Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 456
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Elife ; 122024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38358793

RESUMEN

Background: Primary angle closure glaucoma (PACG) is the leading cause of irreversible blindness in Asia, and no reliable, effective diagnostic, and predictive biomarkers are used in clinical routines. A growing body of evidence shows metabolic alterations in patients with glaucoma. We aimed to develop and validate potential metabolite biomarkers to diagnose and predict the visual field progression of PACG. Methods: Here, we used a five-phase (discovery phase, validation phase 1, validation phase 2, supplementary phase, and cohort phase) multicenter (EENT hospital, Shanghai Xuhui Central Hospital), cross-sectional, prospective cohort study designed to perform widely targeted metabolomics and chemiluminescence immunoassay to determine candidate biomarkers. Five machine learning (random forest, support vector machine, lasso, K-nearest neighbor, and GaussianNaive Bayes [NB]) approaches were used to identify an optimal algorithm. The discrimination ability was evaluated using the area under the receiver operating characteristic curve (AUC). Calibration was assessed by Hosmer-Lemeshow tests and calibration plots. Results: Studied serum samples were collected from 616 participants, and 1464 metabolites were identified. Machine learning algorithm determines that androstenedione exhibited excellent discrimination and acceptable calibration in discriminating PACG across the discovery phase (discovery set 1, AUCs=1.0 [95% CI, 1.00-1.00]; discovery set 2, AUCs = 0.85 [95% CI, 0.80-0.90]) and validation phases (internal validation, AUCs = 0.86 [95% CI, 0.81-0.91]; external validation, AUCs = 0.87 [95% CI, 0.80-0.95]). Androstenedione also exhibited a higher AUC (0.92-0.98) to discriminate the severity of PACG. In the supplemental phase, serum androstenedione levels were consistent with those in aqueous humor (r=0.82, p=0.038) and significantly (p=0.021) decreased after treatment. Further, cohort phase demonstrates that higher baseline androstenedione levels (hazard ratio = 2.71 [95% CI: 1.199-6.104], p=0.017) were associated with faster visual field progression. Conclusions: Our study identifies serum androstenedione as a potential biomarker for diagnosing PACG and indicating visual field progression. Funding: This work was supported by Youth Medical Talents - Clinical Laboratory Practitioner Program (2022-65), the National Natural Science Foundation of China (82302582), Shanghai Municipal Health Commission Project (20224Y0317), and Higher Education Industry-Academic-Research Innovation Fund of China (2023JQ006).


Asunto(s)
Androstenodiona , Glaucoma de Ángulo Cerrado , Humanos , Teorema de Bayes , Biomarcadores , China , Estudios Transversales , Glaucoma de Ángulo Cerrado/diagnóstico , Estudios Prospectivos , Campos Visuales
2.
Biomolecules ; 13(11)2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-38002324

RESUMEN

BACKGROUND: Androgen deprivation therapy (ADT) is a mainstay of prostate cancer in both adjuvant and palliative settings. Since androgens are crucial for functional status and psychological functions, we evaluated whether blood testosterone, androstenedione, or DHEA concentrations were associated with functional status and psychological alterations in patients with localised (PCa) or metastatic prostate cancer (mPCa) receiving ADT with analogues of luteinising hormone-releasing hormone (LHRH). METHODS: The five Fried criteria were considered to identify frailty syndrome. In addition, complementary evaluations were carried out to measure other variables of interest. Sleep quality was assessed using the Athens Insomnia Scale, cognitive functions were assessed using the Mini-Mental State Examination, and symptoms of depression were measured using the Yesavage Geriatric Depression Scale. Logistic regression analysis was performed to determine if the androgens level could be related to frailty syndrome, sleep impairment, depressive symptoms, and cognitive functions. RESULTS: The results of the multivariate analyses show that high concentrations of androstenedione were significantly associated with frailty syndrome in both groups (p = 0.018; odds ratio = 4.66, 95% confidence interval [1.30-16.6]). There were significant relationships between frailty syndrome and the systemic concentration of androstenedione (p = 0.01), but not the concentration of testosterone (p = 0.60) or DHEA (p = 0.42). In addition, the results of the non-parametric tests show significant results between a decreased gait speed in the two groups (metastatic and localised) and the concentration of androstenedione (p = 0.015). High androstenedione levels were associated with a slow walking speed in the mCaP group (p = 0.016), while high testosterone levels were associated with a better walking speed in the localised CaP group (p = 0.03). For the concentration of androstenedione in plasma, the area under the curve was 0.72, with a 95% CI of 0.55-0.88 with acceptable values, and with a cut-off point of 4.51 pg/mL, a sensitivity of 82.9%, and specificity of 53.8%. No relationships between the concentration of androgens in plasma and sleep quality, cognitive functions, or symptoms of depression suggest that the changes were specific to frailty syndrome. CONCLUSIONS: Further research into the role of androstenedione should be evaluated in follow-up studies in order to recommend its use as a suitable biomarker of frailty syndrome in prostate cancer patients.


