat Mirhashemi
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
6
|
Bar-Sadeh B, Pnueli L, Keestra S, Bentley GR, Melamed P. Srd5a1 is Differentially Regulated and Methylated During Prepubertal Development in the Ovary and Hypothalamus. J Endocr Soc 2023; 7:bvad108. [PMID: 37646011 PMCID: PMC10461783 DOI: 10.1210/jendso/bvad108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Indexed: 09/01/2023] Open
Abstract
5α-reductase-1 catalyzes production of various steroids, including neurosteroids. We reported previously that expression of its encoding gene, Srd5a1, drops in murine ovaries and hypothalamic preoptic area (POA) after early-life immune stress, seemingly contributing to delayed puberty and ovarian follicle depletion, and in the ovaries the first intron was more methylated at two CpGs. Here, we hypothesized that this CpG-containing locus comprises a methylation-sensitive transcriptional enhancer for Srd5a1. We found that ovarian Srd5a1 mRNA increased 8-fold and methylation of the same two CpGs decreased up to 75% between postnatal days 10 and 30. Estradiol (E2) levels rise during this prepubertal stage, and exposure of ovarian cells to E2 increased Srd5a1 expression. Chromatin immunoprecipitation in an ovarian cell line confirmed ESR1 binding to this differentially methylated genomic region and enrichment of the enhancer modification, H3K4me1. Targeting dCas9-DNMT3 to this locus increased CpG2 methylation 2.5-fold and abolished the Srd5a1 response to E2. In the POA, Srd5a1 mRNA levels decreased 70% between postnatal days 7 and 10 and then remained constant without correlation to CpG methylation levels. Srd5a1 mRNA levels did not respond to E2 in hypothalamic GT1-7 cells, even after dCas9-TET1 reduced CpG1 methylation by 50%. The neonatal drop in POA Srd5a1 expression occurs at a time of increasing glucocorticoids, and treatment of GT1-7 cells with dexamethasone reduced Srd5a1 mRNA levels; chromatin immunoprecipitation confirmed glucocorticoid receptor binding at the enhancer. Our findings on the tissue-specific regulation of Srd5a1 and its methylation-sensitive control by E2 in the ovaries illuminate epigenetic mechanisms underlying reproductive phenotypic variation that impact life-long health.
Collapse
Affiliation(s)
- Ben Bar-Sadeh
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Lilach Pnueli
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Sarai Keestra
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
- Department of Anthropology, Durham University, Durham, DH1 3LE, UK
| | | | - Philippa Melamed
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
7
|
Yusuf ANM, Amri MF, Ugusman A, Hamid AA, Wahab NA, Mokhtar MH. Hyperandrogenism and Its Possible Effects on Endometrial Receptivity: A Review. Int J Mol Sci 2023; 24:12026. [PMID: 37569402 PMCID: PMC10419014 DOI: 10.3390/ijms241512026] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Endometrial receptivity is a state of the endometrium defined by its readiness for embryo implantation. When the receptivity of the endometrium is impaired due to hyperandrogenism or androgen excess, this condition can lead to pregnancy loss or infertility. Hyperandrogenism encompasses a wide range of clinical manifestations, including polycystic ovary syndrome (PCOS), idiopathic hirsutism, hirsutism and hyperandrogaenemia, non-classical congenital adrenal hyperplasia, hyperandrogenism, insulin resistance, acanthosis nigricans (HAIR-AN), ovarian or adrenal androgen-secreting neoplasms, Cushing's syndrome, and hyperprolactinaemia. Recurrent miscarriages have been shown to be closely related to elevated testosterone levels, which alter the endometrial milieu so that it is less favourable for embryo implantation. There are mechanisms for endometrial receptivity that are affected by excess androgen. The HOXA gene, aVβ3 integrin, CDK signalling pathway, MECA-79, and MAGEA-11 were the genes and proteins affect endometrial receptivity in the presence of a hyperandrogenic state. In this review, we would like to explore the other manifestations of androgen excess focusing on causes other than PCOS and learn possible mechanisms of endometrial receptivity behind androgen excess leading to pregnancy loss or infertility.
Collapse
Affiliation(s)
- Allia Najmie Muhammad Yusuf
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Mohd Fariz Amri
- Department of Pathology, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Adila A. Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Norhazlina Abdul Wahab
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
8
|
Associations of per- and polyfluoroalkyl substances and alternatives with reproductive hormones in women of childbearing age. Int J Hyg Environ Health 2023; 250:114158. [PMID: 36934548 DOI: 10.1016/j.ijheh.2023.114158] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/09/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND Experimental studies suggested that per- and polyfluoroalkyl substances (PFAS) may have endocrine-disrupting effects. However, the epidemiological evidence on the associations of PFAS with female reproductive hormones is sparse and limited to perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid (PFOA). OBJECTIVE To evaluate effects of legacy and emerging PFAS alternatives on female reproductive hormones. METHODS A total of 433 reproductive-aged females were recruited from 2014 to 2016. Information on age, age at menarche, gravity, menstrual cycle, BMI, education, and income was obtained from medical records and questionnaires. Serum samples were collected for reproductive hormones, and plasma samples for PFAS measurement by ultraperformance liquid chromatography - tandem mass spectrometer (UPLC-MS/MS). Multiple linear regression and quantile g-computation (q-gcomp) were used to examine the associations of individual PFAS and their mixture with reproductive hormones. RESULTS Multiple linear regression analysis showed significant effects of certain PFAS on total testosterone (TT): adjusted estimate (β) for perfluoroheptanoic acid (PFHpA) was 0.57 (95% CI: 0.18, 0.97). Moreover, a positive association was detected between PFAS mixture and TT in the q-gcomp model. Higher concentrations of 6:2 chlorinated polyfluorinated ether sulfonic acid (6:2 Cl-PFESA) were associated with significantly lower prolactin level (β = -0.07, 95% CI: -0.14, -0.001). CONCLUSION Our study found that exposure to PFAS alternatives was associated with altered levels of reproductive hormones in women of childbearing age.
Collapse
|
9
|
The Influence of Metabolic Factors and Diet on Fertility. Nutrients 2023; 15:nu15051180. [PMID: 36904180 PMCID: PMC10005661 DOI: 10.3390/nu15051180] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Infertility is a disease globally affecting 20-30% of the reproductive age female population. However, in up to 50% on recorded cases, problems with infertility are ascribed to men; therefore, it is important to popularize healthy eating also in this group. During the last decade, it has been observed that society's lifestyle changed drastically: reduced energy expenditure in physical activity per day, increased consumption of hypercaloric and high-glycemic-index foods with high content of trans fats, and reduced consumption of dietary fiber, which negatively affects fertility. Increasing evidence points to a link between diet and fertility. It is becoming clear that well-planned nutrition can also contribute to the effectiveness of ART. The low-GI plant-based diet appears to have a positive effect, especially when it is based on Mediterranean dietary patterns: rich in antioxidants, vegetable protein, fiber, MUFA fatty acids, omega-3, vitamins, and minerals. Importantly, this diet has been shown to protect against chronic diseases associated with oxidative stress, which also translates into pregnancy success. As lifestyle and nutrition seem to be important factors affecting fertility, it is worth expanding knowledge in this regard among couples trying to conceive a child.
Collapse
|
10
|
Kartal B, Alimoğulları E, Çaylı S. The immunoexpression of valosin-containing protein and small VCP-interacting protein in rat ovaries. Anat Histol Embryol 2023. [PMID: 36843060 DOI: 10.1111/ahe.12914] [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: 03/11/2022] [Revised: 07/07/2022] [Accepted: 02/13/2023] [Indexed: 02/28/2023]
Abstract
Numerous cellular processes are controlled by the ubiquitin-proteasome-mediated degradation pathway, involve the 97-kDa valosin-containing protein (p97/VCP). Small p97/VCP-interacting protein (SVIP) was first discovered as one of the novel androgen-responsive genes as well as one of the many cofactors controlling p97/VCP. The aim of the study was to investigate localization and immunoexpression of p97/VCP and SVIP in rat ovarian tissue. The histomorphological examination of rat ovarian tissue was performed by using haematoxylin-eosin (HE) staining. Using the immunohistochemical technique, cellular location and expression of p97/VCP and SVIP in rat ovarian tissue were examined. The nuclear and cytoplasmic immunoexpression of p97/VCP and SVIP was observed in the different stages of ovarian follicles and corpus luteum in the rat ovaries. The immunolocalization of SVIP and VCP in the rat ovaries suggest that they may be involved in the oogenesis. Further studies should be performed about the function of the VCP and SVIP in the female reproductive tract.
Collapse
Affiliation(s)
- Bahar Kartal
- Department of Histology and Embryology, Ankara Yıldırım Beyazıt University, Medical Faculty, Ankara, Turkey
| | - Ebru Alimoğulları
- Department of Histology and Embryology, Ankara Yıldırım Beyazıt University, Medical Faculty, Ankara, Turkey
| | - Sevil Çaylı
- Department of Histology and Embryology, Ankara Yıldırım Beyazıt University, Medical Faculty, Ankara, Turkey
| |
Collapse
|
11
|
Young AS, Herkert N, Stapleton HM, Coull BA, Hauser R, Zoeller T, Behnisch PA, Felzel E, Brouwer A, Allen JG. Hormone receptor activities of complex mixtures of known and suspect chemicals in personal silicone wristband samplers worn in office buildings. CHEMOSPHERE 2023; 315:137705. [PMID: 36592838 PMCID: PMC9937064 DOI: 10.1016/j.chemosphere.2022.137705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023]
Abstract
Humans are exposed to increasingly complex mixtures of hormone-disrupting chemicals from a variety of sources, yet, traditional research methods only evaluate a small number of chemicals at a time. We aimed to advance novel methods to investigate exposures to complex chemical mixtures. Silicone wristbands were worn by 243 office workers in the USA, UK, China, and India during four work shifts. We analyzed extracts of the wristbands for: 1) 99 known (targeted) chemicals; 2) 1000+ unknown chemical features, tentatively identified through suspect screening; and 3) total hormonal activities towards estrogen (ER), androgen (AR), and thyroid hormone (TR) receptors in human cell assays. We evaluated associations of chemicals with hormonal activities using Bayesian kernel machine regression models, separately for targeted versus suspect chemicals (with detection ≥50%). Every wristband exhibited hormonal activity towards at least one receptor: 99% antagonized TR, 96% antagonized AR, and 58% agonized ER. Compared to men, women were exposed to mixtures that were more estrogenic (180% higher, adjusted for country, age, and skin oil abundance in wristband), anti-androgenic (110% higher), and complex (median 836 detected chemical features versus 780). Adjusted models showed strong associations of jointly increasing chemical concentrations with higher hormonal activities. Several targeted and suspect chemicals were important co-drivers of overall mixture effects, including chemicals used as plasticizers, fragrance, sunscreen, pesticides, and from other or unknown sources. This study highlights the role of personal care products and building microenvironments in hormone-disrupting exposures, and the substantial contribution of chemicals not often identifiable or well-understood to those exposures.
