Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
J Ovarian Res ; 16(1): 207, 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37880784

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is the most common endocrinopathy associated with infertility and metabolic disorder in women of reproductive age. Animal models have been developed and used as tools to unravel the pathogenesis of PCOS, among which most postnatal models employ continuing experimental manipulations. However, the persistence and stability of these animals after modeling is unknown. Dehydroepiandrosterone (DHEA)-induced PCOS mouse model is commonly used in PCOS studies. Thus the aim of the present study was to investigate the reproductive features of DHEA-induced PCOS mice fed a normal chow or an high-fat diet (HFD) with treatment withdrawal or consecutive treatments after PCOS mouse models were established. METHODS: Prepubertal C57BL/6 J mice (age 25 days) were injected (s.c.) daily with DHEA on a normal chow or a 60% HFD for 20 consecutive days to induce PCOS mouse models. Mice injected with the vehicle sesame oil were used as controls. After 20 days, mice were divided into 2 groups, namely "Continue dosing group" and "Stop dosing group". The animals were consecutively treated with DHEA or DHEA + HFD, or housed without any treatment for 2 or 4 weeks. Estrous cycles were evaluated during this period. At the end of the experiment, serum testosterone (T) levels were measured and the morphology of ovaries was evaluated. RESULTS: The mice in Continue dosing groups maintained reproductive phenotypes of PCOS mouse models. In contrast, 2 or 4 weeks after PCOS models were established, the mice with treatment withdrawal in Stop dosing groups exhibited normal serum testosterone levels, regular estrous cycle, and relatively normal ovarian morphology. In addition, even with consecutive treatments, there was no marked difference in body weight between DHEA mice on the normal chow or an HFD in Continue dosing groups and the control animals 3 weeks after modeling. CONCLUSIONS: After PCOS mice were induced with DHEA or DHEA + HFD, the mice still need consecutive treatments to maintain reproductive phenotypes to be regarded as PCOS mice that meet the diagnostic criteria of PCOS defined by the 2003 Rotterdam criteria.


Subject(s)
Polycystic Ovary Syndrome , Humans , Female , Mice , Animals , Infant, Newborn , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Mice, Inbred C57BL , Phenotype , Disease Models, Animal , Testosterone , Dehydroepiandrosterone/adverse effects
2.
Obstet Gynecol ; 142(3): 555-570, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37543737

ABSTRACT

OBJECTIVE: To systematically review the literature and provide clinical practice guidelines regarding various nonestrogen therapies for treatment of genitourinary syndrome of menopause (GSM). DATA SOURCES: MEDLINE, EMBASE, ClinicalTrials.gov , and Cochrane databases were searched from inception to July 2021. We included comparative and noncomparative studies. Interventions and comparators were limited to seven products that are commercially available and currently in use (vaginal dehydroepiandrosterone [DHEA], ospemifene, laser or energy-based therapies, polycarbophil-based vaginal moisturizer, Tibolone, vaginal hyaluronic acid, testosterone). Topical estrogen, placebo, other nonestrogen products, as well as no treatment were considered as comparators. METHODS OF STUDY SELECTION: We double-screened 9,131 abstracts and identified 136 studies that met our criteria. Studies were assessed for quality and strength of evidence by the systematic review group. TABULATION, INTEGRATION, AND RESULTS: Information regarding the participants, details on the intervention and comparator and outcomes were extracted from the eligible studies. Alternative therapies were similar or superior to estrogen or placebo with minimal increase in adverse events. Dose response was noted with vaginal DHEA and testosterone. Vaginal DHEA, ospemifene, erbium and fractional carbon dioxide (CO 2 ) laser, polycarbophil-based vaginal moisturizer, tibolone, hyaluronic acid, and testosterone all improved subjective and objective signs of atrophy. Vaginal DHEA, ospemifene, tibolone, fractional CO 2 laser, polycarbophil-based vaginal moisturizer, and testosterone improved sexual function. CONCLUSION: Most nonestrogen therapies are effective treatments for the various symptoms of GSM. There are insufficient data to compare nonestrogen options to each other.


Subject(s)
Hyaluronic Acid , Menopause , Female , Humans , Hyaluronic Acid/therapeutic use , Hyaluronic Acid/pharmacology , Vagina , Estrogens/therapeutic use , Testosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Dehydroepiandrosterone/adverse effects
3.
BMC Complement Med Ther ; 23(1): 102, 2023 Apr 03.
Article in English | MEDLINE | ID: mdl-37013510

