Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 607
Filter
1.
Nutrients ; 13(7)2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34371958

ABSTRACT

Female reproductive aging is an irreversible process associated with a decrease in oocyte quality, which is a limiting factor for fertility. Previous studies have shown that dehydroepiandrosterone (DHEA) has been shown to improve in vitro fertilization (IVF) outcomes in older women. Herein, we showed that the decline in oocyte quality with age is accompanied by a significant decrease in the level of bioenergetic metabolism genes. We compared the clinical characteristics between groups of infertile women who either received DHEA or did not. Treatment with DHEA may enhance oocyte quality by improving energy production and metabolic reprogramming in cumulus cells (CCs) of aging women. Our results showed that compared with the group without DHEA, the group with DHEA produced a large number of day-three (D3) embryos, top-quality D3 embryos, and had improved ongoing pregnancy rate and clinical pregnancy rate. This may be because DHEA enhances the transport of oxidative phosphorylation and increases mitochondrial oxygen consumption in CCs, converting anaerobic to aerobic metabolism commonly used by aging cells to delay oocyte aging. In conclusion, our results suggest that the benefit of DHEA supplementation on IVF outcomes in aging cells is significant and that this effect may be mediated in part through the reprogramming of metabolic pathways and conversion of anaerobic to aerobic respiration.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Dietary Supplements , Energy Metabolism , Infertility, Female/metabolism , Oocytes/metabolism , Organelle Biogenesis , Adult , Aging , Cellular Senescence , Cumulus Cells/metabolism , Female , Fertilization in Vitro , Humans , Mitochondria/metabolism , Oxidative Phosphorylation , Oxygen Consumption , Pregnancy , Pregnancy Outcome
2.
Gynecol Endocrinol ; 37(8): 746-752, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34169794

ABSTRACT

OBJECTIVE: To develop a best practice document for the management of postmenopausal vulvovaginal atrophy (VVA). METHOD: Literature review carried out using clinical terms, treatments or interventions and comorbidity related to VVA. RESULTS: There is a wide variety of interventions that may produce temporal benefits for VVA. However, there are significant limitations in scientific publications concerning VVA and related issues, including variable outcome evaluations, variability in population age range, and small, often underpowered sample sizes. Therapeutic management of VVA should follow a sequential order, considering women's age, symptoms, general health as well as treatment preference. Beneficial options include lubricants, moisturizers, vaginal estrogens (estradiol, estriol, promestriene, conjugated estrogens), androgens, prasterone, and laser application. In women with general menopausal symptoms who are candidates for systemic hormone therapy, the lowest effective dose should be used. Oral ospemifene is an effective selective estrogen receptor modulator to treat VVA. Systemic androgens have a limited role. Although laser procedures are commonly used, at this moment the International Society for the Study of Vulvovaginal Disease does not endorse its use out of the setting of clinical trials. Pelvic floor muscle training improves blood flow and elasticity of the vulvovaginal tissue. In breast cancer survivors, moisturizers and lubricants are first line therapy. However, limited absorption of low/ultra-low doses of estrogens suggests safety, especially in women under treatment with aromatase inhibitors. As clinical practice and available preparations vary between countries this text should be adapted to local circumstances. CONCLUSIONS: There is a wide range of therapeutic options to individualize VVA treatments.


Subject(s)
Postmenopause/physiology , Vagina/pathology , Vaginal Diseases/therapy , Vulva/pathology , Vulvar Diseases/therapy , Administration, Intravaginal , Atrophy , Breast Neoplasms , Dehydroepiandrosterone/administration & dosage , Estrogens/administration & dosage , Estrogens/therapeutic use , Female , Hormone Replacement Therapy , Humans , Laser Therapy , Lubricants/administration & dosage , Pelvic Floor , Testosterone/administration & dosage
3.
Pharmacol Res Perspect ; 9(2): e00724, 2021 04.
Article in English | MEDLINE | ID: mdl-33638308