Collapse
Affiliation(s)
- Anna S Young
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA.
| | - Nicholas Herkert
- Nicholas School of the Environment, Duke University, 9 Circuit Dr, Durham, NC 27710, USA
| | - Heather M Stapleton
- Nicholas School of the Environment, Duke University, 9 Circuit Dr, Durham, NC 27710, USA
| | - Brent A Coull
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Russ Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Thomas Zoeller
- Department of Biology, University of Massachusetts Amherst, Morrill Science Center, Amherst 01003, USA
| | - Peter A Behnisch
- BioDetection Systems, Science Park 406, 1098 XH Amsterdam, Netherlands
| | - Emiel Felzel
- BioDetection Systems, Science Park 406, 1098 XH Amsterdam, Netherlands
| | - Abraham Brouwer
- BioDetection Systems, Science Park 406, 1098 XH Amsterdam, Netherlands
| | - Joseph G Allen
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| |
Collapse
|
12
|
Higher live birth rate following transdermal testosterone pretreatment in poor responders: a systematic review and meta-analysis. Reprod Biomed Online 2023; 46:81-91. [PMID: 36369150 DOI: 10.1016/j.rbmo.2022.09.022] [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: 07/03/2022] [Revised: 09/08/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
Abstract
A systematic review and meta-analysis was performed aiming to identify good-quality randomized controlled trials (RCT) evaluating testosterone pretreatment in poor responders. Eight RCTs were analysed, evaluating 797 women. Transdermal testosterone gel was used in all studies, with a dose ranging from 10 to 12.5 mg/day for 10-56 days. The main outcome measure was achievement of pregnancy, expressed as clinical pregnancy or live birth. Testosterone pretreatment was associated with a significantly higher live birth (risk ratio [RR] 2.07, 95% confidence interval [CI] 1.09-3.92) and clinical pregnancy rate (RR 2.25, 95% CI 1.54-3.30), as well as a significant increase in the number of cumulus-oocyte complexes retrieved. Significantly fewer days to complete ovarian stimulation, a lower total dose of gonadotrophins, a lower cancellation rate due to poor ovarian response and a thicker endometrium on the day of triggering of final oocyte maturation were observed. No significant differences were observed in oestradiol concentration, the numbers of follicles ≥17 mm, metaphase II oocytes, two-pronuclear oocytes and embryos transferred, and the proportion of patients with embryo transfer. The current study suggests that the probability of pregnancy is increased in poor responders pretreated with transdermal testosterone who are undergoing ovarian stimulation for IVF.
Collapse
|
13
|
Devillers MM, François CM, Chester M, Corre R, Cluzet V, Giton F, Cohen-Tannoudji J, Guigon CJ. Androgen receptor signaling regulates follicular growth and steroidogenesis in interaction with gonadotropins in the ovary during mini-puberty in mice. Front Endocrinol (Lausanne) 2023; 14:1130681. [PMID: 37152943 PMCID: PMC10154677 DOI: 10.3389/fendo.2023.1130681] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
In females, androgens contribute to ovarian diseases such as polycystic ovarian syndrome (PCOS), but their action is also crucial for ovarian physiology, i.e., follicular growth and estradiol (E2) synthesis during reproductive life, in interaction with the gonadotropins LH and FSH. However, it is unclear whether androgens already play a role in the ovary at mini-puberty, a phase of postnatal development with active follicular growth and high E2 levels. Therefore, we analyzed the potential actions of androgens on the ovary and their possible interaction with gonadotropins during this period in mice. We used molecular-based studies and pharmacological approaches in vivo and on cultured ovaries. We found that mini-pubertal ovaries produce significant amounts of testosterone and display androgen receptor (AR) expression in growing follicles, both under the control of LH. By blocking AR signaling either in vivo or in ovarian cultures, we found that this pathway may participate in the regulation of prepubertal E2 synthesis and follicular growth, possibly by regulating the expression of a number of key intra-ovarian regulators, including FSH receptor (Fshr), the aromatase enzyme converting androgens into estrogens (Cyp19a1) and the cell cycle inhibitor p27KIP1 (Cdkn1b). We further showed that AR may stimulate FSH-mediated regulation of Cyp19a1 through its action on Fshr mRNA abundance. Overall, this work supports the idea that AR signaling is already activated in mini-pubertal ovaries to regulate E2 synthesis and follicular growth, at the interplay with LH and FSH signaling. Its early action may, thus, contribute to the implementation of early ovarian function with possible impacts on reproductive function.
Collapse
Affiliation(s)
- Marie M. Devillers
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Charlotte M. François
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Mélanie Chester
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Raphaël Corre
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Victoria Cluzet
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Frank Giton
- AP-HP, Pôle biologie-Pathologie Henri Mondor, Inserm IMRB U955, Créteil, France
| | - Joëlle Cohen-Tannoudji
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Céline J. Guigon
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
- *Correspondence: Céline J. Guigon,
| |
Collapse
|
14
|
Khamoshina MB, Artemenko YS, Bayramova AA, Ryabova VA, Orazov MR. Polycystic ovary syndrome and obesity: a modern paradigm. RUDN JOURNAL OF MEDICINE 2022. [DOI: 10.22363/2313-0245-2022-26-4-382-395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Polycystic ovary syndrome is a heterogeneous endocrine disease that affects women of childbearing age. The pathogenesis of polycystic ovary syndrome has not been fully studied to date, its paradigm considers the genetic determinism of the manifestation of hormonal and metabolic disorders, which are considered to be criteria for the verification of the disease (hyperandrogenism, oligo/anovulation and/or polycystic ovarian transformation during ultrasound examination (ultrasound). This review discusses the main ways of interaction between hyperandrogenism, insulin resistance and obesity and their role in the pathogenesis of polycystic ovary syndrome, as well as possible methods of treatment for this category of patients. The review analyzes the role of hyperandrogenism and insulin resistance in the implementation of the genetic scenario of polycystic ovary syndrome and finds out the reasons why women with polycystic ovary syndrome often demonstrate the presence of a «metabolic trio» - hyperinsulinemia, insulin resistance and type 2 diabetes mellitus. It is noted that obesity is not included in the criteria for the diagnosis of polycystic ovary syndrome, but epidemiological data confirm the existence of a relationship between these diseases. Obesity, especially visceral, which is often found in women with polycystic ovary syndrome, enhances and worsens metabolic and reproductive outcomes with polycystic ovary syndrome, as well as increases insulin resistance and compensatory hyperinsulinemia, which, in turn, stimulates adipogenesis and suppresses lipolysis. Obesity increases the sensitivity of tech cells to luteinizing hormone stimulation and enhances functional hyperandrogenism of the ovaries, increasing the production of androgens by the ovaries. Excess body weight is associated with a large number of inflammatory adipokines, which, in turn, contribute to the growth of insulin resistance and adipogenesis. Obesity and insulin resistance exacerbate the symptoms of hyperandrogenism, forming a vicious circle that contributes to the development of polycystic ovary syndrome. These data allow us to conclude that bariatric surgery can become an alternative to drugs (metformin, thiazolidinedione analogs of glucagon-like peptide-1), which has shown positive results in the treatment of patients with polycystic ovary syndrome and obesity.
Collapse
|
15
|
Xu S, Liu Y, Xue K, Liu X, Jia G, Zeng Y, Chen Y. Diagnostic value of total testosterone and free androgen index measured by LC-MS/MS for PCOS and insulin resistance. J Clin Lab Anal 2022; 36:e24739. [PMID: 36258308 DOI: 10.1002/jcla.24739] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The objective of the study was to explore the clinical significance of steroid hormones in the diagnosis of PCOS and PCOS-related insulin resistance through liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) and chemiluminescent immunoassay (CLIA). METHODS The study included 114 patients with PCOS and 100 controls. Steroid hormone levels in serum were measured using LC-MS/MS and CLIA. The Bland-Altman method was used to check the consistency between the two methods. The diagnostic value of the LC-MS/MS method for female hyperandrogenemia and PCOS was evaluated. RESULTS Women with PCOS were younger than controls on average (p < 0.001). PCOS patients had higher luteal hormone (LH, p < 0.001), insulin (p = 0.002), estradiol (E2, p < 0.001), total testosterone (TT, p < 0.001), free androgen index (FAI, p = 0.021), dehydroepiandrosterone sulfate (DHEA, p = 0.021), insulin resistance index (HOMA-IR) (p = 0.034), and fasting glucose (p = 0.017) levels than controls as measured by CLIA. The diagnostic value of TT was the best, and the area under the AUC curve was 0.766. Women with PCOS had higher androstenedione (A2, p < 0.001), FAI (p < 0.001), TT (p < 0.001), and 17-hydroxyprogesterone (17-OHP, p < 0.001) levels than controls as measured by LC-MS/MS. The ROC curve showed that the diagnostic efficacy of A2, TT, and 17-OHP was 0.830, 0.851, and 0.714, respectively. The consistency of TT detected by LC-MS/MS and CLIA was poor according to the Bland-Altman method. Detected TT by LC-MS/MS had the highest diagnostic efficiency for PCOS. The diagnostic power of the LC-MS/MS results for PCOS-related insulin resistance was analyzed. The results showed that the FAI had the highest diagnostic power, with an ROC curve of 0.798. CONCLUSION LC-MS/MS is more sensitive and accurate than CLIA in the determination of serum TT and FAI. TT is more effective for the diagnosis of PCOS, whereas FAI is more valuable in the diagnosis of insulin resistance.