Asunto(s)
Fragilidad , Neoplasias de la Próstata , Masculino , Anciano , Humanos , Neoplasias de la Próstata/patología , Andrógenos , Androstenodiona , Antagonistas de Andrógenos , Anciano Frágil , Testosterona , Deshidroepiandrosterona
3.
Food Chem Toxicol ; 178: 113927, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37406756

RESUMEN

Formestane (4-OHA) has been proven to be highly effective with high systemic tolerability in treating ER+ breast cancer. However, its intramuscular administration and associated side effects make it unsuitable for adjuvant treatment, leading to its withdrawal from the market. In contrast, Formestane cream may offer a solution by providing a more convenient route of administration and retaining its tumor-shrinking effects. This suggests that 4-OHA cream could have promising clinical applications. However, before clinical application, it is necessary to evaluate the potential toxicity of the cream in animals. This study evaluated the toxicity of 4-OHA cream on female Bama minipigs in vivo by analyzing hematology, biochemistry, and histopathology. The results showed that there was no significant difference between the cream-treated group and the control normal group for each parameter analyzed, indicating that 4-OHA cream was non-dermal toxic to minipigs. This finding provides a basis for the safe clinical use of the cream.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Femenino , Porcinos , Porcinos Enanos , Androstenodiona/efectos adversos , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico
4.
Gynecol Endocrinol ; 39(1): 2227277, 2023 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-37356455

RESUMEN

OBJECTIVE: To evaluate the efficacy of dietary supplementation with a combination of antioxidants (lipoic acid, N-acetylcysteine, vitamin B6, and S-adenosyl-L-methionine) for the modulation of metabolic, endocrine, and clinical parameters in comparison with oral contraception in non-diabetic women newly diagnosed with polycystic ovary syndrome (PCOS). METHODS: This was a prospective, partially randomized, multicenter study in which non-diabetic women with PCOS were recruited under routine clinical practice conditions and distributed in three groups to receive the following regimen for 6 months: 1) antioxidant combination (MN group); 2) oral contraception (OC group); or 3) oral contraception and the antioxidant combination (MN + OC group). General recommendation of healthy diet and regular exercise was given to all patients. Metabolic, endocrine, clinical, and quality of life parameters were recorded at baseline and after 6 months of therapy. RESULTS: A total of 96 women with PCOS were included in the study. After 6 months of treatment, the homeostasis model assessment-estimated insulin resistance (HOMA-IR) level was reduced only in the MN group, with a significant mean reduction of -0.92 points. Androstenedione was significantly reduced in all groups. Clinical parameters that significantly improved in all groups were hirsutism, acne, irregular menstruation, and quality of life, with no statistical differences between the groups. CONCLUSIONS: This study showed that the antioxidant combination might be a suitable therapy for patients with PCOS when oral contraceptive is not indicated, because in all groups clinical parameters, irregular menstruation as well as androstenedione and quality of life were significantly improved with no statistical difference between groups.


Asunto(s)
Antioxidantes , Anticonceptivos Orales , Resistencia a la Insulina , Trastornos de la Menstruación , Síndrome del Ovario Poliquístico , Femenino , Humanos , Androstenodiona , Antioxidantes/uso terapéutico , Síndrome del Ovario Poliquístico/diagnóstico , Estudios Prospectivos , Calidad de Vida , Anticonceptivos Orales/uso terapéutico , Suplementos Dietéticos
5.
Hum Reprod ; 38(9): 1807-1815, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37354554

RESUMEN

STUDY QUESTION: Does 8 weeks of daily low-dose hCG administration affect androgen or inhibin B levels in serum and/or follicular fluid (FF) during the subsequent IVF/ICSI cycle in women with low ovarian reserve? SUMMARY ANSWER: Androgen levels in serum and FF, and inhibin B levels in serum, decreased following 8 weeks of hCG administration. WHAT IS KNOWN ALREADY: Recently, we showed that 8 weeks of low-dose hCG priming, in between two IVF/ICSI treatments in women with poor ovarian responder (anti-Müllerian hormone (AMH) <6.29 pmol/l), resulted in more follicles of 2-5 mm and less of 6-10-mm diameter at the start of stimulation and more retrieved oocytes at oocyte retrieval. The duration of stimulation and total FSH consumption was increased in the IVF/ICSI cycle after priming. Hypothetically, hCG priming stimulates intraovarian androgen synthesis causing upregulation of FSH receptors (FSHR) on granulosa cells. It was therefore unexpected that antral follicles were smaller and the stimulation time longer after hCG priming. This might indicate a different mechanism of action than previously suggested. STUDY DESIGN, SIZE, DURATION: Blood samples were drawn on stimulation day 1, stimulation days 5-6, trigger day, day of oocyte retrieval, and oocyte retrieval + 5 days in the IVF/ICSI cycles before and after hCG priming (the control and study cycles, respectively). FF was collected from the first aspirated follicle on both sides during oocyte retrieval in both cycles. The study was conducted as a prospective, paired, non-blinded, single-center study conducted between January 2021 and July 2021 at a tertiary care center. The 20 participants underwent two identical IVF/ICSI treatments: a control cycle including elective freezing of all blastocysts and a study cycle with fresh blastocyst transfer. The control and study cycles were separated by 8 weeks (two menstrual cycles) of hCG priming by daily injections of 260 IU recombinant hCG. PARTICIPANTS/MATERIALS, SETTING, METHODS: Women aged 18-40 years with cycle lengths of 23-35 days and AMH <6.29 pmol/l were included. Control and study IVF/ICSI cycles were performed in a fixed GnRH-antagonist protocol. MAIN RESULTS AND THE ROLE OF CHANCE: Inhibin B was lower on stimulation day 1 after hCG priming (P = 0.05). Dehydroepiandrosterone sulfate (DHEAS) was significantly lower on stimulation day 1 (P = 0.03), and DHEAS and androstenedione were significantly lower on stimulation days 5-6 after priming (P = 0.02 and P = 0.02) The testosterone level in FF was significantly lower in the study cycle (P = 0.008), while the concentrations of inhibin B and androstenedione in the FF did not differ between the study and control cycles. A lower serum inhibin B in the study cycle corresponds with the antral follicles being significantly smaller after priming, and this probably led to a longer stimulation time in the study cycle. This contradicts the theory that hCG priming increases the intraovarian androgen level, which in turn causes more FSHR on developing (antral up to preovulatory) follicles. However, based on this study, we cannot rule out that an increased intra-follicular androgen level was present at initiation of the ovarian stimulation, without elevating the androgen level in serum and that an increased androgen level may have rescued some small antral follicles that would have otherwise undergone atresia by the end of the previous menstrual cycle. We retrieved significantly more oocytes in the Study cycle, and the production of estradiol per follicle ≥10-mm diameter on trigger day was comparable in the study and control cycles, suggesting that the rescued follicles were competent in terms of producing oocytes and steroid hormones. LIMITATIONS, REASONS FOR CAUTION: The sample size was small, and the study was not randomized. Our study design did not allow for the measurement and comparison of androgen levels or FSHR expression in small antral follicles before and immediately after the hCG-priming period. WIDER IMPLICATIONS OF THE FINDINGS: The results make us question the mechanism of action behind hCG priming prior to IVF. It is important to design a study with the puncture of small antral follicles before and immediately after priming to investigate the proposed hypothesis. Improved cycle outcomes, i.e. more retrieved oocytes, must be confirmed in a larger, preferably randomized study. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by an unrestricted grant from Gedeon Richter awarded to the institution. A.P. reports personal consulting fees from PregLem SA, Novo Nordisk A/S, Ferring Pharmaceuticals A/S, Gedeon Richter Nordics AB, Cryos International, and Merck A/S outside the submitted work and payment or honoraria for lectures from Gedeon Richter Nordics AB, Ferring Pharmaceuticals A/S, Merck A/S, and Theramex and Organon & Co and payment for participation in an advisory board for Preglem. Grants to the institution have been provided by Gedeon Richter Nordics AB, Ferring Pharmaceuticals A/S, and Merck A/S, and equipment and travel support has been given to the institution by Gedeon Richter Nordics AB. The remaining authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov Identifier: NCT04643925.