ABSTRACT

BNN27 is a novel 17-spiroepoxy derivative of the neurosteroid Dehydroepiandrosterone with neuroprotective properties. The purpose of this study was the detection and quantification of BNN27 after single intraperitoneal administration, in the serum and retina of normal rodents. Forty-two C57BL/6 mice and 48 Sprague-Dawley rats were used for the quantification of BNN27 in the blood serum and retina, respectively. BNN27 was injected intraperitoneally (i.p.) at concentrations of 100 and 30 mg/kg of body weight (b.w.), respectively. The blood was collected with retro-orbital bleeding and the retina was isolated after enucleation at various time points. The molecule concentrations were measured with Liquid chromatography-mass spectrometry (LC-MS). Non-compartmental analysis was used to determine pharmacokinetic parameters. BNN27 was found to have an elimination constant kel  = 0.465 h-1 and mean residence time (MRT) 2.154 h in the mouse serum. The maximum concentration (Cmax ) in the retina was detected at 2 h ( tCmax ) after intraperitoneal administration and was equal to 1100 ng/g. BNN27 is rapidly eliminated from both blood and retina. In the retina specifically, it is undetectable 6 h after injection. BNN27 shows a rapid systemic elimination as anticipated by its small size and lipophilicity. It is measurable in small peripheral tissues such as the rat retina, after one single i.p. injection, using a simple method such as LC-MS. Its detection in the retina corroborates the existing biological data that the molecule crosses the blood-retinal barrier, highlighting it as a potential neuroprotective agent for retinal disease.


Subject(s)
Dehydroepiandrosterone/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Animals , Area Under Curve , Blood-Retinal Barrier/metabolism , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/analysis , Female , Injections, Intraperitoneal , Male , Metabolic Clearance Rate , Mice , Models, Animal , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/analysis , Permeability , Rats , Retina/chemistry , Tissue Distribution
4.
J Neurosci Res ; 99(5): 1474-1495, 2021 05.
Article in English | MEDLINE | ID: mdl-33583101

ABSTRACT

BNN20, a C17-spiroepoxy derivative of the neurosteroid dehydroepiandrosterone, has been shown to exhibit strong neuroprotective properties but its role in glial populations has not been assessed. Our aim was to investigate the effect of BNN20 on glial populations by using in vitro and in vivo approaches, taking advantage of the well-established lysophosphatidylcholine (LPC)-induced focal demyelination mouse model. Our in vivo studies, performed in male mice, showed that BNN20 treatment leads to an increased number of mature oligodendrocytes (OLs) in this model. It diminishes astrocytic accumulation during the demyelination phase leading to a faster remyelination process, while it does not affect oligodendrocyte precursor cell recruitment or microglia/macrophage accumulation. Additionally, our in vitro studies showed that BNN20 acts directly to OLs and enhances their maturation even after they were treated with LPC. This beneficial effect of BNN20 is mediated, primarily, through the neurotrophin receptor TrkA. In addition, BNN20 reduces microglial activation and their transition to their pro-inflammatory state upon lipopolysaccharides stimulation in vitro. Taken together our results suggest that BNN20 could serve as an important molecule to develop blood-brain barrier-permeable synthetic agonists of neurotrophin receptors that could reduce inflammation, protect and increase the number of functional OLs by promoting their differentiation/maturation.


Subject(s)
Dehydroepiandrosterone/analogs & derivatives , Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Animals , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism
5.
Complement Ther Med ; 56: 102620, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33220453

ABSTRACT

BACKGROUND: Despite the fact that numerous clinical studies have evaluated the positive effects of dehydroepiandrosterone (DHEA) supplementation on testosterone concentrations and on the body mass index (BMI), more evidence is needed to certify that DHEA is a BMI-reducing agent in the elderly. This meta-analysis aims to clarify the various incompatible results and investigate the impact of DHEA supplementation on serum testosterone levels and lean body mass in elderly women. METHODS: Four scientific databases (EMBASE, PubMed/MEDLINE, Scopus and Web of Science) were searched from inception until 20 August 2020 for trials comparing DHEA with placebo. Results were presented as weighted mean differences (WMDs) and 95 % confidence intervals (CIs) based on the random effects model (DerSimonian-Laird approach). RESULTS: Nine arms with 793 subjects reported testosterone as an outcome measure. The overall results demonstrated that testosterone levels increased significantly after DHEA administration in elderly women (WMD: 17.52 ng/dL, 95 % CI: 6.61, 28.43, P = 0.002). In addition, DHEA administration significantly decreased the BMI (WMD:-0.39 kg/m2, I2 = 0.0 %). CONCLUSION: The results of the current meta-analysis support the use of DHEA supplementation for increasing testosterone concentrations in elderly women.