Collapse
Affiliation(s)
- Sujuan Xu
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Yun Liu
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Kai Xue
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Xiaoguang Liu
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Genmei Jia
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Yu Zeng
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Yajun Chen
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| |
Collapse
|
16
|
Walters KA, Moreno-Asso A, Stepto NK, Pankhurst MW, Rodriguez Paris V, Rodgers RJ. Key signalling pathways underlying the aetiology of polycystic ovary syndrome. J Endocrinol 2022; 255:R1-R26. [PMID: 35980384 DOI: 10.1530/joe-22-0059] [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: 06/09/2022] [Accepted: 07/11/2022] [Indexed: 11/08/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine condition characterised by a range of reproductive, endocrine, metabolic and psychological abnormalities. Reports estimate that around 10% of women of reproductive age are affected by PCOS, representing a significant prevalence worldwide, which poses a high economic health burden. As the origin of PCOS remains largely unknown, there is neither a cure nor mechanism-based treatments leaving patient management suboptimal and focused solely on symptomatic treatment. However, if the underlying mechanisms underpinning the development of PCOS were uncovered then this would pave the way for the development of new interventions for PCOS. Recently, there have been significant advances in our understanding of the underlying pathways likely involved in PCOS pathogenesis. Key insights include the potential involvement of androgens, insulin, anti-Müllerian hormone and transforming growth factor beta in the development of PCOS. This review will summarise the significant scientific discoveries on these factors that have enhanced our knowledge of the mechanisms involved in the development of PCOS and discuss the impact these insights may have in shaping the future development of effective strategies for women with PCOS.
Collapse
Affiliation(s)
- Kirsty A Walters
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Alba Moreno-Asso
- Institute for Health and Sport, Victoria University, Footscray, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Victoria University, St. Albans, Victoria, Australia
| | - Nigel K Stepto
- Institute for Health and Sport, Victoria University, Footscray, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Victoria University, St. Albans, Victoria, Australia
- Monash Centre for Health Research and Implementation, Monash University and Monash Health, Clayton, Victoria, Australia
- Medicine at Western Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael W Pankhurst
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Valentina Rodriguez Paris
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Raymond J Rodgers
- The Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
17
|
Association between testosterone levels and bone mineral density in females aged 40-60 years from NHANES 2011-2016. Sci Rep 2022; 12:16426. [PMID: 36180560 PMCID: PMC9525583 DOI: 10.1038/s41598-022-21008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/21/2022] [Indexed: 11/22/2022] Open
Abstract
Growing evidence indicates that testosterone is a conspicuous marker for assessing male bone mineral density (BMD). However, research regarding testosterone levels and BMD is sparse and controversial for females. Hence, we aimed to investigate the association between testosterone levels and BMD among adult females aged 40–60 years in the United States. In this cross-sectional study, all participants were part of the National Health and Nutrition Examination Survey (2011–2016). A weighted general linear model was used to estimate the association between testosterone levels and lumbar BMD. Age, race, income level, education level, body mass index (BMI), blood urea nitrogen (BUN) level, serum uric acid (UA) level, serum calcium (Ca) level, serum phosphorus (P) level, the use of oral contraceptive pills, the use of hormone replacement therapy (HRT), smoking status, drinking status, and the use of corticosteroids were adjusted using a weighted multiple regression model. Subgroup analyses were performed using the same regression model. We included 2198 female participants in the study, and testosterone levels were positively associated with lumbar BMD after adjusting for all the covariates (β = 1.12, 95% CI 0.31, 1.93). In subgroup analyses, the associations in the fourth quartile of testosterone levels were stronger for the participants aged 40–50 years old (quartile 4, β = 42.92, 95% CI 7.53, 78.30 vs. quartile 1) and 50 to 60-year-old (quartile 4, β = 32.41, 95% CI 0.14, 64.69 vs. quartile 1). Similar results were found in other subgroups, including subgroups for race (Non-Hispanic Black, Other), income level (income ≤ 1.3, income > 3.5), education level (college or higher), BMI > 25 kg/m2, BUN levels ≤ 20 mg/dL, UA levels ≤ 6 mg/dL, Ca levels ≤ 10.1 mg/dL, P levels ≤ 5 mg/dL, drinking status, never smoker, never taking birth control pills, and HRT user. There was no interaction among the covariates in the association between lumbar BMD and testosterone levels (P for interaction > 0.05). In US adult females aged 40–60 years, the testosterone level was a positive predictor of the lumbar BMD after adjusting for covariates.
Collapse
|
18
|
Nasimi Doost Azgomi R, Moini Jazani A, Karimi A, Pourreza S. Potential roles of genistein in polycystic ovary syndrome: A comprehensive systematic review. Eur J Pharmacol 2022; 933:175275. [PMID: 36108737 DOI: 10.1016/j.ejphar.2022.175275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 12/09/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most prevalent polygenic endocrine disorders in reproductive-age women. Genistein is a soy-isolated phytoestrogen and isoflavone with antioxidant, anti-inflammatory, estrogenic, and antineoplastic activity. This systematic review aimed to investigate the therapeutic effects and mechanisms of actions of genistein in PCOS. The present study was conducted according to the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) protocol. We searched PubMed, Scopus, Embase, and Google Scholar databases up to February 2022 using relative keywords. Studies published in English evaluated genistein's effects on PCOS, and its related symptoms were considered. Out of 298 records screened, only 13 articles met the inclusion criteria: Nine animal and 4 human studies. The results of the current study indicated that genistein supplementation may effectively improve PCOS-related symptoms by decreasing insulin resistance and anthropometric indices, improving ovarian morphology and regulating reproductive hormones, and reducing oxidative stress and inflammation by influencing biological pathways. According to the current literature, genistein may diminish the dues of PCOS. Therefore, this study shows that genistein can be considered an effective agent. in reducing the complications of PCOS. However, further studies are recommended for a broad conclusion on the exact mechanism of genistein in PCOS patients.
Collapse
Affiliation(s)
- Ramin Nasimi Doost Azgomi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Arezoo Moini Jazani
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Arash Karimi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sanaz Pourreza
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Li L, Zhang J, Lu C, Wang B, Guo J, Zhang H, Cui S. MicroRNA-7a2 Contributes to Estrogen Synthesis and Is Modulated by FSH via the JNK Signaling Pathway in Ovarian Granulosa Cells. Int J Mol Sci 2022; 23:ijms23158565. [PMID: 35955699 PMCID: PMC9369042 DOI: 10.3390/ijms23158565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022] Open
Abstract
MicroRNA-7a2 (miR-7a2) plays fundamental roles in the female reproductive axis, and estrogen is indispensable for maintaining ovary function. However, the interaction between miR-7a2 and ovarian function is unclear. The present study aimed to determine whether and how miR-7a2 functions in estrogen synthesis. Firstly, the results verified that miR-7a was highly expressed in ovarian granulosa cells. The knockout (KO) of miR-7a2 caused infertility and abnormal ovarian function in mice. Concomitantly, the Cyp19a1 expression and estrogen synthesis were significantly inhibited, which was validated in primary granulosa cells. The mice transplanted with miR-7a2 KO ovaries showed similar results; however, estrogen supplementation reversed infertility. In the in vitro experiment, follicle-stimulating hormone (FSH) significantly improved the expression of miR-7a and Cyp19a1 and the synthesis of estrogen. However, the miR-7a2 KO markedly reversed the function of FSH. Also, FSH upregulated miR-7a by activating the (c-Jun N-terminal kinase) JNK signaling pathway. In addition, Golgi apparatus protein 1 (Glg1) was shown to be the target gene of miR-7a2. These findings indicated that miR-7a2 is essential for ovarian functions with respect to estrogen synthesis through the targeted inhibition of the expression of Glg1 and then promoting Cyp19a1 expression; the physiological process was positively regulated by FSH via the JNK signaling pathway in granulosa cells.
Collapse
Affiliation(s)
- Liuhui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Jinglin Zhang
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou 225009, China;
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Chenyang Lu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Bingjie Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Jiajia Guo
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Haitong Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
20
|
Garzia E, Galiano V, Guarnaccia L, Marfia G, Murru G, Guermandi E, Riparini J, Sulpizio P, Marconi AM. Basal serum level of Δ4-androstenedione reflects the ovaries' ability to respond to stimulation in IVF cycles: setting up a new reliable index of both ovarian reserve and response. J Assist Reprod Genet 2022; 39:1917-1926. [PMID: 35759063 PMCID: PMC9428103 DOI: 10.1007/s10815-022-02546-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/09/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Adequate androgen levels are necessary for regular follicular growth, progression beyond the pre-antral stage, and prevention of follicular atresia. The main purpose of this study was to investigate whether baseline androgen levels had a predictive value on stimulation outcomes in IVF cycles. The secondary purpose was to compare the possible predictive value of androgens with that of already known markers. METHODS The study included 91 infertile patients aged 30-45 years awaiting the first IVF cycle. All women underwent the same stimulation protocol and the same starting dose of recombinant FSH. As stimulation outcomes, the number of follicles recruited, estradiol and progesterone levels on the day of trigger, the total dose of gonadotropins administered, and the number of oocytes collected were recorded. Multiple linear regression and multivariate logistic regression were used to evaluate the significant predictive value of the variables for response to controlled ovarian stimulation (COS). By studying the reliability of different markers, an attempt was made to develop a single index with the highest predictive value. RESULTS Pearson's correlation revealed a statistically significant inverse correlation between oocytes collected and age (r = - 0.333, p < 0.001) and a positive correlation with AMH (anti-müllerian hormone) (r = 0.360, p < 0.001), antral follicle count (AFC) (r = 0.639, p < 0.001), and androstenedione (Δ4-A) (r = 0.359, p < 0.001). No significant correlation was reported with FSH (r = - 0.133, p = 0.207) and total testosterone (r = 0.180, p = 0.088). In COS good responders, the G-index (= AMH ng/mL*AFC/Δ4-A ng/dL) revealed a significantly higher level (p < 0.001) than AMH, AFC, and Δ4-A alone. CONCLUSION Baseline serum Δ4-A, presumably crucial for ensuring a regular follicular growth, is a reliable marker of ovarian response to stimulation. Since the ovarian capacity to respond to gonadotropins does not depend exclusively on the presence of follicles, we suggest a new index, the G-index, able to contemplate both the ovarian reserve and the Δ4-A level.