Asunto(s)
Andrógenos , Reserva Ovárica , Humanos , Femenino , Embarazo , Androstenodiona , Estudios Prospectivos , Inyecciones de Esperma Intracitoplasmáticas/métodos , Inducción de la Ovulación/métodos , Fertilización In Vitro/métodos , Índice de Embarazo
6.
Microb Cell Fact ; 22(1): 19, 2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36710325

RESUMEN

4-Androstene-3,17-dione (4-AD) and 22-hydroxy-23,24-bisnorchol-4-ene-3-one (BA) are the most important and representative C19- and C22-steroidal materials. The optimalization of sterol production with mycobacterial phytosterol conversion has been investigated for decades. One of the major challenges is that current industrial mycobacterial strains accumulate unignorable impurities analogous to desired sterol intermediates, significantly hampering product extractions and refinements. Previously, we identified Mycobacterium neoaurum HGMS2 as an efficient 4-AD-producing strain (Wang et al. in Microb Cell Fact. 19:187, 2020). Recently, we have genetically modified the HGMS2 strain to remove its major impurities including ADD and 9OH-AD (Li et al. in Microb Cell Fact. 20:158, 2021). Unexpectedly, the modified mutants started to significantly accumulate BA compared with the HGMS2 strain. In this work, while we attempted to block BA occurrence during 4-AD accumulation in HGMS2 mutants, we identified a few loop pathways that regulated metabolic flux switching between 4-AD and BA accumulations and found that both the 4-AD and BA pathways shared a 9,10-secosteroidial route. One of the key enzymes in the loop pathways was Hsd4A1, which played an important role in determining 4-AD accumulation. The inactivation of the hsd4A1 gene significantly blocked the 4-AD metabolic pathway so that the phytosterol degradation pathway flowed to the BA metabolic pathway, suggesting that the BA metabolic pathway is a complementary pathway to the 4-AD pathway. Thus, knocking out the hsd4A1 gene essentially made the HGMS2 mutant (HGMS2Δhsd4A1) start to efficiently accumulate BA. After further knocking out the endogenous kstd and ksh genes, an HGMS2Δhsd4A1 mutant, HGMS2Δhsd4A1/Δkstd1, enhanced the phytosterol conversion rate to BA in 1.2-fold compared with the HGMS2Δhsd4A1 mutant in pilot-scale fermentation. The final BA yield increased to 38.3 g/L starting with 80 g/L of phytosterols. Furthermore, we knocked in exogenous active kstd or ksh genes to HGMS2Δhsd4A1/Δ kstd1 to construct DBA- and 9OH-BA-producing strains. The resultant DBA- and 9OH-BA-producing strains, HGMS2Δhsd4A1/kstd2 and HGMS2Δkstd1/Δhsd4A1/kshA1B1, efficiently converted phytosterols to DBA- and 9OH-BA with the rates of 42.5% and 40.3%, respectively, and their final yields reached 34.2 and 37.3 g/L, respectively, starting with 80 g/L phytosterols. Overall, our study not only provides efficient strains for the industrial production of BA, DBA and 9OH-BA but also provides insights into the metabolic engineering of the HGMS2 strain to produce other important steroidal compounds.