Subject(s)
Body Mass Index , Dehydroepiandrosterone , Testosterone/blood , Aged , Body Composition/drug effects , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Female , Humans , Middle Aged , Randomized Controlled Trials as Topic
6.
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
7.
J Clin Pharmacol ; 60 Suppl 2: S74-S85, 2020 12.
Article in English | MEDLINE | ID: mdl-33274517

ABSTRACT

Every woman, if she lives long enough, will transition into menopause, and as the US population ages, women will be spending more time in a postmenopausal state than before. For postmenopausal women, the decision to initiate menopausal hormone therapy should be individualized. A thorough evaluation of the patient's cardiovascular, venous thromboembolic, cancer, and fracture risk should be considered along with the woman's quality of life. Hormone therapy exerts its therapeutic effects on vasomotor symptoms, the skeleton, and the genitourinary system independent of age since menopause and these benefits are lost once hormone therapy is stopped. Here we review the pharmacologic properties dose, formulation, mode of administration, timing of initiation, and duration of hormonal therapies in regard to optimizing benefit and minimizing risk to the patient. This discussion will focus on the effects of common hormonal therapies including estrogen (local and systemic), progesterone, estrogen receptor agonist/antagonist, and local dehydroepiandrosterone and include a brief review of compounded bioidentical hormone therapy.


Subject(s)
Estrogen Replacement Therapy/methods , Menopause/drug effects , Dehydroepiandrosterone/administration & dosage , Estrogens/administration & dosage , Estrogens/adverse effects , Estrogens/metabolism , Estrogens/pharmacokinetics , Female , Female Urogenital Diseases/drug therapy , Humans , Progestins/administration & dosage , Progestins/adverse effects , Progestins/metabolism , Progestins/pharmacokinetics , Receptors, Estrogen/drug effects , Testosterone/administration & dosage
8.
Complement Ther Med ; 55: 102583, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33220623

ABSTRACT

AIMS: The effect of DHEA supplementation on fasting plasma glucose (FPG), insulin levels (IN) and the homeostasis model assessment-estimated insulin resistance (HOMA-IR) index in humans has not been assessed so far. Thus, we aimed to conduct a systematic review and meta-analysis of the randomized controlled trials (RCT) which assessed the effects of DHEA supplementation on FPG, IN and the HOMA-IR index in humans. METHODS: An extensive search was performed in Scopus, PubMed/MEDLINE, and Web of Science from inception to June 2020. Data was combined using the random effects model. RESULTS: 14 publications were included in this study. Overall results demonstrated that FPG was significantly altered after DHEA consumption (WMD: -2.185 mg/dl, P = 0.029). DHEA administration did not result in any significant changes in IN (WMD: 0.057 µU/mL, P = 0.067), and the HOMA - IR index (WMD: 0.174, P = 0.060). In the subgroup analyses, FPG significantly decreased in the subgroup who received DHEA supplementation in dosages of ≤50 mg/day (WMD: -2.29 mg/dl), when the treatment duration was <12 weeks (WMD: -5.25 mg/dl), and in subjects aged ≥60 years (WMD: -2.94 mg/dl). CONCLUSION: This systematic review evaluated the association between FPG and DHEA, revealing that the administration of DHEA reduces FPG levels. However, we found no association between DHEA administration and IN levels or insulin resistance.


Subject(s)
Blood Glucose/drug effects , Dehydroepiandrosterone/administration & dosage , Insulin Resistance , Insulin/metabolism , Adjuvants, Immunologic/administration & dosage , Dose-Response Relationship, Drug , Humans , Randomized Controlled Trials as Topic
9.
Ginekol Pol ; 91(9): 554-562, 2020.
Article in English | MEDLINE | ID: mdl-33030737