Collapse
Affiliation(s)
- Emanuele Garzia
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
- Istituto Di Medicina Aerospaziale “A. Mosso,” Aeronautica Militare, Milan, Italy
| | - Valentina Galiano
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
- Gynecology and Obstetrics Unit, San Carlo Borromeo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Aldo Ravelli Research Center, Milan, Italy
| | - Giovanni Marfia
- Istituto Di Medicina Aerospaziale “A. Mosso,” Aeronautica Militare, Milan, Italy
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Aldo Ravelli Research Center, Milan, Italy
| | - Giulia Murru
- Gynecology and Obstetrics Unit, ASST Rhodense, Ospedale G. Salvini, Garbagnate Milanese, Italy
| | - Ellade Guermandi
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Jennifer Riparini
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Patrizia Sulpizio
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Anna Maria Marconi
- Gynecology and Obstetrics Unit, Department of Mother and Child, Department of Health Sciences, San Paolo Hospital Medical School, University of Milan, Milan, Italy
| |
Collapse
|
21
|
AOP key event relationship report: Linking decreased androgen receptor activation with decreased granulosa cell proliferation of gonadotropin-independent follicles. Reprod Toxicol 2022; 112:136-147. [PMID: 35868514 DOI: 10.1016/j.reprotox.2022.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 02/08/2023]
Abstract
We recently proposed to formally recognize Key Event Relationships (KERs) as building blocks of Adverse Outcome Pathways (AOPs) that can be independently developed and peer-reviewed. Here, we follow this approach and provide an independent KER from AOP345, which describes androgen receptor (AR) antagonism leading to decreased female fertility. This KER connects AR antagonism to reduced granulosa cell proliferation of gonadotropin-independent follicles (KER2273). We have developed both the KER and the two adjacent Key Events (KEs). A systematic approach was used to ensure that all relevant supporting evidence for KER2273 was retrieved. Supporting evidence for the KER highlights the importance of AR action during the early stages of follicular development. Both biological plausibility and empirical evidence are presented, with the latter also assessed for quality. We believe that tackling isolated KERs instead of whole AOPs will accelerate the AOP development. Faster AOP development will lead to the development of simple test methods that will aid screening of chemicals, endocrine disruptor identification, risk assessment, and subsequent regulation.
Collapse
|
22
|
Sex Steroid Receptors in Polycystic Ovary Syndrome and Endometriosis: Insights from Laboratory Studies to Clinical Trials. Biomedicines 2022; 10:biomedicines10071705. [PMID: 35885010 PMCID: PMC9312843 DOI: 10.3390/biomedicines10071705] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/13/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) and endometriosis are reproductive disorders that may cause infertility. The pathology of both diseases has been suggested to be associated with sex steroid hormone receptors, including oestrogen receptors (ER), progesterone receptors (PRs) and androgen receptors (ARs). Therefore, with this review, we aim to provide an update on the available knowledge of these receptors and how their interactions contribute to the pathogenesis of PCOS and endometriosis. One of the main PCOS-related medical conditions is abnormal folliculogenesis, which is associated with the downregulation of ER and AR expression in the ovaries. In addition, metabolic disorders in PCOS are caused by dysregulation of sex steroid hormone receptor expression. Furthermore, endometriosis is related to the upregulation of ER and the downregulation of PR expression. These receptors may serve as therapeutic targets for the treatment of PCOS-related disorders and endometriosis, considering their pathophysiological roles. Receptor agonists may be applied to increase the expression of a specific receptor and treat endometriosis or metabolic disorders. In contrast, receptor antagonist functions to reduce receptor expression and can be used to treat endometriosis and induce ovulation. Understanding PCOS and the pathological roles of endometriosis sex steroid receptors is crucial for developing potential therapeutic strategies to treat infertility in both conditions. Therefore, research should be continued to fill the knowledge gap regarding the subject.
Collapse
|
23
|
Ramadhan RS, Algafari RN, Jarallah AL. Investigating pathogenic SNPs in androgen receptor with direct influence on polycystic ovary syndrome (PCOS) in women. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00292-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Polycystic ovary syndrome (PCOS) became one of the main reasons for infertility in women. It has an obvious effect on phenotype represented by hirsutism, increased body mass index, obesity, and acne, while biochemical tests show adverse hormonal imbalance with hyperandrogenism as testosterone levels increases. From molecular level point of view, pathogenic SNPs may change CAG repeats number along androgen receptor (AR) resulting in altered function of the gene causing different affinity to androgen hormones.
Methods
Recruiting 150 patients diagnosed with PCOS for the study, genomic DNA was extracted and amplified using specifically designed exon 1 PCR primers employing gene walking technique. The resulting amplicons were sequenced and thoroughly analyzed for polymorphism and CAG repeats number.
Results
Data obtained from recruiting 150 patients diagnosed with PCOS showed that sequences X:67545209–67545742; X:67545503–67545739 of exon 1 harbored 7 SNPs altered secondary structure of the resulting protein and forced toward the use of CAA as synonymous codon instead of the normal CAGs stretches. This led to produced alternative mRNA that eventually changed nonsense-mediated mRNA decay mechanism.
Conclusion
Probability of PCOS in women with polymorphic AR gene is higher than others, especially women with high number of CAG stretches. The new finding and highlight of this study is that alternative codon usage (CAAs) to produce the same amino acid (Gln) and compensate the reduced number of CAG repeats number may be attributed to epigenetic mechanism to mitigate the adverse effect of such change and maintain a normal function of AR gene. This finding was not previously reported in former studies.
Collapse
|
24
|
Neves AR, Montoya-Botero P, Polyzos NP. Androgens and diminished ovarian reserve: the long road from basic science to clinical implementation. A comprehensive and systematic review with meta-analysis. Am J Obstet Gynecol 2022; 227:401-413.e18. [PMID: 35364061 DOI: 10.1016/j.ajog.2022.03.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/16/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE This study aimed to present a narrative review regarding androgen production, androgens' role in folliculogenesis, and the available therapeutic approaches for androgen supplementation, and to perform a systematic review and meta-analysis regarding the impact of androgens (dehydroepiandrosterone/testosterone) compared with placebo or no treatment on ovarian response and pregnancy outcomes in patients with diminished ovarian reserve and/or poor ovarian responders. DATA SOURCES An electronic search of MEDLINE, Embase, Cochrane Library, Cochrane Central Register of Controlled Trials, Scopus, ClinicalTrials.gov, the ISRCTN registry, and the World Health Organization International Clinical Trials Registry, was conducted for studies published until September 2021. STUDY ELIGIBILITY CRITERIA Randomized controlled trials that compared ovarian response and/or pregnancy outcomes between the different in vitro fertilization protocols using androgens (ie, dehydroepiandrosterone and testosterone) and conventional in vitro fertilization stimulation in patients with diminished ovarian reserve and/or poor ovarian responders were included. METHODS The quality of each study was evaluated with the revised Cochrane risk-of-bias tool for randomized trials (RoB 2). The meta-analysis used random-effects models. All results were interpreted on the basis of intention-to-treat analysis (defined as the inclusion of all randomized patients in the denominator). Risk ratios and 95% confidence intervals were used and combined for meta-analysis. RESULTS No significant differences were found regarding the number of oocytes retrieved (mean difference, 0.76; 95% confidence interval, -0.35 to 1.88), mature oocytes retrieved (mean difference, 0.25; 95% confidence interval, -0.27 to 0.76), clinical pregnancy rate (risk ratio, 1.17; 95% confidence interval, 0.87-1.57), live-birth rate (risk ratio, 0.97; 95% confidence interval, 0.47-2.01), or miscarriage rate (risk ratio, 0.80; 95% confidence interval, 0.29-2.22) when dehydroepiandrosterone priming was compared with placebo or no treatment. Testosterone pretreatment yielded a higher number of oocytes retrieved (mean difference, 0.94; 95% confidence interval, 0.46-1.42), a higher clinical pregnancy rate (risk ratio, 2.07; 95% confidence interval, 1.33-3.20), and higher live-birth rate (risk ratio, 2.09; 95% confidence interval, 1.11-3.95). CONCLUSION Although dehydroepiandrosterone did not present a clear effect on outcomes of assisted reproductive techniques, we found a potentially beneficial effect of testosterone priming on ovarian response and pregnancy outcomes. However, results should be interpreted with caution, taking into account the low to moderate quality of the available evidence.
Collapse
Affiliation(s)
- Ana Raquel Neves
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain; Autonomous University of Barcelona, Cerdanyola del Vallès, Spain
| | - Pedro Montoya-Botero
- Conceptum - Unidad de Fertilidad del Country, Bogotá, Colombia; Department of Epidemiology and Biostatistics, Fundación Universitaria de Ciencias de la Salud - FUCS, Bogotá, Colombia
| | - Nikolaos P Polyzos
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain; Faculty of Medicine and Health Sciences, Ghent University (UZ Gent), Ghent, Belgium.
| |
Collapse
|
25
|
Nabi M, Andrabi SM, Rasool SUA, Ashraf S, Majid I, Amin S. Androgen receptor coregulator long noncoding RNA CTBP1-AS is associated with polycystic ovary syndrome in Kashmiri women. Endocrine 2022; 75:614-622. [PMID: 34611799 DOI: 10.1007/s12020-021-02894-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/24/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is one of the most common reproductive, endocrine, and metabolic disorder in premenopausal women. Even though the pathophysiology of PCOS is complex and obscure, the disorder is prominently considered as the syndrome of hyperandrogenism. C-Terminal binding protein 1 antisense (CTBP1-AS) acts as a novel androgen receptor regulating long noncoding RNA (lncRNA). Therefore, the present study was aimed to establish the possible association of androgen receptor regulating long noncoding RNA CTBP1-AS with PCOS. METHODS A total of 178 subjects including 105 PCOS cases and 73 age-matched healthy controls were recruited for the study. The anthropometric, hormonal, and biochemical parameters of all subjects were analyzed. Total RNA was isolated from peripheral venous blood and expression analysis was done by quantitative real-time PCR. The correlation analysis was performed to evaluate the association between and various clinical parameters and lncRNA CTBP1-AS expression. RESULTS AND CONCLUSION The mean expression level of CTBP1-AS was found to be significantly higher in the PCOS women than in the healthy controls (-lnCTBP1-AS, 4.23 ± 1.68 versus 1.24 ± 0.29, P < 0.001). Furthermore, subjects with higher expression level of CTBP1-AS had significantly higher risk of PCOS compared to subjects with low levels of CTBP1-AS expression (actual OR = 11.36, 95% CI = 5.59-23.08, P < 0.001). The area under receiver operator characteristic (ROC) curve was 0.987 (SE 0.006, 95% CI 0.976-0.99). However, lncRNA CTBP1-AS was found to have no association with different clinical characteristics of PCOS. In conclusion, androgen receptor coregulating lncRNA CTBP1-AS is associated with PCOS women and high expression of CTBP1-AS is a risk factor for PCOS in Kashmiri women.