Asunto(s)
Mycobacterium , Fitosteroles , Fitosteroles/metabolismo , Esteroles/metabolismo , Mycobacterium/genética , Mycobacterium/metabolismo , Esteroides/metabolismo , Redes y Vías Metabólicas , Androstenodiona
7.
J Clin Endocrinol Metab ; 108(2): 331-338, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36201493

RESUMEN

BACKGROUND: Initiating feminizing gender-affirming hormone therapy (GAHT) in transgender women causes a steep decline in serum testosterone. It is unknown if testosterone concentrations change further and whether adrenal androgen levels change during feminizing GAHT and after gonadectomy. This limits clinical decision making in transgender women with symptoms attributed to GAHT or gonadectomy. METHODS: Transgender women (n = 275) initiating estradiol and cyproterone acetate (CPA) were included at baseline, and had follow-up visits after 3 months, 12 months, and 2 to 4 years. During follow-up, 49.5% of transgender women underwent a gonadectomy. Total testosterone (TT), dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEAS), and androstenedione (A4) were measured using liquid chromatography tandem mass spectrometry. RESULTS: After 3 months of GAHT, mean TT, calculated free testosterone (cFT), and A4 decreased by 18.4 nmol/L (95% CI, -19.4 to -17.4, P < 0.001 [ie, -97.1%]), 383 pmol/L (95% CI, -405 to -362, P < 0.001 [ie, -98.3%]), and 1.2 nmol/L (95% CI, -1.4 to -1.0, P < 0.001 [ie, -36.5%]), respectively, and remained stable thereafter. DHEA and DHEAS decreased by 7.4 nmol/L (95% CI, -9.7 to -5.1 [ie, -28.0%]) and 1.8 µmol/L (95% CI, -2.2 to -1.4 [ie, -20.1%]), respectively, after 1 year and did not change thereafter. After gonadectomy, CPA therapy is stopped, which induced no further change in TT, cFT, DHEA, DHEAS, and A4 compared with those who did not undergo gonadectomy. CONCLUSIONS: Our findings confirm that after an initial drop, testosterone levels in transgender women remain stable. Adrenal androgens decrease in the first year of CPA and estrogen supplementation and remain unchanged after gonadectomy. Androgens did not change after gonadectomy and cessation of CPA. Correlates with clinical symptoms remain to be elucidated.


Asunto(s)
Andrógenos , Personas Transgénero , Femenino , Humanos , Testosterona , Androstenodiona , Acetato de Ciproterona/uso terapéutico , Deshidroepiandrosterona , Sulfato de Deshidroepiandrosterona
8.
Arch Womens Ment Health ; 25(5): 911-921, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36074275

RESUMEN

The aim of this study was to determine whether serum androgen levels have an effect on mindfulness and metacognition in adolescents with polycystic ovary syndrome (PCOS). Adolescents diagnosed with PCOS were asked to answer a questionnaire that included socio-demographic information and two scales: the Mindful Attention Awareness Scale (MAAS) and the Metacognition Scale Child and Adolescent form (MCQ-C). The patients were divided into two groups, the hyperandrogenism group and the non-hyperandrogenism group, according to serum androgen levels. The scores of MAAS and MCQ-C were compared between the groups. The study sample consisted of 70 adolescents. Of these, 44 had hyperandrogenism according to a blood test. No statistically significant difference was found in MAAS scores between the hyperandrogenism and the non-hyperandrogenism groups (p = 0.79). However, the level of mindfulness was found to be lower in participants with a higher modified Ferriman-Gallwey score (mFGS) (r = 0.26, p = 0.02). Mindfulness levels were also lower for obese patients with PCOS compared to non-obese patients with PCOS (p = 0.02). Cognitive monitoring (MCQ-C-CM), one of the MCQ-C sub-scales, was significantly higher in the non-hyperandrogenism group (p = 0.03), and similarly, a positive correlation was detected between higher androgen levels and the positive meta-worry (MCQ-C-PM) sub-scale of the MCQ-C (for total testosterone; r = 0.348, p = 0.03, and for androstenedione; r = 0.35, p = 0.03). High serum androgen levels in PCOS had no effect on mindfulness, but as the modified Ferriman Gallwey score increased, mindfulness levels decreased. For the sub-scales of MCQ-C,MCQ-C-CM, and MCQ-C-PM, the scores increased as androgen levels increased. In line with the results of the present study, evaluating mindfulness in PCOS patients with increased hair growth and metacognition in PCOS patients with serum hyperandrogenism may contribute well-being in adulthood by reducing the psychological burden caused by the disease.