ABSTRACT

Dehydroepiandrosterone (DHEA) concentration decreases with age, therefore, DHEA has been considered a hormone that reduces the symptoms associated with aging, so the usefulness of DHEA in premenopausal and postmenopausal women, and the options of hormone therapy have received a large amount of attention. The effectiveness of DHEA in the premenopausal women remains unclear, while in postmenopausal women with coexisting estrogens deficiency is controversial. Despite many years of study, the use of DHEA is still controversial, especially regarding its effectiveness. The aim of present article was to evaluate DHEA specific effects on metabolic parameters, bone mineral density, insulin resistance as well as the therapeutic potential of DHEA in pre- and postmenopausal women using measures of sexual activity, cognition and well-being. The summary of this article is the position statement of expert group of the Polish Menopause and Andropause Society regarding the efficacy and safety of DHEA supplementation in women. We concluded, that currently available clinical trials and meta-analyses indicate that DHEA supplementation is effective in women with adrenal insufficiency and chronically treated with exogenous glucocorticoids, postmenopausal women with low bone mineral density and/or osteoporosis, premenopausal women with sexual disorders and low libido, and in women with vulvovaginal atrophy due to menopause or genitourinary syndrome of menopause. Currently available clinical trials also suggest that DHEA supplementation is probably effective in postmenopausal women with hypoactive sexual disorders, infertile women with diminished ovarian reserve, women suffering from depression and anxiety, and women with obesity and insulin resistance. No serious adverse effects have been reported.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Dietary Supplements , Aged , Female , Humans , Poland , Postmenopause , Practice Guidelines as Topic , Premenopause , Societies, Medical
10.
Int J Rheum Dis ; 23(12): 1692-1697, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32975909

ABSTRACT

AIM: Chronic inflammation and subsequent use of glucocorticoids can lead to relative adrenocortical insufficiency in patients with rheumatoid arthritis (RA). Previously, adrenocortical hormone, dehydroepiandrosterone (DHEA) was shown as a potential therapy for autoimmune disorders. However, data regarding effects of DHEA in RA are limited. The aim of this study was to investigate the effects of DHEA on quality of life (QOL) in premenopausal rheumatoid arthritis patients. METHOD: In this randomized double blinded, controlled trial 46 premenopausal rheumatoid arthritis patients were assigned to receive 50 mg/d DHEA (23 patients) or placebo (23 patients) for 12 weeks. Disease Activity Score of 28 joints - erythrocyte sedimentation rate (DAS28-ESR) questionnaire, visual analog score and swollen and tender joint counts (both 0-28) were used for assessment of disease activity. Persian-validated World Health Organization Quality of Life Brief version (WHOQOL BREF) questionnaire was used to assess quality of life. RESULTS: In comparison to the control group more improvement in QOL (P = .025) and environment health (P = .001) was observed in the DHEA group. After adjustment for age and disease duration DHEA was associated with more improvement in QOL (P = .01), psychological (P = .02) and physical health (P = .03). A trend toward a decrease in ESR was observed in DHEA group (P = .06). DAS was improved in both groups; however, there was no significant change in DAS28 between groups (P = .88). Frequency of adverse events albeit minor was similar in both groups. CONCLUSION: Our study supports a slightly superior effect of DHEA over placebo to improve QOL in premenopausal female patients with rheumatoid arthritis. We did not find improvement in DAS in the DHEA group over placebo.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Dehydroepiandrosterone/administration & dosage , Premenopause/psychology , Quality of Life , Adjuvants, Immunologic/administration & dosage , Adult , Arthritis, Rheumatoid/psychology , Double-Blind Method , Female , Humans , Surveys and Questionnaires , Treatment Outcome
11.
Eur Rev Med Pharmacol Sci ; 24(17): 9104-9111, 2020 09.
Article in English | MEDLINE | ID: mdl-32965000