Collapse
Affiliation(s)
- Mudasar Nabi
- Department of Biochemistry, University of Kashmir, Hazratbal, Srinagar, 190006, India
| | - Syed Mudasir Andrabi
- Division of Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Shuhama, Kashmir, 190006, India
| | | | - Sairish Ashraf
- Department of Biochemistry, University of Kashmir, Hazratbal, Srinagar, 190006, India
| | - Imran Majid
- Cutis Institute of Dermatology, Hyderpora, Srinagar, 190014, India
| | - Shajrul Amin
- Department of Biochemistry, University of Kashmir, Hazratbal, Srinagar, 190006, India.
| |
Collapse
|
26
|
Kuan KKW, Saunders PTK. Female Reproductive Systems: Hormone Dependence and Receptor Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:21-39. [PMID: 36107311 DOI: 10.1007/978-3-031-11836-4_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The female reproductive system which consists of the ovaries, uterus (myometrium, endometrium), Fallopian tubes, cervix and vagina is exquisitely sensitive to the actions of steroid hormones. The ovaries play a key role in the synthesis of bioactive steroids (oestrogens, androgens, progestins) that act both within the tissue (intracrine/paracrine) as well as on other reproductive organs following release into the blood stream (endocrine action). Sex steroid receptors encoded by the oestrogen (ESR1, ESR2), progesterone (PR) and androgen (AR) receptor genes, which are members of the superfamily of ligand activated transcription factors are widely expressed within these tissues. These receptors play critical role(s) in regulation of cell proliferation, ovulation, endometrial receptivity, myometrial cell function and inflammatory cell infiltration. Our understanding of their importance has been informed by studies on human tissues and cells, which have employed immunohistochemistry as well as a wide range of molecular and genetic methods to identify which processes are dependent steroid ligand activation. The development of mice with targeted deletions of each of these receptors has provided complementary data that has extended our appreciation of cell-cell interactions in the fine tuning of reproductive tissue function. This large body of work has formed the basis of new and improved therapeutics to treat conditions such as infertility.
Collapse
Affiliation(s)
- Kevin K W Kuan
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
27
|
Skoracka K, Ratajczak AE, Rychter AM, Dobrowolska A, Krela-Kaźmierczak I. Female Fertility and the Nutritional Approach: The Most Essential Aspects. Adv Nutr 2021; 12:2372-2386. [PMID: 34139003 PMCID: PMC8634384 DOI: 10.1093/advances/nmab068] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/19/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022] Open
Abstract
Infertility is an increasing problem that affects couples attempting pregnancy. A growing body of evidence points to a link between diet and female fertility. In fact, data show that a diet high in trans fats, refined carbohydrates, and added sugars can negatively affect fertility. Conversely, a diet based on the Mediterranean dietary patterns, i.e., rich in dietary fiber, omega-3 (ɷ-3) fatty acids, plant-based protein, and vitamins and minerals, has a positive impact on female fertility. An unhealthy diet can disrupt microbiota composition, and it is worth investigating whether the composition of the gut microbiota correlates with the frequency of infertility. There is a lack of evidence to exclude gluten from the diet of every woman trying to become pregnant in the absence of celiac disease. Furthermore, there are no data concerning adverse effects of alcohol on female fertility, and caffeine consumption in the recommended amounts also does not seem to affect fertility. On the other hand, phytoestrogens presumably have a positive influence on female fertility. Nevertheless, there are many unanswered questions with regard to supplementation in order to enhance fertility. It has been established that women of childbearing age should supplement folic acid. Moreover, most people experience vitamin D and iodine deficiency; thus, it is vital to control their blood concentrations and consider supplementation if necessary. Therefore, since diet and lifestyle seem to be significant factors influencing fertility, it is valid to expand knowledge in this area.
Collapse
Affiliation(s)
- Kinga Skoracka
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, the Heliodor Swiecicki Hospital, Poznan, Poland
| | - Alicja Ewa Ratajczak
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, the Heliodor Swiecicki Hospital, Poznan, Poland
| | - Anna Maria Rychter
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, the Heliodor Swiecicki Hospital, Poznan, Poland
| | - Agnieszka Dobrowolska
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, the Heliodor Swiecicki Hospital, Poznan, Poland
| | - Iwona Krela-Kaźmierczak
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, the Heliodor Swiecicki Hospital, Poznan, Poland
| |
Collapse
|
28
|
Abstract
The approach to hyperandrogenism in women varies depending on the woman's age and severity of symptoms. Once tumorous hyperandrogenism is excluded, the most common cause is PCOS. Hirsutism is the most common presenting symptom. The woman's concern about her symptoms plays an important role in the management of disease. Although measurement of testosterone is useful in identifying an underlying cause, care must be taken when interpreting the less accurate assays that are available commercially. Surgical resection is curative in tumorous etiologies, whereas medical management is the mainstay for non-tumorous causes.
Collapse
Affiliation(s)
- Anu Sharma
- Division of Endocrinology, Metabolism and Diabetes, University of Utah, EIHG 2110A, 15 N 2030 E, Salt Lake City, UT 84112, USA
| | - Corrine K Welt
- Division of Endocrinology, Metabolism and Diabetes, University of Utah, EIHG 2110A, 15 N 2030 E, Salt Lake City, UT 84112, USA.
| |
Collapse
|
29
|
Jin X, Cheng J, Shen J, Lv X, Li Q, Mu Y, Bai H, Liu Y, Xia Y. Moxibustion improves ovarian function based on the regulation of the androgen balance. Exp Ther Med 2021; 22:1230. [PMID: 34539826 PMCID: PMC8438671 DOI: 10.3892/etm.2021.10664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 06/18/2021] [Indexed: 12/26/2022] Open
Abstract
The effect of androgens on follicular development and female reproduction has become an active research topic. Moxibustion is a Traditional Chinese Medicine therapy that has been reported to be able to prevent and treat numerous ovary-related problems. However, studies on the effect of moxibustion for diminished ovarian reserve (DOR) on androgen balance are still lacking. The present study aimed to assess the efficacy of moxibustion intervention prior to disease onset and at the early stage of disease in a rat model of DOR and explore the mechanisms of its effect on ovarian function. A total of 32 rats were randomly divided into four groups: Blank group, Model group (a drug-induced model of DOR), Moxibustion group 1 and Moxibustion group 2. Moxibustion was performed on the BL23 and RN4 acupoints of female rats daily for a total of 20 days (once a day, five times a week for a total of 4 weeks). The two moxibustion groups were established with different intervention times: One group was subjected to pre-disease intervention and the other group to early-disease intervention. The ovarian function was evaluated by detecting anti-Mullerian hormone (AMH), follicle-stimulating hormone (FSH), estradiol (E2), testosterone (T), dehydroepiandrosterone (DHEA), dihydrotestosterone (DHT) and androgen receptor (AR) levels in the serum or the ovary samples. To further investigate the downstream regulatory factors for AR after moxibustion treatment for pre-disease or early-disease intervention, FSH receptor (FSHR) and microRNA (miR)-125b expression in ovaries were also analyzed. The results indicated that AMH and DHT levels were reduced in the model group compared with those in the blank group, while FSH, T and DHEA levels were increased. AMH and DHT levels were increased in Moxibustion group 1 compared with those in the model group, while FSH, T and DHEA levels were reduced. There was no difference in E2 levels between Moxibustion group 1 and the model group. Compared with that in the model group, the AR content in the ovary was increased in Moxibustion group 1. There was no difference in FSHR mRNA in the ovaries between Moxibustion group 1 and the model group. miR-125b levels were significantly increased in Moxibustion group 1 as compared with those in the model group. Furthermore, AMH and DHT levels were increased in Moxibustion group 2 compared with those in the model group, while FSH, T and DHEA levels were reduced. E2 levels were significantly decreased in Moxibustion group 2 compared with those in the model group. The relative mRNA expression of AR, FSHR and miR-125b was decreased following establishment of the model. Compared with that in the model group, the AR content in the ovary was increased in Moxibustion group 2. In comparison with the blank and model groups, the FSHR content in the ovary of Moxibustion group 2 was significantly increased. miR-125b levels were not obviously altered in Moxibustion group 2 as compared with those in the model group. In addition, there was no significant difference in AMH, FSH, T and DHEA levels between the two moxibustion groups. E2 and DHT levels were higher in Moxibustion group 1 than in Moxibustion group 2. There was no difference in AR mRNA expression between the two moxibustion groups. FSHR mRNA levels were lower in Moxibustion group 1 than in Moxibustion group 2, while miR-125b mRNA levels were higher in Moxibustion group 1 than in Moxibustion group 2. In conclusion, the present study suggested that moxibustion intervention prior to disease onset and at the early disease stage was able to improve ovarian function via modulation of the AR-mediated stable equilibrium of androgens. However, the effects and mechanisms of moxibustion intervention for pre-disease and early-disease intervention of DOR appear to be different. The appropriate duration of treatment and the time-effect relationship require to be further studied.
Collapse
Affiliation(s)
- Xun Jin
- College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Jie Cheng
- College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Jie Shen
- College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xing Lv
- Central Research Institute of Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 200020, P.R. China
| | - Qian Li
- College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yanyun Mu
- College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Hua Bai
- College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yan Liu
- College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Youbing Xia
- Traditional Chinese Medicine Department, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| |
Collapse
|
30
|
Evliyaoglu O, Imöhl M, Weiskirchen R, van Helden J. Age-specific reference values improve the diagnostic performance of AMH in polycystic ovary syndrome. Clin Chem Lab Med 2021; 58:1291-1301. [PMID: 32069226 DOI: 10.1515/cclm-2019-1059] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022]
Abstract
Background The increased secretion of anti-Müllerian hormone (AMH) by the growing follicles has been supposed as a determinative feature of polycystic ovary syndrome (PCOS). The diagnostic performance of AMH in PCOS is superior compared to the free androgen index (FAI) and luteinizing hormone (LH)/follicle-stimulating hormone (FSH) quotient. We established age-dependent reference ranges to further improve the diagnostic performance of AMH. Methods In a cross-sectional study, in samples of 4712 reproductive age patients, ranging from 14 to 50 years, BMI, AMH and other reproductive hormones were determined by immunoassay or tandem mass spectrometry (LC-MS/MS) to calculate age-specific reference ranges and the diagnostic performance. Results Age-specific diagnostic performances for Elecsys® AMH, FAI and LH/FSH ratio were established in the reference group. No significant difference in BMI was found between the groups. AMH values were significantly negatively correlated with age (r = -0.628, p < 0.001) in patients with normal ovarian function, but there was no correlation between age and AMH levels in PCOS patients (r = - 0.041, p < 0.174). In all the study groups, AMH showed a weak correlation between FAI and LH/FSH ratio (r = 0.302, p < 0.001 and r = 0.434, p < 0.001, respectively). The sensitivity/specificity for AMH, FAI and LH/FSH ratio were 89/96%, 71/69% and 75/72%, respectively, according to the Youden index. Conclusions We determined the age-dependent reference ranges for serum AMH levels in a large population-based study and calculated the age-specific diagnostic performance of FAI and LH/FSH ratio, which allows physicians to evaluate patients with PCOS who have normal AMH levels. AMH is suggested as the strongest diagnostic marker in patients with PCOS compared to FAI and LH/FSH ratio.