Asunto(s)
Hiperandrogenismo , Metacognición , Atención Plena , Síndrome del Ovario Poliquístico , Adolescente , Andrógenos , Androstenodiona , Femenino , Humanos , Hiperandrogenismo/complicaciones , Hiperandrogenismo/diagnóstico , Obesidad , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/diagnóstico , Testosterona
9.
Drug Test Anal ; 14(11-12): 1891-1903, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36001066

RESUMEN

In 2020, the confirmation of the non-endogenous origin of several pseudo-endogenous steroids by means of isotope ratio mass spectrometry (IRMS) was recommended by the World Anti-Doping Agency (WADA), in addition to previously established target analytes for IRMS in sports drug testing. To date, however, IRMS-based methods validated in accordance with current WADA regulations have not been available. Therefore, the aim of this research project was the development and validation of a method to determine the carbon isotope ratios (CIR) of all newly considered pseudo-endogenous steroids, encompassing the anabolic androgenic steroids comprising a 1-ene-core structure (5α-androst-1-ene-3ß,17ß-diol, 5α-androst-1-ene-3,17-dione [1AD], 17ß-hydroxy-5α-androst-1-en-3-one, 3α-hydroxy-5α-androst-1-ene-17-one [1AND], and 3ß-hydroxy-5α-androst-1-ene-17-one [1EpiAND]), as well as steroids referred to as hormone and metabolic modulators (androsta-1,4,6-triene-3,17-dione [TRD] and its main metabolite 17ß-hydroxy-androsta-1,4,6-triene-3-one) and 6α- and 6ß-hydroxy-androst-4-ene-3,17-dione. With peak purity of target analytes being critical for IRMS analyses, a twofold high-performance liquid chromatography (HPLC)-based sample purification was employed, with all analytes being acetylated between the first and second HPLC fractionation. Using established gas chromatography/combustion/IRMS instrumentation, limits of quantification were estimated at 10 ng/ml for a 20 ml urine aliquot for all analytes, except for 1AND (20 ng/ml), and combined measurement uncertainties were estimated between 0.4‰ and 0.9‰. For proof-of-concept, samples collected after the single oral administration of a nutritional supplement containing 1AD and 1EpiAND were analyzed as well as existing excretion study urine samples obtained after the administration of 4-androstenedione and TRD. Based on the obtained results, the developed method was considered to be fit-for-purpose.


Asunto(s)
Androstenodiona , Doping en los Deportes , Esteroides/orina , Cromatografía de Gases y Espectrometría de Masas/métodos , Detección de Abuso de Sustancias/métodos , Isótopos de Carbono
10.
Hormones (Athens) ; 21(4): 545-554, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35841524

RESUMEN

OBJECTIVE: To date, no meta-analysis has been carried out to collect evidence from randomized placebo-controlled trials (RCTs) for the purpose of comprehensively summarizing the effect of androstenedione supplementation. Therefore, the aim of this research was to perform a systematic review and meta-analysis of all RCTs that explored the effect of androstenedione supplementation on individual hormonal, lipid, and anthropometric indices. METHODS: We searched five databases (Web of Science, SCOPUS, Embase, PubMed/MEDLINE, and Google Scholar) using a combination of medical subject headings (MeSH) and non-MeSH terms. Using the random-effects model, we summarized the outcomes as weighted mean difference (WMD) with 95% confidence interval (CI). RESULTS: Eight eligible articles were included in the meta-analysis. The pooled effect sizes suggested a significant effect of androstenedione supplementation on serum estradiol concentrations (WMD: 20.82 ng/ml, 95% CI: 7.25 to 34.38, p = 0.003), triglycerides (TG, WMD: -0.19 mg/dl, 95% CI: - 0.96, 0.57, p = 0.000), and high-density lipoprotein (HDL)-cholesterol (WMD: - 0.13 mg/dl, 95% CI: - 0.23 to - 0.03, p = 0.009); however, it had no effect on testosterone (WMD: 0.098 ng/ml, 95% CI: - 0.499 to 0.696, p = 0.748), body weight (WMD: 0.579 kg, 95% CI: - 4.02 to 5.17, p = 0.805), body mass index (BMI, WMD: - 0.73 kg/m2, 95% CI: - 2.98, 1.50, p = 0.519), low-density lipoprotein (LDL)-cholesterol (WMD: - 0.074 mg/dl, 95% CI: - 0.37 to 0.22, p = 0.622), and total cholesterol (TC, WMD: - 0.15 mg/dl, 95% CI: - 0.49, 0.17, p = 0.198). CONCLUSION: These findings indicate that androstenedione supplementation can lower TG and HDL-cholesterol and increase estradiol concentrations.


Asunto(s)
Androstenodiona , Lípidos , Humanos , Testosterona , Estradiol , Suplementos Dietéticos , Ensayos Clínicos Controlados Aleatorios como Asunto , HDL-Colesterol , Colesterol , Congéneres de la Testosterona , Composición Corporal , Andrógenos
11.
J Steroid Biochem Mol Biol ; 218: 106049, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34990809

RESUMEN

Recent studies have highlighted the potential role of 11oxygenated (keto or hydroxy) androgens in human reproductive function with 11keto androgens circulating at concentrations comparable with testosterone in women and children. However, the intrinsic androgenic bioactivities of 11 keto and hydroxy androgens are not fully characterized. We therefore investigated the full androgen dose-response curves using complementary in vitro yeast and mammalian (HEK293) host cell bioassays of 11 keto and hydroxy derivatives of the potent androgens, testosterone (T) and dihydrotestosterone (DHT), compared with their parent non-11 oxygenated steroids together with the pro-androgen precursor (androstenedione (A4)) and metabolites (androstanedione, androsterone). For potent androgens, the mammalian HEK293 host cell bioassay was 22-138 times more sensitive than the yeast host cell bioassay. In both androgen bioassays, 11keto derivatives displayed androgenic bioactivity but significantly lower molar potency than their parent non-keto steroids. By contrast, the 11hydroxy derivatives had minimal or no androgenic bioactivity. In both bioassays 5α-reduction increased androgenic potency. These findings confirm that that 11keto androgens may contribute directly to androgen status in women, children, and other conditions apart from healthy eugonadal men whereas 11hydroxy androgens have negligible androgenic potency although it cannot be excluded that they may be converted to more potent androgens in vivo.