ABSTRACT

OBJECTIVE: We investigated whether DHEA supplementation had an impact on ovarian reserve parameters and pregnancy rates in patients with poor ovarian response (POR) and primary ovarian insufficiency (POI). PATIENTS AND METHODS: A total of 34 people, 6 patients with POI and 28 patients with POR, were included in the study. The patients in the POR group consisted of two different groups: diminished ovarian reserve (DOR) and premature ovarian failure (PMOF). Patients in the POI and POR group were given 50 mg DHEA supplementation daily for 5 months. The primary outcome was to determine spontaneous clinical pregnancy rates. The monthly changes in the serum hormone levels and AFC were recorded for five months. AMH levels were also measured before and after treatment. RESULTS: The total follow-up time was 152 cycles. The number of pregnancies during the follow-up period was 9. The ratio of pregnancies to the number of patients was 26.5% and the rate per cycle was 5.9%. While 8 of 9 pregnancies resulted in a live birth, one resulted in a miscarriage. The rate of abortion was 11.1%. The mean AFC was 0 to 5 before treatment. Following DHEA administration, a significant increase was detected in 30.8% of the patients. There was an increase in AMH levels after DHEA, but this was not significant. The live birth rate and pregnancy rate per cycle were significantly higher in POR patients than those in POF. Patients with POF had no pregnancy. Although the PMOF patients were younger than the DOR patients, the rate of pregnancy (36% vs. 29%), and pregnancy rates per cycle (8.5% vs. 6.35%) were higher in the DOR group. The rates of live birth were the same in the PMOF and DOR groups (29% vs. 29%). CONCLUSIONS: Oral DHEA supplementation improves both ovarian reserve and pregnancy rates in women with POR.


Subject(s)
Dehydroepiandrosterone/pharmacology , Dietary Supplements , Infertility, Female/drug therapy , Ovarian Reserve/drug effects , Pregnancy Rate , Primary Ovarian Insufficiency/drug therapy , Adult , Dehydroepiandrosterone/administration & dosage , Female , Humans , Pregnancy
13.
Life Sci ; 249: 117515, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32147428

ABSTRACT

AIMS: This study investigated the effects of curcumin-loaded super-paramagnetic iron oxide (Fe3O4) nanoparticles (NPs) (SPIONs) on histological parameters and apoptosis-inducing factors (AIFs) in an experimental mouse model of polycystic ovary syndrome (PCOS). MATERIALS AND METHODS: A total number of 40 female prepuberal BALB/c mice were randomly divided into four groups. Group 1 was selected as control and Group 2 was considered as a vehicle taking sesame oil, in the form of a curcumin carrier. Moreover, Group 3 was administered with dehydroepiandrosterone (DHEA) at 6 mg/100 g of the body weight and Group 4 received the DHEA plus the NPs of curcumin (5.4 mg/100 g) for twenty consecutive days. Finally, histology, stereology, and apoptosis of the ovary were evaluated. KEY FINDINGS: The results revealed that the NPs of curcumin had reduced ovarian volume (p < 0.05) and a total number of primary, secondary, antral, and primordial follicles in comparison with the PCOS and vehicle groups (p < 0.05). Furthermore, curcumin treatment following administration of the DHEA resulted in a significant decrease in BAX (p < 0.001) and levels of expression of Caspase3 (CASP3) protein, increased levels of B-cell lymphoma 2 (Bcl2) expression (p < 0.05), and moderated apoptosis in granulosa cells in comparison with the ones seen in the PCOS group. SIGNIFICANCE: Ovarian injuries and DHEA-induced apoptosis were efficiently suppressed by curcumin, indicating the probable protective property of NPs of curcumin against PCOS.


Subject(s)
Apoptosis/drug effects , Curcumin/administration & dosage , Dehydroepiandrosterone/administration & dosage , Ferric Compounds/administration & dosage , Metal Nanoparticles/chemistry , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Sexual Maturation , Animals , Antioxidants/administration & dosage , Apoptosis/genetics , Caspase 3/metabolism , Disease Models, Animal , Female , Ferric Compounds/chemistry , Gene Expression/drug effects , Mice , Mice, Inbred BALB C , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Oxidative Stress
14.
Med J Malaysia ; 75(1): 24-28, 2020 01.
Article in English | MEDLINE | ID: mdl-32008015