Collapse
Affiliation(s)
- Osman Evliyaoglu
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH, Aachen, Germany
| | - Mathias Imöhl
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH, Aachen, Germany
| | - Josef van Helden
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany
| |
Collapse
|
31
|
Salinas I, Sinha N, Sen A. Androgen-induced epigenetic modulations in the ovary. J Endocrinol 2021; 249:R53-R64. [PMID: 33764313 PMCID: PMC8080881 DOI: 10.1530/joe-20-0578] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/24/2021] [Indexed: 12/16/2022]
Abstract
In recent years, androgens have emerged as critical regulators of female reproduction and women's health in general. While high levels of androgens in women are associated with polycystic ovary syndrome (PCOS), recent evidence suggests that a certain amount of direct androgen action through androgen receptor is also essential for normal ovarian function. Moreover, prenatal androgen exposure has been reported to cause developmental reprogramming of the fetus that manifests into adult pathologies, supporting the Developmental Origins of Health and Disease (DOHaD) hypothesis. Therefore, it has become imperative to understand the underlying mechanism of androgen actions and its downstream effects under normal and pathophysiological conditions. Over the years, there has been a lot of studies on androgen receptor function as a transcriptional regulator in the nucleus as well as androgen-induced rapid extra-nuclear signaling. Conversely, new evidence suggests that androgen actions may also be mediated through epigenetic modulation involving both the nuclear and extra-nuclear androgen signaling. This review focuses on androgen-induced epigenetic modifications in female reproduction, specifically in the ovary, and discusses emerging concepts, latest perceptions, and highlight the areas that need further investigation.
Collapse
Affiliation(s)
- Irving Salinas
- Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Niharika Sinha
- Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Animal Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Aritro Sen
- Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Animal Sciences, Michigan State University, East Lansing, MI 48824, USA
- Corresponding author and person to whom reprint request should be addressed: Aritro Sen Ph.D., Reproductive and Developmental Sciences Program, 3013 Interdisciplinary Science & Technology Building, 766 Service Road, Michigan State University, East Lansing, MI 48824, Ph:517-432-4585;
| |
Collapse
|
32
|
Serum testosterone levels are positively associated with serum anti-mullerian hormone levels in infertile women. Sci Rep 2021; 11:6336. [PMID: 33737663 PMCID: PMC7973568 DOI: 10.1038/s41598-021-85915-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/05/2021] [Indexed: 11/09/2022] Open
Abstract
Anti-Mullerian hormone (AMH) and testosterone (T) both play distinct roles in the early stages of folliculogenesis. However, the relationship between serum T and AMH levels is poorly understood. This study aimed to investigate the association between serum T and AMH levels in infertile women. A total of 1935 infertile women aged 20-46 years were included in the cross-sectional study and divided into four quartile groups (Q1 to Q4) based on serum T levels. Compared to the subjects in the highest T quartile (Q4), those in the lowest T quartile (Q1) showed significantly lower AMH levels. After adjustment for age, body weight, body mass index and FSH, increasing T quartile categories were associated with higher AMH levels. Binary logistic regression analyses revealed that the odds for the risk of diminished ovarian reserve (DOR) were 11.44-fold higher in Q1 than in Q4 and the odds for the risk of excess ovarian reserve (EOR) were 10.41-fold higher in Q4 than in Q1. Our data show that serum T levels are positively associated with serum AMH levels and suggest that androgen insufficiency may be a potential risk factor for DOR; androgen excess may lead to EOR in infertile women.
Collapse
|
33
|
The Relationships Between Serum DHEA-S and AMH Levels in Infertile Women: A Retrospective Cross-Sectional Study. J Clin Med 2021; 10:jcm10061211. [PMID: 33803980 PMCID: PMC7998713 DOI: 10.3390/jcm10061211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
The relationship between serum dehydroepiandrosterone sulphate (DHEA-S) and anti-Mullerian hormone (AMH) levels has not been fully established. Therefore, we performed a large-scale cross-sectional study to investigate the association between serum DHEA-S and AMH levels. The study included a total of 2155 infertile women aged 20 to 46 years who were divided into four quartile groups (Q1 to Q4) based on serum DHEA-S levels. We found that there was a weak positive association between serum DHEA-S and AMH levels in infertile women (r = 0.190, p < 0.001). After adjusting for potential confounders, serum DHEA-S levels positively correlated with serum AMH levels in infertile women (β = 0.103, p < 0.001). Infertile women in the highest DHEA-S quartile category (Q4) showed significantly higher serum AMH levels (p < 0.001) compared with women in the lowest DHEA-S quartile category (Q1). The serum AMH levels significantly increased across increasing DHEA-S quartile categories in infertile women (p = 0.014) using generalized linear models after adjustment for potential confounders. Our data show that serum DHEA-S levels are positively associated with serum AMH levels.
Collapse
|
34
|
Ozgen Saydam B, Yildiz BO. Polycystic Ovary Syndrome and Brain: An Update on Structural and Functional Studies. J Clin Endocrinol Metab 2021; 106:e430-e441. [PMID: 33205212 DOI: 10.1210/clinem/dgaa843] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Indexed: 12/25/2022]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is the most common endocrine disorder of women in reproductive age and is associated with reproductive, endocrine, metabolic, cardiovascular, and psychological outcomes. All these disorders are thought to be affected by central mechanisms which could be a major contributor in pathogenesis of PCOS. EVIDENCE ACQUISITION This mini-review discusses the relevance of central nervous system imaging modalities in understanding the neuroendocrine origins of PCOS as well as their relevance to understanding its comorbidities. EVIDENCE SYNTHESIS Current data suggest that central nervous system plays a key role in development of PCOS. Decreased global and regional brain volumes and altered white matter microstructure in women with PCOS is shown by structural imaging modalities. Functional studies show diminished reward response in corticolimbic areas, brain glucose hypometabolism, and greater opioid receptor availability in reward-related regions in insulin-resistant patients with PCOS. These structural and functional disturbances are associated with nonhomeostatic eating, diminished appetitive responses, as well as cognitive dysfunction and mood disorders in women with PCOS. CONCLUSION Structural and functional brain imaging is an emerging modality in understanding pathophysiology of metabolic disorders such as diabetes and obesity as well as PCOS. Neuroimaging can help researchers and clinicians for better understanding the pathophysiology of PCOS and related comorbidities as well as better phenotyping PCOS.
Collapse
Affiliation(s)
- Basak Ozgen Saydam
- Division of Endocrinology and Metabolism, Dokuz Eylul University School of Medicine, İzmir, Turkey
| | - Bulent Okan Yildiz
- Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
35
|
Neves AR, Montoya-Botero P, Polyzos NP. The Role of Androgen Supplementation in Women With Diminished Ovarian Reserve: Time to Randomize, Not Meta-Analyze. Front Endocrinol (Lausanne) 2021; 12:653857. [PMID: 34079524 PMCID: PMC8165260 DOI: 10.3389/fendo.2021.653857] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/23/2021] [Indexed: 11/24/2022] Open
Abstract
The management of patients with diminished ovarian reserve (DOR) remains one of the most challenging tasks in IVF clinical practice. Despite the promising results obtained from animal studies regarding the importance of androgens on folliculogenesis, the evidence obtained from clinical studies remains inconclusive. This is mainly due to the lack of an evidence-based methodology applied in the available trials and to the heterogeneity in the inclusion criteria and IVF treatment protocols. In this review, we analyze the available evidence obtained from animal studies and highlight the pitfalls from the clinical studies that prevent us from closing the chapter of this line of research.
Collapse
Affiliation(s)
- Ana Raquel Neves
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
- Faculty of Medicine, Autonomous University of Barcelona, Cerdanyola del Vallès, Spain
| | - Pedro Montoya-Botero
- Department of Reproductive Medicine, Conceptum – Unidad de Fertilidad del Country, Bogotá, Colombia
| | - Nikolaos P. Polyzos
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
- Faculty of Medicine and Health Sciences, Ghent University (UZ Gent), Gent, Belgium
- *Correspondence: Nikolaos P. Polyzos,
| |
Collapse
|
36
|
Ye W, Xie T, Song Y, Zhou L. The role of androgen and its related signals in PCOS. J Cell Mol Med 2020; 25:1825-1837. [PMID: 33369146 PMCID: PMC7882969 DOI: 10.1111/jcmm.16205] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/29/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women at reproductive age. However, the underlying pathogenic mechanisms have not been completely understood. Hyperandrogenism is an important clinic feature in patients with PCOS, suggesting its pathologic role in the development and progression of PCOS. However, the actual role of androgen and the related signals in PCOS and PCOS-related complications have not yet been clarified. In this review, we surveyed the origin and effects of androgen on PCOS and the related complications, highlighted the cellular signals affecting androgen synthesis and summarized the pathological processes caused by hyperandrogenism. Our review well reveals the important mechanisms referring the pathogenesis of PCOS and provides important clues to the clinic strategies in patients with PCOS.