ABSTRACT

BACKGROUND: One of the novel mechanisms in the pathogenesis of Polycystic ovary syndrome (PCOS) is low-grade chronic inflammation. Chamomile (Matricaria recutita L.) and Nettle (Urtica dioica), with phytoestrogenic and antioxidant properties, are traditionally used to treat gynecological diseases. This study investigated the immune-modulating effects of these two plants. METHODS: Following the induction of PCOS by subcutaneous injection (SC) of Dehydroepiandrosterone (DHEA) in BALB / C mice. Mice were treated in five groups: Sham, PCOS, PCOS + Chamomile, PCOS + Nettle, and PCOS + Chamomile and Nettle for 21 days. Ovarian morphology, blood antioxidant capacity, the abundance of Treg cells, and expression of matrix metalloproteinase-9 (MMP-9), transforming growth factor-ß (TGF-ß), cyclooxygenase-2 genes (COX-2), and tumor necrosis factor-alpha (TNF-α) were measured. RESULTS: Folliculogenesis, Cystic follicles, and corpus luteum improved in the treatment groups (P < 0. 05). Treg cells in the DHEA group were significantly reduced compared to the Sham group (P < 0. 01). However, this decrease was not corrected in treatment groups (P > 0. 05). Total serum antioxidant capacity was significantly increased in the treatment group of Nettle and Chamomile + Nettle (P < 0. 05). The expression of MMP9 and TGFß genes in the PCOS group was significantly higher than the Sham group (P < 0. 05), which the expression of MMP9 was corrected by treatment with Chamomile + Nettle extract (P < 0. 05). CONCLUSION: Chamomile and Nettle extract may be an effective supplement in improving the histological and immunological changes of PCOS. However, more research is needed to confirm its effectiveness in humans.


Subject(s)
Polycystic Ovary Syndrome , Urtica dioica , Female , Humans , Mice , Animals , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Matrix Metalloproteinase 9 , Antioxidants/pharmacology , Chamomile , Dehydroepiandrosterone/adverse effects
4.
Drug Dev Res ; 84(2): 226-237, 2023 04.
Article in English | MEDLINE | ID: mdl-36621953

ABSTRACT

This study was implemented to address the role of Roflumilast in polycystic ovary syndrome (PCOS) as well as to discuss its reaction mechanism in vivo and in vitro. In vivo, mice were administrated with 6 mg dehydroepiandrosterone (DHEA) per 100 g body weight and fed with 60% high fat diet to induce PCOS. The expression of phosphodiesterases 4 (PDE4) was assessed with RT-qPCR. The ovary pathology was observed by hematoxylin and eosin staining and follicles were counted. Enzyme-linked immunosorbent assay was adopted for the estimation of progesterone, testosterone and inflammatory factors and lipid accumulation was observed by Oil Red O staining. With the application of reverse transcription-quantitative PCR (RT-qPCR) and western blot, the messenger RNA (mRNA) and protein expressions of low-density lipoprotein receptor (LDLR) was resolved. In vitro, Cell counting kit-8 and flow cytometry analysis were applied for the assessment of cell proliferation and apoptosis. The proliferation- and apoptosis-related proteins were appraised with western blot. Additionally, the expressions of PDE-4 at both mRNA and protein levels were tested with RT-qPCR and western blot. Here, it was discovered that PDE4 was greatly elevated in PCOS mice and DHEA-induced ovarian granulosa cells (KGN). In PCOS mice, PDE4 was negative correlated with progesterone and had positive correlation with testosterone. Roflumilast could enhanced progesterone expression, increased the number of primary follicles, preantral follicles and antral follicles but reduced testosterone and decreased the number of cystic follicles in PCOS mice. It was also testified that Roflumilast could inhibit the release of inflammatory factors and lipid accumulation in PCOS mice. Besides, the proliferation of DHEA-induced KGN cells was enhanced while the apoptosis was declined by Roflumilast, accompanied by elevated contents of PCNA, Ki67 and antiapoptotic protein Bcl-2. Collectively, Roflumilast inhibited inflammation and lipid accumulation in PCOS mice to improve ovarian function and reduce DHEA-induced granulosa cell apoptosis.


Subject(s)
Phosphodiesterase 4 Inhibitors , Polycystic Ovary Syndrome , Humans , Female , Mice , Animals , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Progesterone/adverse effects , Progesterone/metabolism , Phosphodiesterase 4 Inhibitors/adverse effects , Granulosa Cells/metabolism , Granulosa Cells/pathology , Testosterone/adverse effects , Testosterone/metabolism , Inflammation/metabolism , Apoptosis , Dehydroepiandrosterone/adverse effects , Dehydroepiandrosterone/metabolism , Lipids
5.
Br J Pharmacol ; 179(18): 4563-4574, 2022 09.
Article in English | MEDLINE | ID: mdl-35751868

ABSTRACT

BACKGROUND AND PURPOSE: Polycystic ovary syndrome (PCOS) is a common metabolic and endocrine disease affecting women of reproductive age. Due to its complex aetiology, there is no currently effective cure for PCOS. Brown adipose tissue (BAT) activity is significantly decreased in PCOS patients, and BAT activation has beneficial effects in animal models of PCOS. Here, we investigated the effect of ginsenoside compound K (CK) in an animal model of PCOS and its mechanism of BAT activation. EXPERIMENTAL APPROACH: Primary brown adipocytes, Db/Db mice and dehydroepiandrosterone (DHEA)-induced PCOS rats were used. The core body temperature, oxygen consumption, energy metabolism related gene and protein expression were assessed to identify the effect of CK on overall energy metabolism. Oestrous cycle, serum sex hormone, ovarian steroidogenic enzyme gene expression and ovarian morphology were also evaluated following CK treatment. KEY RESULTS: Our results indicated that CK treatment could significantly protect against body weight gain in Db/Db mice via BAT activation. Furthermore, we found that CK treatment could normalize hyperandrogenism, oestrous cyclicity, normalize steroidogenic enzyme expression and decrease the number of cystic follicles in PCOS rats. Interestingly, as a potential endocrine intermediate, C-X-C motif chemokine ligand-14 protein (CXCL14) was significantly up-regulated following CK administration. In addition, exogenous CXC14 supplementation was found to reverse DHEA-induced PCOS in a phenotypically similar manner to CK treatment. CONCLUSION AND IMPLICATIONS: In summary, CK treatment significantly activates BAT, increases CXCL14 expression and ameliorates PCOS. These findings suggest that CK might be a potential drug candidate for PCOS treatment.