Asunto(s)
Andrógenos , Saccharomyces cerevisiae , Andrógenos/metabolismo , Androstenodiona/metabolismo , Animales , Niño , Dihidrotestosterona/metabolismo , Dihidrotestosterona/farmacología , Femenino , Células HEK293 , Humanos , Masculino , Mamíferos/metabolismo , Saccharomyces cerevisiae/metabolismo , Esteroides/metabolismo , Testosterona/metabolismo
12.
Molecules ; 26(20)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34684800

RESUMEN

Androstenedione is a steroidal hormone produced in male and female gonads, as well as in the adrenal glands, and it is known for its key role in the production of estrogen and testosterone. Androstenedione is also sold as an oral supplement, that is being utilized to increase testosterone levels. Simply known as "andro" by athletes, it is commonly touted as a natural alternative to anabolic steroids. By boosting testosterone levels, it is thought to be an enhancer for athletic performance, build body muscles, reduce fats, increase energy, maintain healthy RBCs, and increase sexual performance. Nevertheless, several of these effects are not yet scientifically proven. Though commonly used as a supplement for body building, it is listed among performance-enhancing drugs (PEDs) which is banned by the World Anti-Doping Agency, as well as the International Olympic Committee. This review focuses on the action mechanism behind androstenedione's health effects, and further side effects including clinical features, populations at risk, pharmacokinetics, metabolism, and toxicokinetics. A review of androstenedione regulation in drug doping is also presented.


Asunto(s)
Androstenodiona/farmacología , Anabolizantes/farmacología , Androstenodiona/metabolismo , Androstenodiona/toxicidad , Animales , Atletas , Rendimiento Atlético , Suplementos Dietéticos/toxicidad , Doping en los Deportes , Femenino , Humanos , Masculino , Factores Sexuales , Testosterona/metabolismo
13.
Anim Reprod Sci ; 229: 106760, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33962315

RESUMEN

Effects of nutrition on insulin-like growth factor-I (IGF-I), IGF binding proteins (IGFBP), and insulin in plasma and dominant follicles were evaluated at day 72 and 56 (Exp. 1, n = 12 and Exp. 2, n = 28, respectively) postpartum in anovulatory primiparous beef cows. Cows were stratified based on body condition score at calving and randomly assigned to nutritional treatments: maintain (M), 2.27 kg of a 40 % CP supplement per day and ad libitum hay; or gain (G), ad libitum access to a 50 % concentrate diet and ad libitum hay. Blood samples were collected twice weekly starting 30 days postpartum. Ovarian follicles were evaluated using ultrasonography commencing 42 (Exp. 1) or 30 (Exp. 2) days postpartum. Body weight and condition score were greater (P < 0.05) for cows of G than M groups and postpartum interval to luteal function was longer for cows of the M than G group. Insulin and IGF-I concentrations in follicular fluid (FF) and plasma were greater (P < 0.05) for cows of the G than M group at follicular aspiration. Plasma and FF IGFBP4 and IGFBP5 concentrations were greater (P <  0.05) in Exp. 2, and IGFBP5 was greater in Exp. 1 for cows of the G than M group. Treatment did not affect FF steroid concentrations or granulosal cell CYP19A1, PAPPA, IGFBP4, and IGFBP5 mRNA abundance. These results indicate concentrations of IGF-I, insulin, IGFBP4, and IGFBP5 in FF and plasma are affected by nutritional intake and may be related to follicular function.


Asunto(s)
Bovinos/fisiología , Dieta/veterinaria , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Folículo Ovárico/efectos de los fármacos , Periodo Posparto , Somatomedinas/metabolismo , Androstenodiona/química , Androstenodiona/metabolismo , Alimentación Animal/análisis , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Composición Corporal , Peso Corporal , Bovinos/sangre , Estradiol/química , Estradiol/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Folículo Ovárico/metabolismo , Progesterona/química , Progesterona/metabolismo , Somatomedinas/genética
14.
J Obstet Gynaecol ; 41(5): 684-689, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32811221

RESUMEN

The aim of this systematic review is to assess the effect of Berberine (BBR) on women's health to provide greater insights about its effect on women with polycystic syndrome for both patients and health care providers. Electronic databases such as PubMed, Web of Science, Scopus, Cochrane Central Register of Controlled Trials (CENTRAL) were systematically searched from the base to July 1th, 2019 to identify clinical trials and randomised controlled trials that had explored the effect of BBR on the polycystic syndrome. With regard to the weight and composition body, BBR did not have any significant effect on reducing body weight and conflicting findings had been reported about waist circumference (WC) and body mass index (BMI). However, BBR led to a significant decrease in waist to hip ratio (WHR), profile hormonal insulin resistance (IR), and insulin resistance (HOMA-IR). Further, androstenedione dropped significantly following treatment with BBB. However, BBB did not have a significant effect on follicle stimulating hormone (FSH) and luteinizing hormone (LH).