Collapse
Affiliation(s)
- Wenting Ye
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Xie
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yali Song
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
37
|
Tian L, Zou Y, Tan J, Wang Y, Chen J, Xia L, Xu L, Chen G, Wu Q, Huang O. Androgen receptor gene mutations in 258 Han Chinese patients with polycystic ovary syndrome. Exp Ther Med 2020; 21:31. [PMID: 33262817 PMCID: PMC7690241 DOI: 10.3892/etm.2020.9463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/15/2020] [Indexed: 01/12/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) affects 8-13% of reproductive-age females worldwide and mutations or aberrant expression of androgen receptor (AR) may cause the onset of this disease. In the present study, 258 samples from Han Chinese patients with PCOS were analyzed for the presence of AR mutations via sequencing of all coding exons of the AR gene. A total of five heterozygous missense mutations, namely p.V3M, p.Q72R, p.S158L, p.S176R and p.G396R, were identified in five of the patients. Among these, p.S158L was a novel mutation that, to the best of our knowledge, has not been reported previously. Although the remaining four mutations have been reported previously, they existed at low frequencies or were absent in the control subjects and in the Exome Aggregation Consortium database. The results of evolutionary conservation and in silico analysis revealed that the p.V3M, p.S158L and p.S176R mutations were pathogenic, whereas the p.Q72R and p.G396R mutations were benign. Compared with the patients with PCOS without AR mutations or with benign AR mutations, markedly lower estrogen levels on the day of human chorionic gonadotropin injection were observed in the three patients with PCOS with potentially pathogenic mutations. In addition, patients with PCOS with pathogenic mutations had lower numbers of oocytes; however, the difference was not statistically significant. Of note, these observations should be interpreted with caution due to the relatively small sample size in the present study. Therefore, a larger number of samples should be collected to validate the results of the present study in future studies. In summary, the present study identified three potential pathogenic mutations in 258 Han Chinese patients with PCOS and these mutations may have an implication in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Lifeng Tian
- Department of Gynecology and Obstetrics, Jiangxi Medical College, Graduate School of Nanchang University, Nanchang, Jiangxi 330031, P.R. China.,Reproductive Medicine Center, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Jun Tan
- Reproductive Medicine Center, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Yaoqing Wang
- Department of Gynecology and Obstetrics, Jiangxi Medical College, Graduate School of Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Jia Chen
- Department of Gynecology and Obstetrics, Jiangxi Medical College, Graduate School of Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Leizhen Xia
- Reproductive Medicine Center, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Lixian Xu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Ge Chen
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Qiongfang Wu
- Reproductive Medicine Center, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Ouping Huang
- Department of Gynecology and Obstetrics, Jiangxi Medical College, Graduate School of Nanchang University, Nanchang, Jiangxi 330031, P.R. China.,Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
38
|
Hou E, Zhao Y, Hang J, Qiao J. Metabolomics and correlation network analysis of follicular fluid reveals associations between l-tryptophan, l-tyrosine and polycystic ovary syndrome. Biomed Chromatogr 2020; 35:e4993. [PMID: 32986877 DOI: 10.1002/bmc.4993] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 12/24/2022]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine and metabolic disorder in women of reproductive age. Some studies have investigated metabolic alterations in plasma and follicular fluid from PCOS patients, but they did not control for obesity or insulin resistance (IR); additionally, correlation analysis of metabolites is sparse. Accordingly, in this study, we aimed to examine metabolic differences owing to the pathogenesis of PCOS, identify the hub metabolites and investigate its associations with androgens. We applied GC-MS platform coupled with a correlation network approach to analyze follicular fluid samples from 32 PCOS patients without obesity and IR and 31 healthy women. Thirty significantly altered metabolites in PCOS patients were enriched in amino acid metabolism. l-Phenylalanine, l-tryptophan, pyroglutamic acid, l-tyrosine, l-leucine and l-valine were screened as hub metabolites in metabolic correlation network. Among them, increased l-tryptophan and l-tyrosine were altered hub metabolites, and they had a more significant impact on the metabolic change of PCOS. In addition, l-tryptophan and l-tyrosine were significantly positively associated with serum androgens levels in the PCOS. Our results suggest that disorders of amino acid metabolism, especially tryptophan and tyrosine metabolism, might play an important role in the development of PCOS in predisposed women without obesity and IR.
Collapse
Affiliation(s)
- Entai Hou
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction (Peking University Third Hospital), Beijing, China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction (Peking University Third Hospital), Beijing, China
| | - Jing Hang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction (Peking University Third Hospital), Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction (Peking University Third Hospital), Beijing, China
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an overview of androgen supplementation in ART with the most updated evidence, from animal studies to its clinical applications in poor ovarian responders (POR) and the future studies to be published. RECENT FINDINGS Animal studies, has shown that testosterone supplementation, can be an option to increase the recruitable follicular pool in POR. However, the potential mechanism of action, dose, and duration of treatment is still under investigation. Early studies in humans reported promising results in favor of androgens [dehydroepiandrosterone (DHEA) or testosterone] in POR. Nevertheless, recent evidence does not appear to follow the initial results, whereas the type, dose, and duration of testosterone administration appear to be crucial for treatment effect. SUMMARY Testosterone seems to play an essential role in regulating ovarian function. However, it is worrisome that androgens are used off-label, despite that the available evidence is weak. Although testosterone supplementation may be beneficial in POR, published studies have used inconsistent doses and duration of administration. An ongoing trial (T-TRANSPORT trial) for the first time aims to provide conclusive evidence on whether transdermal testosterone administration can improve the reproductive outcomes in patients undergoing IVF/ICSI.
Collapse
|
40
|
Elucidating the roles of naturally occurring silent mutations in Polycystic Ovary Syndrome (PCOS). Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
41
|
Cox MJ, Edwards MC, Rodriguez Paris V, Aflatounian A, Ledger WL, Gilchrist RB, Padmanabhan V, Handelsman DJ, Walters KA. Androgen Action in Adipose Tissue and the Brain are Key Mediators in the Development of PCOS Traits in a Mouse Model. Endocrinology 2020; 161:bqaa061. [PMID: 32301482 DOI: 10.1210/endocr/bqaa061] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a complex disorder characterized by endocrine, reproductive, and metabolic abnormalities. Despite PCOS being the most common endocrinopathy affecting women of reproductive age, the etiology of PCOS is poorly understood, so there is no cure and symptomatic treatment is suboptimal. Hyperandrogenism is the most consistent feature observed in PCOS patients, and recently aberrant neuroendocrine signaling and adipose tissue function have been proposed as playing a role in the development of PCOS. To investigate the role of adipose tissue and the brain as key sites for androgen receptor (AR)-mediated development of PCOS, we combined a white and brown adipose and brain-specific AR knockout (AdBARKO) mouse model with a dihydrotestosterone (DHT)-induced mouse model of PCOS. As expected, in wildtype (WT) control females, DHT exposure induced the reproductive PCOS traits of cycle irregularity, ovulatory dysfunction, and reduced follicle health, whereas in AdBARKO females, DHT did not produce the reproductive features of PCOS. The metabolic PCOS characteristics of increased adiposity, adipocyte hypertrophy, and hepatic steatosis induced by DHT in WT females were not evident in DHT-treated AdBARKO females, which displayed normal white adipose tissue weight and no adipocyte hypertrophy or liver steatosis. Dihydrotestosterone treatment induced increased fasting glucose levels in both WT and AdBARKO females. These findings demonstrate that adipose tissue and the brain are key loci of androgen-mediated actions involved in the developmental origins of PCOS. These data support targeting adipocyte and neuroendocrine AR-driven pathways in the future development of novel therapeutic strategies for PCOS.
Collapse
Affiliation(s)
- Madeleine J Cox
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, Australia
| | - Melissa C Edwards
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Valentina Rodriguez Paris
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, Australia
| | - Ali Aflatounian
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, Australia
| | - William L Ledger
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, Australia
| | - Robert B Gilchrist
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, Australia
| | | | - David J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Kirsty A Walters
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
42
|
Gao XY, Liu Y, Lv Y, Huang T, Lu G, Liu HB, Zhao SG. Role of Androgen Receptor for Reconsidering the "True" Polycystic Ovarian Morphology in PCOS. Sci Rep 2020; 10:8993. [PMID: 32488141 PMCID: PMC7265442 DOI: 10.1038/s41598-020-65890-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/12/2020] [Indexed: 11/21/2022] Open
Abstract
Purpose: Polycystic ovarian morphology (PCOM) is one of the key features of polycystic ovary syndrome (PCOS). The diagnosis of PCOM according to the Rotterdam criteria (≥12 antral follicles per ovary) is debated because of the high prevalence of PCOM in the general population. Androgen receptor (AR) is associated with the PCOS phenotype and might as well play a role during folliculogenesis. This study is aimed to investigate the expression of the AR in PCOS granulosa cells (GCs) and its relationship with the PCOM phenotype. Methods: 106 PCOS cases and 63 controls were included from the Center for Reproductive Medicine, Shandong University. The diagnosis of PCOS was following the Rotterdam criteria (2003). Total RNA was extracted from GCs retrieved from ovarian stimulation. The expression of AR was amplified by means of quantitative real-time polymerase chain reaction. Results: The AR expression was significantly decreased in PCOS cases, especially in the tPCOM subgroup (≥20 antral follicles per ovary). Correlation analyses showed that AR expression was significantly correlated with serum FSH levels in controls and non-tPCOM. In the tPCOM subgroup, the AR expression was significantly correlated with serum LH levels. Interestingly, the significance of these correlations gradually disappeared as the threshold of antral follicles increased above 24 for PCOM. Conclusions:AR was differently expressed in PCOS and especially in the tPCOM subtype. The correlation of AR expression with serum FSH and LH might be associated with the number of follicles in PCOM.
Collapse
Affiliation(s)
- Xue-Ying Gao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Yue Liu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Yue Lv
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Tao Huang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hong-Bin Liu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Shi-Gang Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
43
|
Abstract
Fertility rates in classic congenital adrenal hyperplasia caused by 21-hydroxylase deficiency are substantially decreased for various reasons, including hormonal, anatomic, psychosocial, and psychosexual causes. However, fecundity is comparable with the general population. Under optimal hormone replacement, the course and outcome of pregnancies is also good. This article summarizes successful gestational management, including preconceptional considerations, adjustment of hormone replacement during pregnancy, delivery and lactation, as well as the prevention of adrenal crises. In nonclassic 21-hydroxylase deficiency, preconceptional low-dose hydrocortisone replacement normalizes the otherwise increased miscarriage rate. Pregnancy reports in rarer forms of congenital adrenal hyperplasia are summarized as well.