Subject(s)
Ginsenosides , Polycystic Ovary Syndrome , Adipose Tissue, Brown/metabolism , Animals , Dehydroepiandrosterone/adverse effects , Disease Models, Animal , Female , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Humans , Mice , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Rats
6.
Mol Cell Endocrinol ; 550: 111645, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35413388

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder in reproductive-aged women. In this study, a rat model of PCOS was established by subcutaneous injection of dehydroepiandrosterone (DHEA). NOX4 was highly expressed in PCOS rat ovaries, while its specific role in PCOS remains unclear. Lentivirus-mediated shRNA targeting NOX4 inhibited oxidative stress by reducing ROS, 4-HNE and MDA levels, and increasing SOD and GPX activities in rat ovaries. NOX4 deficiency increased Bcl-2 levels and decreased Bax, cleaved caspase-3 and cleaved caspase-9 levels and DHEA-induced cell apoptosis in rat ovaries. Similar to the in vivo results, NOX4 silencing inhibited oxidative stress and cell apoptosis in DHEA-treated rat granulosa cells. Moreover, NOX4 silencing promoted Nrf-2 translocation, and the expression of Nrf-2 and HO-1 both in vivo and in vitro. Thus, NOX4 deficiency may ameliorate PCOS in rats by reducing oxidative stress and cell apoptosis via activating the Nrf-2/HO-1 signal pathway.


Subject(s)
Polycystic Ovary Syndrome , Adult , Animals , Apoptosis , Dehydroepiandrosterone/adverse effects , Female , Humans , NADP/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Oxidative Stress , Polycystic Ovary Syndrome/metabolism , Rats , Signal Transduction/genetics
7.
J Clin Endocrinol Metab ; 107(6): 1679-1685, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35254428

ABSTRACT

CONTEXT: Androgen prohormones such as dehydroepiandrosterone (DHEA) increase in early puberty, peak in the second and third decade, and thereafter decline, independent of menopausal status. Investigators have examined their potential beneficial effects in normal women and those with DHEA-deficient states. EVIDENCE ACQUISITION: A review of the literature from 1985 to 2021 on the potential benefits and risks of androgen prohormones in women. EVIDENCE SYNTHESIS: Studies have examined the potential benefit of DHEA therapy for anti-aging, sexual dysfunction, infertility, metabolic bone health, cognition, and wellbeing in hormone-deficient states such as primary adrenal insufficiency, hypopituitarism, and anorexia as well as administration to normal women across the lifespan. CONCLUSIONS: Data support small benefits in quality of life and mood but not for anxiety or sexual function in women with primary or secondary adrenal insufficiency or anorexia. No consistent beneficial effects of DHEA administration have been observed for menopausal symptoms, sexual function, cognition, or overall wellbeing in normal women. Local administration of DHEA shows benefit in vulvovaginal atrophy. Use of DHEA to improve induction of ovulation response in women with diminished ovarian reserve is not recommended. Risks of high physiologic or pharmacologic use of DHEA include androgenic and estrogenic side effects which are of concern for long-term administration. CLINICAL CASE: A 49-year-old woman with Addison's disease who is on low dose estrogen with cyclic progesterone therapy for menopausal symptoms returns for follow-up. She is on a stable glucocorticoid replacement strategy of hydrocortisone 10 mg in the morning and 5 mg in the early afternoon and fludrocortisone 0.05 mg each morning. She has read on the internet that additional therapy with DHEA may help her overall quality of life and libido. She asks whether she should add this therapy to her regimen and at what dose.


Subject(s)
Androgens , Dehydroepiandrosterone , Anorexia/chemically induced , Anorexia/drug therapy , Dehydroepiandrosterone/adverse effects , Estrogens , Female , Hormone Replacement Therapy/adverse effects , Humans , Middle Aged , Quality of Life
8.
Int Immunopharmacol ; 107: 108717, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35334358

ABSTRACT

Gut microbiota dysbiosis is critical in the etiology of polycystic ovary syndrome (PCOS). However, the mechanisms of gut microbiota in PCOS pathogenesis have not been fully elucidated. We aimed to explore the role of gut microbiota-derived macrophage pyroptosis in PCOS. This study conducted dehydroepiandrosterone (DHEA) induced PCOS mice model, 16S rDNA sequencing, western blot, genetic knocking out, transcriptome and translatome profiling, et al. to evaluate the underlying mechanisms. 16S rDNA sequencing showed reduced gut Akkermansia and elevated gram-negative bacteria (Desulfovibrio and Burkholderia) abundances in DHEA induced PCOS mice, which was accompanied by increased serum lipopolysaccharide (LPS). LPS could induce macrophage pyroptosis in mice ovaries, also activated in PCOS. Gasdermin D (GSDMD) is the final executor of macrophage pyroptosis. We demonstrated that Gsdmd knockout in mice could dramatically ameliorate PCOS. Mechanistically, transcriptome and translatome profiling revealed that macrophage pyroptosis disrupted estrogen production and promoted apoptosis of granulosa cells. Interferon (IFN)-γ, which was elevated in PCOS mice serum and ovaries, enhanced macrophage pyroptosis and exacerbated its effect on estrogen receptor in granulosa cells. Inspiringly, we identified that disulfiram and metformin could augment gut Akkermansia abundance, reduce serum IFN-γ level, inhibit macrophage pyroptosis in ovaries, therefore ameliorating PCOS. Collectively, this study emphasizes that macrophage pyroptosis, which was induced by gut microbiota dysbiosis and enhanced by IFN-γ, plays a key role in PCOS pathogenesis through estrogen synthesis dysfunction and apoptosis of granulosa cells. Disulfiram and metformin, which enhanced gut Akkermansia abundance and suppressed macrophage pyroptosis, may be considered as potential therapeutic strategies for PCOS.