ABSTRACT

BACKGROUND: In reproductive medicine poor ovarian response (POR) among women undergoing in vitro fertilisation (IVF) is of great concern. Meta-analysis showed that Dehydroepiandrosterone (DHEA) administration resulted in a significant increase in the number of oocytes retrieved in women with POR. The aim of this study was to assess the effectiveness of DHEA supplementation on IVF outcomes among poor responders undergoing IVF. METHODS: Sixteen patients who were diagnosed with POR scheduled to undergo their second cycle of Intracytoplasmic sperm injection (ICSI)/embryo transfer cycle were enrolled. All enrolled patients had earlier undergone their first ICSI/embryo transfer cycle at least four months prior to this study. All subjects were given DHEA supplementation of 25mg three times daily for at least three months prior to their second ICSI/embryo transfer cycle. Statistical analysis of various ovarian response and ICSI outcomes parameter were compared pre and post DHEA. RESULTS: Sixteen women with the mean age of 35 years were enrolled in the study. The comparative analysis of results showed a significant increase in the number of good quality of embryos obtained (p<0.05). After the treatment with DHEA, there was an improvement in the number of oocytes retrieved, Metaphase II (MII) oocyte (mature) oocytes obtained, fertilised and transferrable embryos and the pregnancy rate. There was no significant effect of DHEA treatment on the number of days of stimulation and cumulative dose of gonadotrophins used. CONCLUSION: Our results is able to show that DHEA supplementation may help to enhance IVF-ICSI outcomes in women with POR especially in those age 35 years and below.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/pharmacology , Fertilization in Vitro/drug effects , Oocytes/drug effects , Adult , Female , Humans , Malaysia , Outcome Assessment, Health Care , Retrospective Studies
15.
Expert Opin Pharmacother ; 21(4): 409-415, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31928093

ABSTRACT

Introduction: Dyspareunia caused by vulvovaginal atrophy is a primary symptom of genitourinary syndrome of menopause (GSM), a chronic, progressive medical condition that results from estrogen and androgen deficiency at menopause. Dehydroepiandrosterone (DHEA, prasterone) is an endogenous precursor steroid hormone that is metabolized into both androgens and estrogens that has been recently been approved by the FDA for the treatment of moderate to severe dyspareunia caused by vulvovaginal atrophy secondary to menopause.Areas covered: This is a comprehensive drug evaluation describing the chemical composition, pharmacokinetics, metabolism, clinical efficacy and safety of dehydroepiandrosterone (prasterone) in the treatment of dyspareunia and VVA secondary to menopause. Preclinical and clinical data suggesting further potential uses, benefits, and contraindications in the genitourinary health of postmenopausal women are also considered.Expert opinion: Intravaginal dehydroepiandrosterone (prasterone) is effective for the management of dyspareunia secondary to menopause and may be effective in the treatment of other types of sexual dysfunction that are secondary to menopause. Further studies should explore additional dosing regimens and different indications.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Female Urogenital Diseases/drug therapy , Menopause/metabolism , Vagina/drug effects , Administration, Intravaginal , Androgens/metabolism , Atrophy , Dehydroepiandrosterone/administration & dosage , Dyspareunia/drug therapy , Dyspareunia/metabolism , Estrogens/metabolism , Female , Female Urogenital Diseases/metabolism , Humans , Treatment Outcome , Vagina/pathology
16.
Cells ; 9(1)2020 01 14.
Article in English | MEDLINE | ID: mdl-31947651

ABSTRACT

Advanced glycation end-products (AGEs) are involved in the pathogenesis and consequences of polycystic ovary syndrome (PCOS), a complex metabolic disorder associated with female infertility. The most powerful AGE precursor is methylglyoxal (MG), a byproduct of glycolysis, that is detoxified by the glyoxalase system. By using a PCOS mouse model induced by administration of dehydroepiandrosterone (DHEA), we investigated whether MG-dependent glycative stress contributes to ovarian PCOS phenotype and explored changes in the Sirtuin 1 (SIRT1) functional network regulating mitochondrial functions and cell survival. In addition to anovulation and reduced oocyte quality, DHEA ovaries revealed altered collagen deposition, increased vascularization, lipid droplets accumulation and altered steroidogenesis. Here we observed increased intraovarian MG-AGE levels in association with enhanced expression of receptor for AGEs (RAGEs) and deregulation of the glyoxalase system, hallmarks of glycative stress. Moreover, DHEA mice exhibited enhanced ovarian expression of SIRT1 along with increased protein levels of SIRT3 and superoxide dismutase 2 (SOD2), and decreased peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC1α), mitochondrial transcriptional factor A (mtTFA) and translocase of outer mitochondrial membrane 20 (TOMM20). Finally, the presence of autophagy protein markers and increased AMP-activated protein kinase (AMPK) suggested the involvement of SIRT1/AMPK axis in autophagy activation. Overall, present findings demonstrate that MG-dependent glycative stress is involved in ovarian dysfunctions associated to PCOS and support the hypothesis of a SIRT1-dependent adaptive response.