Collapse
Affiliation(s)
- Nicole Reisch
- Medizinische Klinik IV, Department of Endocrinology, Klinikum der Universität München, Ziemssenstraße 1, München 80336, Germany.
| |
Collapse
|
44
|
Zhang B, Shen S, Gu T, Hong T, Liu J, Sun J, Wang H, Bi Y, Zhu D. Increased circulating conjugated primary bile acids are associated with hyperandrogenism in women with polycystic ovary syndrome. J Steroid Biochem Mol Biol 2019; 189:171-175. [PMID: 30849463 DOI: 10.1016/j.jsbmb.2019.03.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 02/25/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common metabolic disorder with hyperandrogenism as the core pathophysiologic feature. Increased bile acids and altered bile acid pool composition have been implicated in the pathogenesis of several metabolic diseases. Thus, this study aimed to clarify the specific alterations in circulating bile acids in patients with PCOS and investigate its associations with hyperandrogenism. We recruited 37 patients with PCOS and 35 age- and BMI-matched control subjects. Serum bile acids and androgen profiles were assessed by ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS). Compared with control subjects, glycine- and taurine-conjugated primary bile acids were significantly elevated in patients with PCOS (P = 0.001 and P < 0.001). In addition, these conjugated primary bile acids were significantly and positively associated with serum concentrations of total testosterone and androstenedione in the PCOS group. In conclusion, increased circulating conjugated primary bile acids are positively associated with hyperandrogenism in women with PCOS. Further studies are warranted to explore the underlying mechanism.
Collapse
Affiliation(s)
- Bingjie Zhang
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China
| | - Shanmei Shen
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China
| | - Tianwei Gu
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China
| | - Ting Hong
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China
| | - Jiayi Liu
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China
| | - Jie Sun
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China
| | - Yan Bi
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China.
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower hospital affiliated to Nanjing University Medical School, No 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
45
|
Duffy DM, Ko C, Jo M, Brannstrom M, Curry TE. Ovulation: Parallels With Inflammatory Processes. Endocr Rev 2019; 40:369-416. [PMID: 30496379 PMCID: PMC6405411 DOI: 10.1210/er.2018-00075] [Citation(s) in RCA: 264] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/18/2018] [Indexed: 12/14/2022]
Abstract
The midcycle surge of LH sets in motion interconnected networks of signaling cascades to bring about rupture of the follicle and release of the oocyte during ovulation. Many mediators of these LH-induced signaling cascades are associated with inflammation, leading to the postulate that ovulation is similar to an inflammatory response. First responders to the LH surge are granulosa and theca cells, which produce steroids, prostaglandins, chemokines, and cytokines, which are also mediators of inflammatory processes. These mediators, in turn, activate both nonimmune ovarian cells as well as resident immune cells within the ovary; additional immune cells are also attracted to the ovary. Collectively, these cells regulate proteolytic pathways to reorganize the follicular stroma, disrupt the granulosa cell basal lamina, and facilitate invasion of vascular endothelial cells. LH-induced mediators initiate cumulus expansion and cumulus oocyte complex detachment, whereas the follicular apex undergoes extensive extracellular matrix remodeling and a loss of the surface epithelium. The remainder of the follicle undergoes rapid angiogenesis and functional differentiation of granulosa and theca cells. Ultimately, these functional and structural changes culminate in follicular rupture and oocyte release. Throughout the ovulatory process, the importance of inflammatory responses is highlighted by the commonalities and similarities between many of these events associated with ovulation and inflammation. However, ovulation includes processes that are distinct from inflammation, such as regulation of steroid action, oocyte maturation, and the eventual release of the oocyte. This review focuses on the commonalities between inflammatory responses and the process of ovulation.
Collapse
Affiliation(s)
- Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - CheMyong Ko
- Department of Comparative Biosciences, University of Illinois Urbana Champaign, Urbana, Illinois
| | - Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, Kentucky
| | - Mats Brannstrom
- Department of Obstetrics and Gynecology, University of Gothenburg, Gothenburg, Sweden.,Stockholm IVF, Stockholm, Sweden
| | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
46
|
Bertoldo MJ, Caldwell ASL, Riepsamen AH, Lin D, Gonzalez MB, Robker RL, Ledger WL, Gilchrist RB, Handelsman DJ, Walters KA. A Hyperandrogenic Environment Causes Intrinsic Defects That Are Detrimental to Follicular Dynamics in a PCOS Mouse Model. Endocrinology 2019; 160:699-715. [PMID: 30657917 DOI: 10.1210/en.2018-00966] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common cause of female infertility. Hyperandrogenism is both a major symptom and key diagnostic trait of PCOS; however, the direct impact of this androgen excess on ovarian dynamics is unclear. By combining a DHT-induced PCOS mouse model with an ex vivo follicle culture system, we investigated the impact of hyperandrogenism on ovarian function. Ovaries from PCOS mice exhibited the characteristic polycystic ovary morphology with numerous large cystic follicles and no corpora lutea present. Isolation and individual culture of preantral and antral follicles from PCOS mice resulted in slower growth rates during 5 days compared with the follicles isolated from control mice (P < 0.01). In contrast, preovulatory follicles from PCOS mice exhibited a significant increase in growth rate compared with controls (P < 0.01). Preantral follicles from PCOS ovaries maintained comparable follicular health as control follicles, but antral and preovulatory PCOS follicles exhibited reduced follicle health (P < 0.01) and survival rates (P < 0.01). Compared with controls, PCOS females also exhibited a poorer response to hyperstimulation (P < 0.01), impaired oocyte function evident by increased levels of reactive oxygen species (P < 0.01), and a reduction in on-time embryo development (P < 0.01). These results demonstrate that prolonged exposure to androgen excess leads to aberrant follicle development, which persists even after removal from the hyperandrogenic environment, causing perturbed follicular developmental trajectories. These findings indicate that an in vivo hyperandrogenic environment in patients with PCOS may intrinsically induce detrimental effects on follicles and oocytes.
Collapse
Affiliation(s)
- Michael J Bertoldo
- Fertility & Research Centre, School of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Aimee S L Caldwell
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Angelique H Riepsamen
- Fertility & Research Centre, School of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Dianliang Lin
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
- Fujian Provincial Reproductive Medicine Center, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Macarena B Gonzalez
- School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Rebecca L Robker
- School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - William L Ledger
- Fertility & Research Centre, School of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Robert B Gilchrist
- Fertility & Research Centre, School of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - David J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Kirsty A Walters
- Fertility & Research Centre, School of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
47
|
Gomes LG, Bachega TA, Mendonca BB. Classic congenital adrenal hyperplasia and its impact on reproduction. Fertil Steril 2019; 111:7-12. [DOI: 10.1016/j.fertnstert.2018.11.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/25/2018] [Accepted: 11/27/2018] [Indexed: 01/30/2023]
|
48
|
Ignatiuk VM, Izvolskaya MS, Sharova VS, Voronova SN, Zakharova LA. Disruptions in the reproductive system of female rats after prenatal lipopolysaccharide-induced immunological stress: role of sex steroids. Stress 2019; 22:133-141. [PMID: 30369279 DOI: 10.1080/10253890.2018.1508440] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Stress signals during fetal or early postnatal periods may disorganize reproductive axis development at different levels. This study was aimed to test the hypothesis that prenatal immunological stress induced by bacterial endotoxin, lipopolysaccharide (LPS), has impact on structure and function of the reproductive system in female offspring. Adult female Wistar rats were divided into two groups, a control group (n = 5) and a LPS group (n = 12). Rats were injected with LPS 50 μg/kg body or 0.9% saline intraperitoneally on the 12th day of pregnancy. After birth the female pups (n = 20 in each group) were divided into four groups: (group 1) 0.9% saline prenatally, sesame oil (vehicle) postnatally; (group 2) LPS prenatally, sesame oil postnatally; (group 3) LPS prenatally, fulvestrant postnatally; (group 4) LPS prenatally, flutamide postnatally. Pups were injected subcutaneously into the neck with fulvestrant (estrogen receptor antagonist), 1.5 mg/kg in sesame oil, from postnatal day (PND) 5 to PND14; or flutamide (androgen receptor antagonist), 20 mg/kg in sesame oil, from PND14 to PND30. Rats of the control group were injected with sesame oil during the same time period. Parameters were evaluated by ELISA (serum estradiol and testosterone) and ovarian histology. The main findings were: (1) prenatal stress during the critical period resulted in delayed vaginal opening, decreased body weight and serum concentrations of sex steroids, and significant disorders in ovarian development; (2) postnatal estradiol and testosterone antagonist treatments decreased follicular atresia through increasing the number of healthy follicles and restored endogenous steroid production. Lay summaryImmunological stress, caused by simulating infection through exposure to a bacterial toxin (LPS), during a critical period of fetal development in laboratory rats results in delayed reproductive maturity, decreased body weight and decreased secretion of sex steroids in female offspring, and abnormalities in the ovaries like those in polycystic ovarian syndrome. These prenatally toxin-induced sexual disorders in females could be corrected by estradiol/testosterone antagonists during the postnatal period.
Collapse
Affiliation(s)
- V M Ignatiuk
- a Moscow State University GSP-1 , Moscow , Russia
| | - M S Izvolskaya
- b Koltsov Institute of Developmental Biology , Russian Academy of Sciences , Moscow , Russia
| | - V S Sharova
- b Koltsov Institute of Developmental Biology , Russian Academy of Sciences , Moscow , Russia
| | - S N Voronova
- b Koltsov Institute of Developmental Biology , Russian Academy of Sciences , Moscow , Russia
| | - L A Zakharova
- b Koltsov Institute of Developmental Biology , Russian Academy of Sciences , Moscow , Russia
| |
Collapse
|
49
|
Puttabyatappa M, Padmanabhan V. Ovarian and Extra-Ovarian Mediators in the Development of Polycystic Ovary Syndrome. J Mol Endocrinol 2018; 61:R161-R184. [PMID: 29941488 PMCID: PMC6192837 DOI: 10.1530/jme-18-0079] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/14/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous endocrine disorder affecting women of reproductive age. The origin of PCOS is still not clear and appears to be a function of gene x environment interactions. This review addresses the current knowledge of the genetic and developmental contributions to the etiology of PCOS, the ovarian and extra-ovarian mediators of PCOS and the gaps and key challenges that need to be addressed in the diagnosis, treatment and prevention of PCOS.
Collapse
|
50
|
Affiliation(s)
- Douglas A Gibson
- Centre for Inflammatory Research, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France, Crescent, Edinburgh, EH16 4TJ, UK.
| | - Philippa T K Saunders
- Centre for Inflammatory Research, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France, Crescent, Edinburgh, EH16 4TJ, UK.
| | - Iain J McEwan
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|