Asunto(s)
Berberina/uso terapéutico , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Adulto , Androstenodiona/sangre , Índice de Masa Corporal , Femenino , Hormona Folículo Estimulante/sangre , Humanos , Insulina/análisis , Resistencia a la Insulina , Hormona Luteinizante/sangre , Síndrome del Ovario Poliquístico/sangre , Resultado del Tratamiento , Circunferencia de la Cintura/efectos de los fármacos , Relación Cintura-Cadera
15.
Steroids ; 165: 108758, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33161054

RESUMEN

There is some evidence that marketable supplements contain hormones not declared on the product label. The presence of these androgenic anabolic steroids (AAS) in sports supplements can be considered an adulteration and affect the health of consumers, who are predominantly athletes. This study aimed to measure anabolic hormones (methyltestosterone and 4-androstenedione) in sport supplements. Ultra Performance Liquid chromatography coupled mass spectrometry (UPLC-MS/MS) with electrospray ionization (ESI) in positive mode was employed under the Multiple Reaction Monitoring (MRM) ion program. To overcome matrix effects and quantify the selected analyte, the calibration curve was made using Matrix Match method. The LOQ and LOD were 1 ng/g and 0.3 ng/g for both analytes. The recovery of 4-androstenedione and methyltestosterone was in the range of 86.87-107.35 and 77.31-113.98, respectively. In terms of reproducibility, CV % for 4-androstenedione and methyltestosterone ranged from 6.56 to 16.87% and 1.45-15.12%, respectively. 4-androstenedione was found in 11 samples including 9 whey as 1.578 ±â€¯0.154 ng/g and 2 whey albumin samples with an amount of 1.134 ng/g and 1.474 ng/g. Consequently, continuous controlling of sport supplements comprising intentionally or unintentionally added androgens could be important for health and discuss in the context of compliance with anti-doping.


Asunto(s)
Androstenodiona , Doping en los Deportes , Metiltestosterona , Reproducibilidad de los Resultados
16.
Mol Med Rep ; 22(5): 3645-3658, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32901880

RESUMEN

Eurycoma (E.) longifolia Jack (Tongkat Ali) is a widely applied medicine that has been reported to boost serum testosterone and increase muscle mass. However, its actual biological targets and effects on an in vitro level remain poorly understood. Therefore, the present study aimed to investigate the effects of a standardised E. longifolia extract (F2) on the growth and its associated gene expression profile in mouse Leydig cells. F2, even at lower doses, was found to induce a high level of testosterone by ELISA. The level was as high as the levels induced by eurycomanone and formestane in Leydig cells. However, Leydig cells treated with F2 demonstrated reduced viability, which was likely due to the diminished cell population at the G0/G1 phase and increased cell population arrested at the S phase in the cell cycle, as assessed by MTT assay and flow cytometry, respectively. Cell viability was revived when the treatment time­point was prolonged to 96 h. Genome­wide gene analysis by reverse transcription­quantitative PCR of F2­treated Leydig cells at 72 h, when the cell growth was not revived, and 96 h, when the cell growth had started to revive, revealed cyclin­dependent kinase­like 2 (CDKL2) to be a potential target in regulating the viability of F2­treated Leydig cells. Functional analysis, as analysed using GeneMANIA Cytoscape program v.3.6.0 (https://genemania.org/), further suggested that CDKL2 could act in concert with Casitas B­lineage lymphoma and sphingosine kinase 1 interactor­A­kinase anchoring protein domain­containing genes to regulate the viability of F2­treated Leydig cells. The findings of the present study provide new insights regarding the potential molecular targets associated with the biological effect of E. longifolia extract on cell growth, particularly on the cell cycle, which could aid in enhancing the bioefficacy and reducing the toxicity of this natural product in the future.


Asunto(s)
Eurycoma/química , Redes Reguladoras de Genes/efectos de los fármacos , Células Intersticiales del Testículo/efectos de los fármacos , Fitoquímicos/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Androstenodiona/análogos & derivados , Androstenodiona/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/genética , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-cbl/genética , Testosterona/metabolismo
17.
Biochemistry ; 59(33): 2999-3009, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32786398

RESUMEN

Aromatase (CYP19A1) catalyzes the synthesis of estrogens from androgens and is an invaluable target of pharmacotherapy for estrogen-dependent cancers. CYP19A1 is also one of the most primordial human CYPs and, to the extent that its fundamental dynamics are conserved, is highly relevant to understanding those of the more recently evolved and promiscuous enzymes. A complementary approach employing molecular dynamics simulations and hydrogen-deuterium exchange mass spectrometry (HDX-MS) was employed to interrogate the changes in CYP19A1 dynamics coupled to binding androstenedione (ASD). Gaussian-accelerated molecular dynamics and HDX-MS agree that ASD globally suppresses CYP19A1 dynamics. Bimodal HDX patterns of the B'-C loop potentially arising from at least two conformations are present in free 19A1 only, supporting the possibility that conformational selection is operative. Random-acceleration molecular dynamics and adaptive biasing force simulations illuminate ASD's binding pathway, predicting ASD capture in the lipid headgroups and a pathway to the active site shielded from solvent. Intriguingly, the predicted access channel in 19A1 aligns well with the steroid binding sites of other human sterol-oxidizing CYPs.