Subject(s)
Glycation End Products, Advanced/metabolism , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , Pyruvaldehyde/metabolism , Sirtuin 1/metabolism , Animals , Dehydroepiandrosterone/administration & dosage , Disease Models, Animal , Female , Glycosylation , Mice , Polycystic Ovary Syndrome/chemically induced
17.
Gynecol Endocrinol ; 36(2): 162-165, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31311350

ABSTRACT

Premature ovarian insufficiency (POI) is a delicate medical problem in young women. This condition is not unchangeable and permanent but is associated with intermittent and unpredictable ovarian activity, resulting in low conception rate. Over the period of 8 years, the evaluation of secondary amenorrhea was conducted in 90 patients below the age of 40 who wished to restore fertility. Having confirmed the diagnosis and investigated the etiology of POI, hormone replacement therapy was applied (sequential administration of estradiol and norethisterone acetate) in the first 30 patients (group A). Estrogen-progestogen therapy with daily supplementation of 25 mg of micronized oral dehydroepiandrosterone (DHEA) was conducted in 44 patients (group B), whereas a combined regime (estrogen-progestogen therapy, DHEA supplementation in daily dose of 25 mg, and melatonin supplementation in daily dose of 3 mg) was conducted in 16 patients (group C). In the course of our study, 16 pregnancies were realized (18% of all cases: 17% in group A; 18% in group B; 19% in group C) 6 to 20 months after the initiation of hormone therapy, and there have been 13 completed term pregnancies so far with normal fetal growth and development. We concluded that estrogen-progestogen therapy combined with DHEA and melatonin could optimize fertility and lead to successful pregnancy in POI patients.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Estradiol/therapeutic use , Fertility/drug effects , Primary Ovarian Insufficiency/drug therapy , Progesterone/therapeutic use , Adult , Dehydroepiandrosterone/administration & dosage , Estradiol/administration & dosage , Female , Hormone Replacement Therapy , Humans , Progesterone/administration & dosage , Treatment Outcome
18.
Neuropharmacology ; 165: 107919, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31877321

ABSTRACT

BNN-20 is a synthetic microneurotrophin, long-term (P1-P21) administration of which exerts potent neuroprotective effect on the "weaver" mouse, a genetic model of progressive, nigrostriatal dopaminergic degeneration. The present study complements and expands our previous work, providing evidence that BNN-20 fully protects the dopaminergic neurons even when administration begins at a late stage of dopaminergic degeneration (>40%). Since neuroinflammation plays a critical role in Parkinson's disease, we investigated the possible anti-neuroinflammatory mechanisms underlying the pharmacological action of BNN-20. The latter was shown to be microglia-mediated, at least in part. Indeed, BNN-20 induced a partial, but significant, reversal of microglia hyperactivation, observed in the untreated "weaver" mouse. Furthermore, it induced a shift in microglia polarization towards the neuroprotective M2 phenotype, suggesting a possible beneficial shifting of microglia activity. This observation was further supported by morphometric measurements. Moreover, BDNF levels, which were severely reduced in the "weaver" mouse midbrain, were restored to normal even after short-term BNN-20 administration. Experiments in "weaver"/NGL (dual GFP/luciferase-NF-κВ reporter) mice using bioluminescence after a short BNN-20 treatment (P60-P74), have shown that the increase of BDNF production was specifically mediated through the TrkB-PI3K-Akt-NF-κB signaling pathway. Interestingly, long-term BNN-20 treatment (P14-P60) significantly increased dopamine levels in the "weaver" striatum, which seems to be associated with the improved motor activity observed in the treated mutant animals. In conclusion, our findings suggest that BNN-20 may serve as a lead molecule for new therapeutic compounds for Parkinson's disease, combining strong anti-neuroinflammatory and neuroprotective properties, leading to elevated dopamine levels and improved motor activity.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Dehydroepiandrosterone/analogs & derivatives , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Encephalitis/metabolism , Neuroprotective Agents/administration & dosage , Parkinson Disease/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Dehydroepiandrosterone/administration & dosage , Disease Models, Animal , Encephalitis/complications , Encephalitis/prevention & control , Female , Male , Membrane Glycoproteins/metabolism , Mice, Neurologic Mutants , Microglia/drug effects , Microglia/metabolism , Parkinson Disease/complications , Parkinson Disease/prevention & control , Pars Compacta/drug effects , Pars Compacta/metabolism , Protein-Tyrosine Kinases/metabolism , Tyrosine 3-Monooxygenase/metabolism
19.
J Womens Health (Larchmt) ; 29(1): 57-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31687883

ABSTRACT

Androgens are believed to have an important biologic role in women, particularly in regulation of libido and sexual arousal, although much about their function on other systems in women is unknown. Testosterone, the primary ovarian androgen, has been used to treat carefully selected postmenopausal women with hypoactive sexual desire disorder (HSDD). However, testosterone use in women has not been approved by the United States Food and Drug Administration (FDA) because of uncertainties regarding the effectiveness and long-term safety of this strategy. An intravaginal form of the adrenal androgen, dehydroepiandrosterone (DHEA) has been approved by the FDA to treat genitourinary syndrome of menopause. In this article, we review the current knowledge regarding the role of androgens and their clinical use in women. We conducted a systematic search of PubMed for publications describing the role and clinical use of androgens in women. We used the search terms "HSDD," "DHEA in women," "testosterone in women," and "androgens in women," and reviewed most references from all relevant articles. Most randomized placebo-controlled trials show an improvement in sexual function with low-dose testosterone therapy in select postmenopausal women with HSDD. Although this strategy appears to be safe in the short term and no major safety concerns have emerged thus far, long-term effects on cardiovascular risk and breast cancer incidence are not known. A trial of low-dose testosterone therapy may be considered for carefully selected postmenopausal women with HSDD, as long as other contributors to sexual dysfunction have been adequately addressed. However, patients need careful counseling regarding the lack of long-term safety data, and close clinical and laboratory monitoring of these women is recommended to avoid supraphysiologic dosing.


Subject(s)
Androgens/administration & dosage , Libido/drug effects , Sexual Dysfunctions, Psychological/drug therapy , Testosterone/administration & dosage , Dehydroepiandrosterone/administration & dosage , Female , Hormone Replacement Therapy/methods , Humans , Postmenopause/drug effects
20.
Drug Test Anal ; 11(11-12): 1629-1643, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31701664

ABSTRACT

7-keto-DHEA (3ß-hydroxy-androst-5-ene-7,17-dione) is included in section S1 of the World Antidoping Agency (WADA) List of Prohibited Substances. The detection of its misuse in sports needs special attention, since it is naturally present in urine samples. The main goal of this study is to investigate the in vivo metabolism of 7-keto-DHEA after a single administration to healthy volunteers and to better describe the relationship between arimistane (androst-5-ene-7,17-dione) and 7-keto-DHEA after the application of the common routine procedures to detect anabolic steroids in WADA accredited antidoping laboratories. Free, glucuro-, and sulpho-conjugated steroids extracted from urine samples obtained before and after the administration of 7-keto-DHEA were analyzed by different gas chromatographic (GC)-mass spectrometric (MS) techniques. Gas chromatography coupled to tandem MS to study the effect on the endogenous steroid profile, coupled to isotope ratio mass spectrometry (IRMS) to investigate the potential formation of androgens derived from DHEA and coupled to high resolution accurate mass spectrometry (HRMS) to investigate new diagnostic metabolites. The analysis by IRMS confirmed that there is no formation of DHEA from 7-keto-DHEA. Ten proposed metabolites, not previously reported, were described. These include reduced and hydroxylated structures that are not considered part of the steroid profile in antidoping analyses. They showed considerable responses in all fractions analyzed. Some deoxidation reactions (including arimistane formation) were found and most probably can be linked to the sample preparation or instrumental analysis. This is important when interpreting the results after the application of procedures to detect steroids in urine currently used in antidoping laboratories. 7-keto-DHEA metabolism in humans for antidoping purposes was studied and unexpected results were found. This could lead to a misinterpretation of the data, depending on the procedure applied and the analytical instrumentation used.


Subject(s)
Anabolic Agents/metabolism , Dehydroepiandrosterone/analogs & derivatives , Anabolic Agents/administration & dosage , Anabolic Agents/urine , Chromatography, High Pressure Liquid , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone/urine , Doping in Sports , Gas Chromatography-Mass Spectrometry , Humans , Hydroxylation , Substance Abuse Detection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...