Subject(s)
Gastrointestinal Microbiome , Metformin , Polycystic Ovary Syndrome , Animals , Apoptosis , DNA, Ribosomal/pharmacology , Dehydroepiandrosterone/adverse effects , Disulfiram/adverse effects , Dysbiosis/microbiology , Estrogens/pharmacology , Female , Gastrointestinal Microbiome/physiology , Granulosa Cells/pathology , Humans , Lipopolysaccharides/pharmacology , Macrophages/pathology , Metformin/pharmacology , Mice , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/drug therapy , Pyroptosis
9.
Ann Med ; 54(1): 578-587, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35152800

ABSTRACT

BACKGROUND: Polycystic ovarian syndrome (PCOS) is one of the major causes encouraging the elevation of androgens, obesity along with menstrual complications. Here we study the effect of Apigenin in rat model of polycystic ovarian syndrome. METHODS: Female Sprague Dawley (SD) rats were treated with Dehydroepiandrosterone (DHEA) (6 mg/100g) opting the post-pubertal approach for developing rat model of polycystic ovarian syndrome, Metformin was used as standard. The treatments were given for 21 days along with coloproctological analysis. After the treatment regimen, the biochemical analysis was carried in plasma samples, whereas the ovaries were submitted for histopathological analysis. RESULTS: The treatment of DHEA resulted in disturbed lipid profile and anti-oxidant status along with increased weight, ovarian diameter and cysts in rats confirming the development of PCOS. However, treatment of Apigenin showed ameliorative effect by improving the lipid profile and anti-oxidant status, the treatment also normalised the body weight, reduced ovarian diameter, cysts and restored the healthy follicles compared to control rats. The treatment of Apigenin also suppressed the levels of oestradiol and testosterone compared to control group, also, levels of progesterone were increased in Apigenin treated group of rats. The treatment of Apigenin suppressed the levels of inflammatory cytokines TNF-α and IL-6. It was observed that the effect of Apigenin were to some extent parallel to standard drug Metformin. CONCLUSION: The findings confirmed that Apigenin ameliorates the disturbed hormonal levels, lipid profile and antioxidant status in PCOS rats.


Subject(s)
Polycystic Ovary Syndrome , Animals , Apigenin/adverse effects , Dehydroepiandrosterone/adverse effects , Disease Models, Animal , Female , Humans , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Rats , Rats, Sprague-Dawley
10.
Mol Reprod Dev ; 88(12): 817-829, 2021 12.
Article in English | MEDLINE | ID: mdl-34658106

ABSTRACT

The possible relationship between dehydroepiandrosterone (DHEA)-induced polycystic ovary syndrome (PCOS) and epigenetic changes (ECs) leading to the impaired oocyte quality, has not been investigated yet. So, this study aimed to provide an insight into the relationship of the impaired oocyte quality with ECs in a mice DHEA-induced PCOS model and to further reveal the effect of metformin treatment. For this purpose, 80 female BALB/C mice were randomly divided into four equal groups, named as the control, sham, (DHEA) and DHEA + Metformin groups. The alterations in acetylation of H4K5 and H4K16, and in methylation of DNA (5MeC) and H3K9 were evaluated using immunocytochemical. Moreover, the expression of Hdac1, Hdac2, Dnmt1, and Dnmt3a genes involved in ECs were analyzed using reverse-transcription polymerase chain reaction. As well, the levels of mitochondrial membrane potential (MMP), oxidative stress (OS), embryo development, ovarian morphology, sexual hormone, ovulatory function, and AMPKα phosphorylation activity were compared in all the studied groups. Metformin attenuated the damages induced by DHEA as indicated by the normalized the estrous cycle, the improved ovarian morphology, the decreased sexual hormone and OS levels, and the increased MMP and AMPKα phosphorylation levels. In the metformin group, the Dnmt1, Dnmt3a, and Hdac2 genes have significantly upregulated compared to the DHEA group. However, metformin was found to have no effect on the expression level of Hdac1. In this regard, significant decrease and increase were observed in both the acetylated H4K16 and methylated H3K9 within MII oocytes in the DHEA + Metformin group compared with the DHEA group. Our results show that metformin could enhance the developmental competence of PCOS oocytes via reducing OS and ECs.


Subject(s)
Metformin , Polycystic Ovary Syndrome , Animals , Dehydroepiandrosterone/adverse effects , Embryonic Development , Epigenesis, Genetic , Female , Metformin/pharmacology , Mice , Mice, Inbred BALB C , Oocytes/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism
11.
J Ovarian Res ; 14(1): 64, 2021 May 04.
Article in English | MEDLINE | ID: mdl-33947426

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a multifactorial endocrinopathy that affects reproduction and metabolism. Mammalian target of rapamycin (mTOR) has been shown to participate in female reproduction under physiological and pathological conditions. This study aimed to investigate the role of mTOR complex 1 (mTORC1) signaling in dehydroepiandrosterone (DHEA)-induced PCOS mice. RESULTS: Female C57BL/6J mice were randomly assigned into three groups: control group, DHEA group, and DHEA + rapamycin group. All DHEA-treated mice were administered 6 mg/100 g DHEA for 21 consecutive days, and the DHEA + rapamycin group was intraperitoneally injected with 4 mg/kg rapamycin every other day for the last 14 days of the DHEA treatment. There was no obvious change in the expression of mTORC1 signaling in the ovaries of the control and DHEA groups. Rapamycin did not protect against DHEA-induced acyclicity and PCO morphology, but impeded follicle development and elevated serum testosterone levels in DHEA-induced mice, which was related with suppressed Hsd3b1, Cyp17a1, and Cyp19a1 expression. Moreover, rapamycin also exacerbated insulin resistance but relieved lipid metabolic disturbance in the short term. CONCLUSIONS: Rapamycin exacerbated reproductive imbalance in DHEA-induced PCOS mice, which characterized by elevated testosterone levels and suppressed steroid synthesis. This underscores the need for new mTORC1-specific and tissue-specific mTOR-related drugs for reproductive disorders.


Subject(s)
Dehydroepiandrosterone/adverse effects , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Reproduction/drug effects , Sirolimus/adverse effects , Testosterone/adverse effects , Animals , Disease Models, Animal , Female , Humans , Mice
12.
Endocr Relat Cancer ; 28(2): R47-R53, 2021 02.
Article in English | MEDLINE | ID: mdl-33263566

ABSTRACT

Androgens play a fundamental role in the morbidity and mortality of COVID-19, inducing both the ACE-2 receptor to which SARS-CoV-2 binds to gain entry into the cell, and TMPRS22, the transmembrane protease that primes the viral spike protein for efficient infection. The United States stands alone among developed nations in permitting one androgen, oral DHEA, to be freely available OTC and online as a 'dietary supplement'. DHEA is widely used by males in the US to offset the age-related decline in circulating androgens. This fact may contribute to the disparate statistics of COVID-19 morbidity and mortality in this country. In regulatory antithesis, every other developed nation regulates DHEA as a controlled substance. DHEA is an extremely potent inhibitor of glucose-6-phosphate dehydrogenase (G6PD), with uniquely unstable uncompetitive inhibition kinetics. This has particular relevance to COVID-19 because G6PD-deficient human cells have been demonstrated to be exceptionally sensitive to infection by human coronavirus. Because DHEA is lipophilic and freely passes into cells, oral DHEA bypasses the normal controls regulating androgen biology and uncompetitive G6PD inhibition. DHEA's status as a 'dietary supplement' means that no clinical trials demonstrating safety have been performed, and, in the absence of physician supervision, no data on adverse events have been collected. During the current pandemic, the unrestricted availability of oral DHEA as a 'dietary supplement' cannot be considered safe without proof from placebo-controlled clinical trials that it is not contributing to the severity of COVID-19. US physicians may therefore wish to query their patients' use of DHEA.


Subject(s)
COVID-19/metabolism , Dehydroepiandrosterone/adverse effects , Nonprescription Drugs/adverse effects , Androgens/metabolism , Animals , COVID-19/mortality , Dehydroepiandrosterone/administration & dosage , Female , Humans , Male , Nonprescription Drugs/administration & dosage , SARS-CoV-2/isolation & purification , Survival Rate , United States/epidemiology
13.
Pediatrics ; 146(3)2020 09.
Article in English | MEDLINE | ID: mdl-32868471

ABSTRACT

BACKGROUND: Legal performance-enhancing substance(s) (PES) (eg, creatine) are widely used among adolescent boys and young men; however, little is known about their temporal associations with substance use behaviors. METHODS: We analyzed prospective cohort data from the National Longitudinal Study of Adolescent to Adult Health, Waves I to IV (1994-2008). Logistic regressions were used to first assess adolescent substance use (Wave I) and use of legal PES (Wave III) and second to assess use of legal PES (Wave III) and subsequent substance use-associated risk behaviors (Wave IV), adjusting for potential confounders. RESULTS: Among the sample of 12 133 young adults aged 18 to 26 years, 16.1% of young men and 1.2% of young women reported using legal PES in the past year. Adolescent alcohol use was prospectively associated with legal PES use in young men (odds ratio 1.39; 95% confidence interval [CI] 1.13-1.70). Among young men, legal PES use was prospectively associated with higher odds of problematic alcohol use and drinking-related risk behaviors, including binge drinking (adjusted odds ratio [aOR] 1.35; 95% CI 1.07-1.71), injurious and risky behaviors (aOR 1.78; 95% CI 1.43-2.21), legal problems (aOR 1.52; 95% CI 1.08-2.13), cutting down on activities and socialization (aOR 1.91; 95% CI 1.36-2.78), and emotional or physical health problems (aOR 1.44; 95% CI 1.04-1.99). Among young women, legal PES use was prospectively associated with higher odds of emotional or physical health problems (aOR 3.00; 95% CI 1.20-7.44). CONCLUSIONS: Use of legal PES should be considered a gateway to future problematic alcohol use and drinking-related risk behaviors, particularly among young men.


Subject(s)
Performance-Enhancing Substances/adverse effects , Substance-Related Disorders/etiology , Adolescent , Adult , Alcohol Drinking/adverse effects , Alcoholic Intoxication/complications , Amino Acids/adverse effects , Athletes/statistics & numerical data , Body Mass Index , Confidence Intervals , Creatine/adverse effects , Dehydroepiandrosterone/adverse effects , Female , Humans , Logistic Models , Male , Marijuana Smoking/adverse effects , Odds Ratio , Prospective Studies , Risk-Taking , Sex Factors , Smoking/adverse effects , Steroids/adverse effects , Underage Drinking , Young Adult
14.
Mol Cell Endocrinol ; 518: 110973, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32781251

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with hyperandrogenemia and uterine abnormalities. Our aim was to investigate the uterine effects of PCOS that are mediated through the androgen receptor (AR). After weaning, female rats were treated with sesame oil (Control), dehydroepiandrosterone (DHEA), or DHEA + flutamide (FLU, an AR antagonist) for 20 consecutive days. On postnatal day 41, serum, ovarian and uterine tissues were collected. DHEA and DHEA + FLU rats showed increased testosterone levels. DHEA rats showed increased epithelial height, glandular density, subepithelial stroma and myometrial thickness, associated with decreased nuclei density. These rats also showed increased uterine water content, with decreased aquaporin (AQP) 3, 7 and 8 expression in the uterine epithelium and increased AQP8 expression in the myometrium. DHEA rats also showed decreased uterine collagen remodeling, decreased cell proliferation in the subepithelial stroma, and increased apoptosis in the luminal and glandular epithelium and in the myometrium. They also showed an increase in insulin-like growth factor-1 and a decrease in phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase. The uterine stroma of DHEA rats showed no changes in progesterone receptor or estrogen receptor alpha (ERα) and increased AR expression. DHEA + FLU rats showed a smaller increase in the myometrial thickness, an increase in the uterine water content without AQP8 induction and a smaller decrease in collagen remodeling. These rats also showed no apoptosis induction and decreased proliferation in the myometrium, decreased ERα in the subepithelial stroma and myometrium and no modifications in AR. Our results demonstrate that the uterine cell turnover and collagen remodeling in DHEA rats are regulated through AR, directly or indirectly associated with ERα expression.


Subject(s)
Dehydroepiandrosterone/adverse effects , Estrogen Receptor alpha/metabolism , Polycystic Ovary Syndrome/metabolism , Receptors, Androgen/metabolism , Uterus/pathology , Animals , Animals, Newborn , Aquaporins/metabolism , Collagen/metabolism , Disease Models, Animal , Female , Flutamide/pharmacology , Gene Expression Regulation/drug effects , Humans , Polycystic Ovary Syndrome/chemically induced , Rats , Testosterone/blood , Testosterone/metabolism , Uterus/drug effects , Uterus/metabolism
15.
Nutr Metab Cardiovasc Dis ; 30(9): 1465-1475, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32675010

ABSTRACT

BACKGROUND AND AIMS: Dehydroepiandrosterone (DHEA) supplementation has gained attention in individuals with adrenal insufficiency, and as a tool for increasing androgens and estrogens whereby is proposed to improve the accretion of muscle and bone mass. However, DHEA supplementation has demonstrated negative effects on the lipid profile and, thus, we aimed to analyze the body of evidence in this regard. METHODS AND RESULTS: A systematic review and dose-response meta-analysis of randomized controlled trials (RCTs) was performed employing in Scopus, PubMed/Medline, Web of Science, Embase and Google Scholar, then including relevant articles that addressed the effects of DHEA supplementation on the lipid profile, up to February 2020. Combined findings were generated from 23 eligible articles. Hence, total cholesterol (TC) (weighted mean difference (WMD): -3.5 mg/dl, 95% confidence interval (CI): -8.5 to 1.6)), low-density lipoprotein-cholesterol (LDL-C) (WMD: 0.34 mg/dl, 95% CI: -3 to 3.7) and triglycerides (TG) levels (WMD: -2.85 mg/dl, 95% CI: -9.3 to 3.6) did not alter in DHEA group compared to the control, but HDL-C levels significantly reduced in DHEA group (WMD: -3.1 mg/dl, 95% CI: -4.9 to -1.3). In addition, a significant reduction in HDL-C values was observed in studies comprising women (WMD: -5.1 mg/dl, 95% CI: -7.2 to -3) but not in males (WMD: 0.13 mg/dl, 95% CI: -1.4 to 1.7). CONCLUSIONS: Overall, supplementation with DHEA did not change circulating values of TC, LDL-C and TG, whereas it may decrease HDL-C levels. Further long-term RCTs are required to investigate the effects of DHEA particularly on major adverse cardiac events.


Subject(s)
Cardiovascular Diseases/prevention & control , Dehydroepiandrosterone/therapeutic use , Dietary Supplements , Dyslipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Lipids/blood , Adult , Aged , Biomarkers/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Dehydroepiandrosterone/adverse effects , Dietary Supplements/adverse effects , Dyslipidemias/blood , Dyslipidemias/diagnosis , Dyslipidemias/epidemiology , Female , Humans , Hypolipidemic Agents/adverse effects , Male , Middle Aged , Randomized Controlled Trials as Topic , Risk Factors , Treatment Outcome , Young Adult
16.
J Ovarian Res ; 13(1): 57, 2020 May 09.
Article in English | MEDLINE | ID: mdl-32386521

ABSTRACT

BACKGROUND: Previous studies have shown that chronic inflammation and oxidative stress may play an important role in the pathophysiology of polycystic ovary syndrome (PCOS), and glutamine (Gln) have showed the anti-inflammatory and antioxidant properties. So the aim of this study is to investigate the effect of glutamine supplementation on PCOS rats. METHODS: Female Sprague-Dawley rats were randomly assigned into four groups (n = 10 /group), control group, PCOS group, PCOS+ 0.5 g/kg Gln group and PCOS+ 1.0 g/kg Gln group. All the PCOS rats were administrated with 6 mg/100 g dehydroepiandrosterone (DHEA) for 20 consecutive days, all the PCOS+Gln groups were intraperitoneal injected glutamine twice in the next morning after the last DHEA injection. All the samples were collected 12 h after the last administration. Ovarian histological examinations were analyzed and the concentration of serum hormone, inflammatory and oxidative stress factors were measured. RESULTS: There was no obvious ovarian histological change among the PCOS group and PCOS+Gln groups. All the detected inflammation factors [C-reactive protein, interleukin (IL)-6, IL-18, tumor necrosis factor] showed significantly higher in all the PCOS groups compared to the control group (P < 0.01), and were significantly decreased with the supplementation of 0.5 g/kg glutamine (P < 0.01). Concentrations of superoxide dismutase were significantly lower in all the PCOS groups (P < 0.01) compared to the control group, and increased significantly with the supplementation of 0.5 g/kg glutamine (P < 0.01). Serum concentrations of malondialdehyde, nitric oxide synthase and nitric oxide were significantly higher in PCOS group (P < 0.01) compared with the control group, and significantly decreased to the comparative levels of control group with supplementation of 0.5 g/kg glutamine (P < 0.01). CONCLUSION: There is low-grade inflammation and oxidative stress in DHEA-induced PCOS rats. The supplementation of 0.5 g/kg glutamine could effectively ameliorate the inflammation and oxidative stress conditions of PCOS.


Subject(s)
Dehydroepiandrosterone/adverse effects , Glutamine/therapeutic use , Inflammation/drug therapy , Oxidative Stress/drug effects , Polycystic Ovary Syndrome/chemically induced , Animals , Female , Glutamine/pharmacology , Humans , Polycystic Ovary Syndrome/drug therapy , Rats , Rats, Sprague-Dawley
17.
JBRA Assist Reprod ; 24(1): 41-54, 2020 01 30.
Article in English | MEDLINE | ID: mdl-31608617

ABSTRACT

OBJECTIVES: Polycystic ovary syndrome (PCOS) represents 75% of the cases of anovulatory infertility. The aim of this study was to investigate the role of aspirin on dehydroepiandrosterone (DHEA) - induced polycystic ovary syndrome in Wistar rats. METHODS: Twenty eight (28) pre-pubertal female Wistar rats of 21 days old weighing 16 - 21 g were divided into 4 groups (7 rats/group) and treated as follows; group I received distilled water and served as Control; Group II received 6 mg/100 g body weight DHEA in 0.2 ml of oil subcutaneously to induce PCOS. Group III received 7.5 mg/kg of aspirin orally; Group IV received 6 mg/100kg of body weight of DHEA in 0.2ml of oil subcutaneously and 7.5 mg/kg of aspirin orally. After 15 days of administration, the rats were slaughtered by cervical dislocation. Blood samples and ovaries were collected for reproductive hormonal analysis, biochemical and histopathological analysis. The expressions of mRNA androgen receptor (AR) gene in the ovary were determined by real time reverse transcriptase polymerase chain reaction (qPCR). All the data was analyzed using one way ANOVA with the Graph pad prism software version 6. A p<0.05 was considered significant. RESULTS: The results obtained showed that dehydroepiandrosterone treatment caused significant decrease (p<0.05) in total protein, superoxide Dismutase (SOD), glutathione-s- transferase (GST), Ca2+ ATPase, and significant increase (p<0.05) in malondialdehyde, vascular endothelial growth factor, tumor necrosis factor and estrogen as compared to Controls. The group co-administered with DHEA and aspirin showed significant increases in SOD, GST, CAT, GSH, Progesterone, Ca2+ ATPase, Na+ ATPase, H+ ATPase and significant reduction (p<0.05) in malondialdehyde, VEGF, TNF-α and estrogen as compared with the DHEA group. The histopathological analysis showed reductions in cystic fibrosis, atretic ovaries, increased expression of Bcl-2 and E- Cadherin and reduced Bax expression in the group that received Aspirin and DHEA. CONCLUSION: This study clearly demonstrates that Aspirin has ameliorating effects against polycystic ovary syndrome via anti-inflammatory and hormonal modulatory pathways.


Subject(s)
Aspirin/pharmacology , Dehydroepiandrosterone/adverse effects , Ovary/drug effects , Polycystic Ovary Syndrome , Animals , Antioxidants/analysis , Antioxidants/metabolism , Cytokines/analysis , Cytokines/metabolism , Female , Ovary/cytology , Ovary/metabolism , Oxidative Stress/drug effects , Oxidoreductases/analysis , Oxidoreductases/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Rats , Rats, Wistar
18.
Trials ; 20(1): 50, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30646929

ABSTRACT

BACKGROUND: Women undergoing in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) with poor ovarian respond (POR) always have very low clinical pregnancy rates. In previous data, dehydroepiandrosterone (DHEA) was suggested as a promising treatment and maybe has a good pregnancy outcome. But there is no sufficient evidence from randomized clinical trials evaluating the effect of DHEA preconceptional treatment on live birth in POR. METHODS: This trial is a multicenter active-placebo double-blind clinical trial (1:1 treatment ratio of active versus placebo). The infertile POR patients undergoing IVF or ICSI will be enrolled and randomly assigned to two parallel groups. Participants in these two groups will be given 4-12 weeks' treatment of DHEA or placebo, respectively. The primary outcome is live birth rate. DISCUSSION: The results of this study will provide evidence for the effect of preconceptional DHEA treatment on IVF outcome in POR. TRIAL REGISTRATION: Chinese Clinical Trial Registry, ChiCTR-IPR-15006909 . Registered on November 9, 2015.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Fertility Agents, Female/administration & dosage , Fertilization in Vitro , Infertility, Female/therapy , Ovary/drug effects , Ovulation/drug effects , Preconception Care/methods , Dehydroepiandrosterone/adverse effects , Double-Blind Method , Embryo Culture Techniques , Embryo Transfer , Female , Fertility Agents, Female/adverse effects , Humans , Infertility, Female/diagnosis , Infertility, Female/physiopathology , Live Birth , Multicenter Studies as Topic , Oocyte Retrieval , Ovary/physiopathology , Ovulation Induction , Pregnancy , Pregnancy Rate , Prospective Studies , Randomized Controlled Trials as Topic , Sperm Injections, Intracytoplasmic , Time Factors , Treatment Outcome
19.
Mol Cell Endocrinol ; 483: 64-73, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30654004

ABSTRACT

The use of Bisphenol S (BPS) was proposed as an alternative to Bisphenol A (BPA), a chemical employed in the production of polycarbonate plastics and epoxy resins. BPA is a xenoestrogen that affects normal physiology in several species. It was reported that BPS may also act as a xenoestrogen with harmful effects in the reproductive system. Here we studied the effects of BPS during the induction of a polycystic ovarian syndrome (PCOS)-like condition in rats. Animals were injected daily with vehicle, DHEA 60 mg/kg, BPS 1 µg/kg and DHEA-BPS, for 20 days. Cell apoptosis, cell proliferation, and EZRIN expression were analyzed by immunohistochemistry. We found an increase in PCNA expression, which correlates with cytoplasmic accumulation of the polarization marker, EZRIN, in the BPS treated groups. Additionally, the administration of BPS in the DHEA treated group augmented the stratification and number of "intraepithelial lumina" in the endometrial surface epithelium.


Subject(s)
Cytoskeletal Proteins/metabolism , Dehydroepiandrosterone/administration & dosage , Endometrium/cytology , Phenols/administration & dosage , Polycystic Ovary Syndrome/metabolism , Sulfones/administration & dosage , Up-Regulation , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Dehydroepiandrosterone/adverse effects , Disease Models, Animal , Endometrium/drug effects , Endometrium/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Organ Size/drug effects , Phenols/adverse effects , Polycystic Ovary Syndrome/chemically induced , Proliferating Cell Nuclear Antigen/metabolism , Rats , Sulfones/adverse effects
20.
Mol Reprod Dev ; 86(4): 370-378, 2019 04.
Article in English | MEDLINE | ID: mdl-30633842

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common endocrinopathy and an important metabolic disorder in women of reproductive age. Insulin resistance (IR) is one of its most important clinical features in patients with PCOS. Androgen excess-induced mitochondrial dysfunction contributes to skeletal muscle IR in dehydroepiandrosterone (DHEA)-induced PCOS mice. The effect of androgen excess on the skeletal muscle, however, is incompletely characterized. A nontargeted metabolomics approach was thus applied to analyze the metabolites in skeletal muscle of DHEA-induced PCOS mice. Data from metabolomic analysis revealed the significant changes in 32 metabolites and the marked impact of five metabolic pathways. ATP production was also found to be significantly reduced in skeletal muscle of DHEA mice. Combined with the quantification of type I and II myofibers and lipid measurement in the skeletal muscle of the mice, the results from the present study supported the role of mitochondrial impairment rather than lipid accumulation in the pathogenesis of skeletal muscle IR in DHEA-induced PCOS mice. In summary, we show here for the first time the profile of the metabolites in the skeletal muscle of DHEA-induced PCOS mice which exhibit IR. The work would help better understand the pathology of skeletal muscle IR in PCOS.


Subject(s)
Dehydroepiandrosterone/adverse effects , Metabolomics , Muscle, Skeletal/metabolism , Polycystic Ovary Syndrome/metabolism , Animals , Dehydroepiandrosterone/pharmacology , Disease Models, Animal , Female , Humans , Mice , Muscle, Skeletal/pathology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...