Asunto(s)
Androstenodiona/farmacocinética , Aromatasa/química , Aromatasa/metabolismo , Membranas/metabolismo , Androstenodiona/metabolismo , Dominio Catalítico , Medición de Intercambio de Deuterio , Humanos , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Membranas/química , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica
18.
Steroids ; 162: 108679, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32569733

RESUMEN

Although the discovery of antibiotics has decreased the spread and severity of infectious diseases, their uncontrolled use has lead to the emergence of bacterial resistance to existing chemotherapeutic agents. Bacterial disease thus remains a challenge for health authorities in worldwide and especially in sub-Saharan Africa. Despite their efficacy, the miss-use of medicinal plants for the treatment of infectious diseases couple to the farming and hunting activities has contribute enormously to the destruction of many medicinal plant species. In search of an alternative for new and effective agents against bacterial infection, norandrostenedion (19-nor-4-androsten-3,17-dione) (1), was biotransformed by Cunninghamella blakesleeana ATCC 8688A and yielded a new metabolite, 6α,10 ß -dihydroxy-19-nor-4-androsten-3-one (2), together with three known compounds, 10 ß -hydroxy-19-nor-4-androsten-3,17-dione (3), 6 ß,10 ß,17 ß -trihydroxy-19-nor-4-androsten-3-one (4) and 10 ß,17 ß -dihydroxy-19-nor-4-androsten-3-one (5). Their structures were elucidated on the basis ofspectroscopic techniques: NMR analysis (1D and 2D) and HRIE-MS and by comparison with previously reported data. In addition, the agar diffusion method was used to evaluate the diameter of the inhibition zone and INT colorimetric assay for MIC values. All metabolites obtained showed a potent and varied activity against tested bacteria. These results support the uses of biotransformation to develop new antimicrobial compounds for clinical application.


Asunto(s)
Androstenodiona/análogos & derivados , Antibacterianos/metabolismo , Antibacterianos/farmacología , Cunninghamella/metabolismo , Androstenodiona/química , Androstenodiona/metabolismo , Androstenodiona/farmacología , Antibacterianos/química , Biotransformación , Modelos Moleculares , Conformación Molecular , Estereoisomerismo
19.
Int J Food Sci Nutr ; 71(5): 644-652, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31914834

RESUMEN

A randomised, double-blind and placebo-controlled trial was performed to examine the effects of whole soy and isoflavone daidzein on serum androgenic hormones in Chinese equol-producing post-menopausal women. A total of 270 eligible women aged 45-70 years were randomised to either one of the three iso-caloric supplements: 40 g soy flour (whole soy group), 40 g low-fat milk powder +63 mg daidzein (daidzein group) or 40 g low-fat milk powder (placebo group) daily for 6 months. Fasting venous samples were tested for serum androstenedione (AD), testosterone (T), prolactin, sex hormone binding globulin and dehydroepiandrosterone sulphate. Intention-to-treat analysis indicated that serum T (p = .022) and AD (p = .05) levels modestly but significantly decreased after 6-month daidzein treatment in comparison with placebo, with a mean difference of -0.057 nmol/L (95%CI: -0.185 to 0.070, p = .018) and -0.118 ng/mL (95%CI: -0.240-0.004, p = .045), respectively. This 6-month trial suggested that purified daidzein may exhibit less androgenic effect.Trial registration: The trial was registered in ClinicalTrials.gov with identifier of NCT01270737. (URL: http://clinicaltrials.gov/ct2/show/NCT01270737.).


Asunto(s)
Androstenodiona/sangre , Glycine max/química , Isoflavonas/farmacología , Fitoestrógenos/farmacología , Extractos Vegetales/farmacología , Alimentos de Soja , Testosterona/sangre , Andrógenos/sangre , China , Método Doble Ciego , Equol/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Posmenopausia
20.
Endocrinology ; 160(1): 143-155, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30418546

RESUMEN

Polycystic ovary syndrome (PCOS) results from functional ovarian hyperandrogenism due to dysregulation of androgen secretion. Cultured theca cells from polycystic ovaries of women with the most common form of PCOS overexpress most androgen producing enzymes, particularly CYP450c17. In this study, a murine model was used of PCOS induced by chronic feeding with a high-fat diet that exhibits the reproductive, hyperandrogenic, and metabolic constellation of PCOS symptoms seen in women. Oral administration of KDT501, a hops-derived bitter taste receptor (Tas2R 108) isohumulone ligand resulted in resolution of PCOS-associated endocrine and metabolic disturbances and restored reproductive function. Pioglitazone, a PPARγ agonist, also improved metabolic and reproductive function, though not to the same degree as KDT501. Specifically, treatment of the murine PCOS model with KDT501 resulted in reduced testosterone and androstenedione levels in the absence of significant changes in LH or FSH, improved glucose tolerance and lipid metabolism, and reduced hepatic lipid infiltration and adiposity. There was significant improvement in estrous cyclicity and an increase in the number of ovarian corpora lutea, indicative of improved reproductive function after exposure to KDT501. Finally, ex vivo exposure of murine ovaries to KDT501 attenuated androgen production and ovarian expression of CYP450c17. Interestingly, the ovaries expressed Tas2R 108, suggesting a potential regulation of ovarian steroidogenesis through this chemosensory receptor family. In summary, a therapeutic strategy for PCOS possibly could include direct influences on ovarian steroidogenesis that are independent of gonadotrophic hormone regulation.


Asunto(s)
Extractos Vegetales/administración & dosificación , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Adiposidad/efectos de los fármacos , Androstenodiona/metabolismo , Animales , Familia 17 del Citocromo P450/genética , Familia 17 del Citocromo P450/metabolismo , Modelos Animales de Enfermedad , Ciclo Estral/efectos de los fármacos , Femenino , Humanos , Humulus/química , Ligandos , Ratones , Ratones Endogámicos C57BL , Extractos Vegetales/química , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/fisiopatología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA