Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 555
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Physiol Sci ; 74(1): 22, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38561673

RESUMO

Androgen excess and metabolic abnormality largely contribute to the pathogenesis of polycystic ovarian syndrome (PCOS), which primarily precipitates ovarian dysfunction and infertility in reproductive-age women. Impaired mitochondrial function and epigenetic alteration have been linked to the development of PCOS. However, it is unknown whether acetate would exert a therapeutic effect on ovarian mitochondrial dysfunction in PCOS. Herein, the study hypothesized that acetate reverses ovarian mitochondrial dysfunction in experimental PCOS rat model, possibly through modulation of mitofusin-2 (MFn2). Eight-week-old female Wistar rats were randomized into four groups (n = 5). Induction of PCOS was performed by 1 mg/kg letrozole (p.o.), administered for 21 days. Thereafter, the rats were treated with acetate (200 mg/kg; p.o.) for 6 weeks. The PCOS rats demonstrated androgen excess, multiple ovarian cysts, elevated anti-mullerian hormone and leptin and decreased SHBG, adiponectin and 17-ß estradiol with corresponding increase in ovarian transforming growth factor-ß1. Additionally, inflammation (tumor growth factor and nuclear factor-kB), elevated caspase-6, decreased hypoxia-inducible factor-1α and elevated histone deacetylase-2 (HDAC2) were observed in the ovaries of PCOS rats, while mitochondrial abnormality with evidence of decreased adenosine triphosphate synthase and MFn2 was observed in rats with PCOS. Treatment with acetate reversed the alterations. The present results collectively suggest that acetate ameliorates ovarian mitochondrial abnormality, a beneficial effect that is accompanied by MFn2 with consequent normalization of reproductive-endocrine profile and ovarian function. Perhaps, the present data provide hope for PCOS individuals that suffer infertility.


Assuntos
Infertilidade , Doenças Mitocondriais , Síndrome do Ovário Policístico , Humanos , Feminino , Ratos , Animais , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo , Letrozol/efeitos adversos , Androgênios/efeitos adversos , Ratos Wistar , Infertilidade/complicações , Mitocôndrias/metabolismo , Acetatos/efeitos adversos
2.
Clin Neurol Neurosurg ; 239: 108217, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38452714

RESUMO

INTRODUCTION: Meningiomas frequently occur within the field of neuro-oncology, but it is unclear whether exogenous or imbalanced endogenous hormones are involved in the pathophysiology. A previous case-control study found an almost 20-fold increase in the risk of developing meningioma among users of androgenic anabolic steroids. We, therefore, investigated this hypothesis. METHODS: We compared the incidence rate of meningioma in a cohort of males sanctioned for the use of androgenic anabolic steroids with age- and sex-matched controls with an identical enrollment date. RESULTS: We followed 1189 males sanctioned for using androgenic anabolic steroids for a total of 13,305 person-years and found 0 cases of meningioma. The control cohort of 59,450 males was followed for a total of 654,938 person-years, and 16 were diagnosed with meningioma. Thus, the incidence rate ratio was 0 (95% CI: 0-12.8). CONCLUSION: We did not find any evidence supporting the hypothesis of an increased risk of meningioma development with the use of androgenic anabolic steroids. Due to the limited sample size, we cannot exclude androgenic anabolic steroids as a potential risk factor for meningioma development, despite the lack of apparent evidence in this study.


Assuntos
Anabolizantes , Neoplasias Meníngeas , Meningioma , Masculino , Humanos , Androgênios/efeitos adversos , Estudos de Coortes , Meningioma/induzido quimicamente , Meningioma/epidemiologia , Esteróides Androgênicos Anabolizantes , Anabolizantes/efeitos adversos , Neoplasias Meníngeas/induzido quimicamente , Neoplasias Meníngeas/epidemiologia
3.
Sci Rep ; 14(1): 563, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-38177175

RESUMO

Polycystic ovary syndrome (PCOS) is associated with symptoms of moderate to severe anxiety and depression. Hyperandrogenism is a key feature together with lower levels of the adipocyte hormone adiponectin. Androgen exposure leads to anxiety-like behavior in female offspring while adiponectin is reported to be anxiolytic. Here we test the hypothesis that elevated adiponectin levels protect against the development of androgen-induced anxiety-like behavior. Pregnant mice overexpressing adiponectin (APNtg) and wildtypes were injected with vehicle or dihydrotestosterone to induce prenatal androgenization (PNA) in the offspring. Metabolic profiling and behavioral tests were performed in 4-month-old female offspring. PNA offspring spent more time in the closed arms of the elevated plus maze, indicating anxiety-like behavior. Intriguingly, neither maternal nor offspring adiponectin overexpression prevented an anxiety-like behavior in PNA-exposed offspring. However, adiponectin overexpression in dams had metabolic imprinting effects, shown as lower fat mass and glucose levels in their offspring. While serum adiponectin levels were elevated in APNtg mice, cerebrospinal fluid levels were similar between genotypes. Adiponectin overexpression improved metabolic functions but did not elicit anxiolytic effects in PNA-exposed offspring. These observations might be attributed to increased circulating but unchanged cerebrospinal fluid adiponectin levels in APNtg mice. Thus, increased adiponectin levels in the brain are likely needed to stimulate anxiolytic effects.


Assuntos
Ansiolíticos , Síndrome do Ovário Policístico , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Humanos , Camundongos , Feminino , Animais , Síndrome do Ovário Policístico/metabolismo , Androgênios/efeitos adversos , Adiponectina , Ansiolíticos/efeitos adversos , Ansiedade/metabolismo , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente
5.
J Endocrinol Invest ; 47(3): 603-617, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37642904

RESUMO

PURPOSE: Maternal hyperandrogenism during pregnancy is associated with adverse gestational outcomes and chronic non-communicable diseases in offspring. However, few studies are reported to demonstrate the association between maternal androgen excess and cardiac health in offspring. This study aimed to explore the relation between androgen exposure in utero and cardiac health of offspring in fetal and adult period. Its underlying mechanism is also illustrated in this research. METHODS: Pregnant mice were injected with dihydrotestosterone (DHT) from gestational day (GD) 16.5 to GD18.5. On GD18.5, fetal heart tissue was collected for metabolite and morphological analysis. The hearts from adult offspring were also collected for morphological and qPCR analysis. H9c2 cells were treated with 75 µM androsterone. Immunofluorescence, flow cytometry, qPCR, and western blot were performed to observe cell proliferation and explore the underlying mechanism. RESULTS: Intrauterine exposure to excessive androgen led to thinner ventricular wall, decreased number of cardiomyocytes in fetal offspring and caused cardiac hypertrophy, compromised cardiac function in adult offspring. The analysis of steroid hormone metabolites in fetal heart tissue by ultra performance liquid chromatography and tandem mass spectrometry showed that the content of androgen metabolite androsterone was significantly increased. Mechanistically, H9c2 cells treated with androsterone led to a significant decrease in phosphorylated retinoblastoma protein (pRB) and cell cycle-related protein including cyclin-dependent kinase 2 (CDK2), cyclin-dependent kinase 4 (CDK4), and cyclin D1 (CCND1) in cardiomyocytes. This resulted in cell cycle arrest at G1-S phase, which in turn inhibited cardiomyocyte proliferation. CONCLUSION: Taken together, our results indicate that in utero exposure to DHT, its metabolite androsterone could directly decrease cardiomyocytes proliferation through cell cycle arrest, which has a life-long-lasting effect on cardiac health. Our study highlights the importance of monitoring sex hormones in women during pregnancy and the follow-up of cardiac function in offspring with high risk of intrauterine androgen exposure.


Assuntos
Androgênios , Miócitos Cardíacos , Humanos , Adulto , Gravidez , Feminino , Animais , Camundongos , Androgênios/efeitos adversos , Androsterona , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Proteínas de Ciclo Celular , Di-Hidrotestosterona , Cardiomegalia/induzido quimicamente
7.
Urologia ; 90(4): 715-719, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37606191

RESUMO

OBJECTIVE: Evaluate the association between AGD and the risk of PCa. METHODS: Incident case-control study, conducted on 153 patients. All patients underwent prostate biopsy for abnormal digital rectal exam and/or prostate antigen elevation. Two variants of AGD [anus to scrotum base (AGDas) and anus to dorsal insertion of penis (AGDap) were measured. Student's t-test was used to analyze continuous variables and chi-square for discrete variables. Differences in AGD (raw and adjusted) measures between cases and controls were assessed using the ANCOVA test. RESULTS: There is significant association between the diagnose of PCa and both AGD, patients diagnosed with PCa had both AGD measures shortened, in the crude and the full adjusted models. CONCLUSIONS: This research suggests that the same gestational environmental exposures, could represent a weak androgen signaling and may also have an increased risk of prostate cancer.


Assuntos
Androgênios , Neoplasias da Próstata , Masculino , Humanos , Androgênios/efeitos adversos , Estudos de Casos e Controles , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/diagnóstico , Pênis , Biomarcadores , Canal Anal
9.
Reprod Sci ; 30(11): 3179-3189, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37380913

RESUMO

Hyperandrogenism is an endocrine disorder affecting a large population of reproductive-aged women, thus proportionally high number of fetuses are subjected to prenatal androgenic exposure (PNA). The short-term stimulations at critical ontogenetic stages can wield lasting influences on the health. The most commonly diagnosed conditions in reproductive age women is polycystic ovary syndrome (PCOS). PNA may affect the growth and development of many systems in the whole body and disrupts the normal metabolic trajectory in the offspring of PCOS, contributing to the prevalence of cardiovascular and metabolic diseases (CVMD), including myocardial hypertrophy, hypertension, hyperinsulinemia, insulin resistance, hyperglycemia, obesity, and dyslipidemia, which are the leading causes of hospitalizations in young PCOS offspring. In this review, we focus on the effects of prenatal androgenic exposure on the cardiovascular and metabolic diseases in offspring, discuss the possible pathogenesis respectively, and summarize potential management strategies to improve metabolic health of PCOS offspring. It is expected that the incidence of CVMD and the medical burden will be reduced in the future.


Assuntos
Doenças Cardiovasculares , Hiperandrogenismo , Resistência à Insulina , Doenças Metabólicas , Síndrome do Ovário Policístico , Gravidez , Feminino , Humanos , Adulto , Síndrome do Ovário Policístico/diagnóstico , Androgênios/efeitos adversos , Hiperandrogenismo/complicações , Doenças Metabólicas/etiologia , Doenças Metabólicas/complicações , Obesidade/complicações , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/etiologia
10.
Neuroendocrinology ; 113(12): 1193-1214, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37356425

RESUMO

Hormones are important regulators of key processes during fetal brain development. Thus, the developing brain is vulnerable to the action of chemicals that can interfere with endocrine signals. Epidemiological studies have pointed toward sexually dimorphic associations between neurodevelopmental outcomes, such as cognitive abilities, in children and prenatal exposure to endocrine-disrupting chemicals (EDCs). This points toward disruption of sex steroid signaling in the development of neural structures underlying cognitive functions, such as the hippocampus, an essential mediator of learning and memory processes. Indeed, during development, the hippocampus is subjected to the organizational effects of estrogens and androgens, which influence hippocampal cell proliferation, differentiation, dendritic growth, and synaptogenesis in the hippocampal fields of Cornu Ammonis and the dentate gyrus. These early organizational effects correlate with a sexual dimorphism in spatial cognition and are subject to exogenous chemical perturbations. This review summarizes the current knowledge about the organizational effects of estrogens and androgens on the developing hippocampus and the evidence for hippocampal-dependent learning and memory perturbations induced by developmental exposure to EDCs. We conclude that, while it is clear that sex hormone signaling plays a significant role during hippocampal development, a complete picture at the molecular and cellular levels would be needed to establish causative links between the endocrine modes of action exerted by EDCs and the adverse outcomes these chemicals can induce at the organism level.


Assuntos
Androgênios , Disruptores Endócrinos , Criança , Humanos , Androgênios/efeitos adversos , Disruptores Endócrinos/toxicidade , Estrogênios/farmacologia , Transdução de Sinais , Encéfalo
11.
Phytother Res ; 37(9): 4018-4041, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37165686

RESUMO

Polycystic ovary syndrome (PCOS) is most common in women of reproductive age, giving rise to androgen excess and anovulation, leading to infertility and non-reproductive complications. We explored the ameliorating effect of naringenin in PCOS using the Sprague Dawley (SD) rat model and human granulosa cells. Letrozole-induced PCOS rats were given either naringenin (50 mg/kg/day) alone or in combination with metformin (300 mg/kg/day), followed by the estrous cycle, hormonal analysis, and glucose sensitivity test. To evaluate the effect of naringenin on granulosa cell (hGC) steroidogenesis, we treated cells with naringenin (2.5 µM) alone or in combination with metformin (1 mM) in the presence of forskolin (10 µM). To determine the steroidogenesis of CYP-17A1, -19A1, and 3ßHSD2, the protein expression levels were examined. Treatment with naringenin in the PCOS animal groups increased ovulation potential and decreased cystic follicles and levels of androgens. The expression levels of CYP-17A1, -19A1, and 3ßHSD2, were seen restored in the ovary of PCOS SD rats' model and in the human ovarian cells in response to the naringenin. We found an increased expression level of phosphorylated-AKT in the ovary and hGCs by naringenin. Naringenin improves ovulation and suppress androgens and cystic follicles, involving AKT activation.


Assuntos
Cisto Folicular , Metformina , Síndrome do Ovário Policístico , Humanos , Feminino , Ratos , Animais , Androgênios/efeitos adversos , Ratos Sprague-Dawley , Letrozol/efeitos adversos , Proteínas Proto-Oncogênicas c-akt , Cisto Folicular/complicações , Modelos Animais de Doenças
12.
Eur Urol Focus ; 9(3): 409-410, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37005166

RESUMO

There are varied associations that have been identified between the use of androgen deprivation therapy (ADT) and the development of cognitive decline. We highlight the first studies to evaluate chronic use of ADT, other systemic treatments for prostate cancer, and genetic polymorphisms in this context.


Assuntos
Disfunção Cognitiva , Neoplasias da Próstata , Masculino , Humanos , Neoplasias da Próstata/tratamento farmacológico , Antagonistas de Androgênios/efeitos adversos , Androgênios/efeitos adversos , Disfunção Cognitiva/induzido quimicamente
13.
Transl Res ; 256: 1-13, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36586536

RESUMO

Polycystic ovary syndrome (PCOS), characterized by the androgen excess and arrest of antral follicles, is a common endocrine disorder among women lacking specific diagnostic biomarkers and therapeutic targets. Herein, we studied the molecular mechanism of miR-96-5p in the process of PCOS and its potential applications in PCOS. Clinically, we found that miR-96-5p significantly decreased in serum, follicular fluid and primary human granulosa cells (hGCs) of PCOS patients (n = 70) vs non-PCOS women (n = 60), as well as in the ovaries of 3-types of induced PCOS-like mice. Furthermore, we demonstrated that the elevated circulating miR-96-5p levels were significantly correlated with the PCOS disordered endocrine clinical features, and the area under the curve of receiver operating characteristic was 0.8344, with 75.71% specificity and 80% sensitivity. Mechanically, we identified miR-96-5p as an androgen-regulated miRNA that directly targets the forkhead transcription factor FOXO1. Inhibition of miR-96-5p decreased estrogen synthesis, while decreasing the cell proliferation index of KGN via regulating the expression of FOXO1 and its downstream genes. Inversely, inhibition of FOXO1 abrogated the effect of miR-96-5p on estrogen synthesis and proliferation index. Of note, ovarian intra-bursal injection of miR-96-5p agomir rescued the phenotypes of dehydroepiandrosterone-induced PCOS like mice. In conclusion, our results clarified a vital role of miR-96-5p in the pathogenesis of PCOS and might serve as a novel diagnostic biomarker and therapeutic target for PCOS.


Assuntos
MicroRNAs , Síndrome do Ovário Policístico , Humanos , Feminino , Camundongos , Animais , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/terapia , Androgênios/efeitos adversos , Androgênios/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Células da Granulosa/metabolismo , Estrogênios
14.
Eur Urol ; 83(1): 70-81, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36273937

RESUMO

BACKGROUND: Bladder cancer (BCa) is more common in men and presents differences in molecular subtypes based on sex. Fibroblast growth factor receptor 3 (FGFR3) mutations are enriched in the luminal papillary muscle-invasive BCa (MIBC) and non-MIBC subtypes. OBJECTIVE: To determine whether FGFR3 mutations initiate BCa and impact BCa male sex bias. DESIGN, SETTING, AND PARTICIPANTS: We developed a transgenic mouse model expressing the most frequent FGFR3 mutation, FGFR3-S249C, in urothelial cells. Bladder tumorigenesis was monitored in transgenic mice, with and without carcinogen exposure. Mouse and human BCa transcriptomic data were compared. INTERVENTION: Mutant FGFR3 overexpression in mouse urothelium and siRNA knockdown in cell lines, and N-butyl-N(4-hydroxybutyl)-nitrosamine (BBN) exposure. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Impact of transgene dosage on tumor frequency, synergy with BBN treatment, and FGFR3 pathway activation were analyzed. The sex-specific incidence of FGFR3-mutated tumors was evaluated in mice and humans. FGFR3 expression in FGFR3-S249C mouse urothelium and in various human epithelia was measured. Mutant FGFR3 regulation of androgen (AR) and estrogen (ESR1) receptor activity was evaluated, through target gene expression (regulon) and reporter assays. RESULTS AND LIMITATIONS: FGFR3-S249C expression in mice induced low-grade papillary BCa resembling human luminal counterpart at histological, genomic, and transcriptomic levels, and promoted BBN-induced basal BCa formation. Mutant FGFR3 expression levels impacted tumor incidence in mice, and mutant FGFR3-driven human tumors were restricted to epithelia presenting high normal FGFR3 expression levels. BCa male sex bias, also found in our model, was even higher in human FGFR3-mutated tumors compared with wild-type tumors and was associated with higher AR and lower ESR1 regulon activity. Mutant FGFR3 expression inhibited both ESR1 and AR activity in mouse tumors and human cell lines, demonstrating causation only between FGFR3 activation and low ESR1 activity in tumors. CONCLUSIONS: Mutant FGFR3 initiates luminal papillary BCa formation and favors BCa male sex bias, potentially through FGFR3-dependent ESR1 downregulation. Patients with premalignant lesions or early-stage BCa could thus potentially benefit from FGFR3 targeting. FGFR3 expression level in epithelia could account for FGFR3-driven carcinoma tissue specificity. PATIENT SUMMARY: By developing a transgenic mouse model, we showed that gain-of-function mutations of FGFR3 receptor, among the most frequent genetic alterations in bladder cancer (BCa), initiate BCa formation. Our results could support noninvasive detection of FGFR3 mutations and FGFR3 targeting in early-stage bladder lesions.


Assuntos
Receptor Tipo 3 de Fator de Crescimento de Fibroblastos , Neoplasias da Bexiga Urinária , Feminino , Humanos , Masculino , Camundongos , Animais , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Bexiga Urinária/patologia , Sexismo , Neoplasias da Bexiga Urinária/patologia , Mutação , Camundongos Transgênicos , Androgênios/efeitos adversos
15.
World J Gastroenterol ; 28(26): 3071-3080, 2022 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-36051334

RESUMO

Anabolic androgenic steroids (AASs) are a group of molecules including endogenous testosterone and synthetic derivatives that have both androgenic and anabolic effects. These properties make them therapeutically beneficial in medical conditions such as hypogonadism. However, they are commonly bought illegally and misused for their anabolic, skeletal muscle building, and performance-enhancing effects. Supraphysiologic and long-term use of AASs affects all organs, leading to cardiovascular, neurological, endocrine, gastrointestinal, renal, and hematologic disorders. Hepatotoxicity is one of the major concerns regarding AASs treatment and abuse. Testosterone and its derivatives have been most often shown to induce a specific form of cholestasis, peliosis hepatis, and hepatic benign and malignant tumors. It is currently believed that mechanisms of pathogenesis of these disorders include disturbance of antioxidative factors, upregulation of bile acid synthesis, and induction of hepatocyte hyperplasia. Most toxicity cases are treated with supportive measures and liver function normalizes with discontinuation of AAS. However, some long-term consequences are irreversible. AAS-induced liver injury should be taken in consideration in patients with liver disorders, especially with the increasing unintentional ingestion of supplements containing AAS. In this paper, we review the most current knowledge about AAS-associated adverse effects on the liver, and their clinical presentations, prevalence, and pathophysiological mechanisms.


Assuntos
Anabolizantes , Doença Hepática Crônica Induzida por Substâncias e Drogas , Anabolizantes/efeitos adversos , Androgênios/efeitos adversos , Humanos , Testosterona , Congêneres da Testosterona/efeitos adversos
16.
Curr Opin Endocrinol Diabetes Obes ; 29(6): 560-565, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-35938779

RESUMO

PURPOSE OF REVIEW: Data on the health effects of androgen abuse are mainly derived from lower level evidence, such as case series and cross-sectional studies. In the last few years a relatively large and prospective cohort initiative, the HAARLEM (health risks of anabolic androgenic steroid use by male amateur athletes) study, made an important contribution to current knowledge. RECENT FINDINGS: The HAARLEM study showed that all androgen abusers experience positive and negative effects, such as an increase in strength and acne and gynecomastia, respectively. Effects are generally reversible and acute life-threatening toxicity is rare. There is a distinct but limited impact on liver and kidney function. Gonadal function is disrupted but resumes normally after abuse is discontinued in the majority of athletes. The negative impact of androgens on cardiovascular parameters, such as blood pressure, hematocrit and lipid metabolism, as well as cardiac structure and function, seems to be the mechanism for premature atherosclerosis and cardiomyopathy, respectively, in long-term users. SUMMARY: It is beyond dispute that androgen abuse is harmful and much of the short-term toxicity is well documented. To prevent the long-term health hazards, there should be ample focus on preventive measures, both primary and secondary, and effective harm reduction strategies should be developed.


Assuntos
Anabolizantes , Androgênios , Masculino , Humanos , Androgênios/efeitos adversos , Estudos Transversais , Estudos Prospectivos , Esteroides
17.
Best Pract Res Clin Endocrinol Metab ; 36(5): 101686, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35981955

RESUMO

Male hypogonadism is associated with reduced quality of life and the development of co-morbidities including obesity, diabetes mellitus, and dyslipidaemia. The mainstay of treatment for male hypogonadism is testosterone replacement therapy (TRT). However, TRT has recognised side effects including impaired spermatogenesis and there are concerns regarding its use in men with concurrent cardiovascular disease. Thus, there has been an impetus to develop novel androgen therapies for treating male hypogonadism to mitigate the side effects of TRT. This review will discuss the benefits and adverse effects of TRT, and novel therapies including nasal testosterone, aromatase inhibitors, selective oestrogen receptor modulators, and selective androgen receptor modulators.


Assuntos
Antagonistas de Receptores de Andrógenos , Androgênios , Inibidores da Aromatase , Terapia de Reposição Hormonal , Hipogonadismo , Moduladores Seletivos de Receptor Estrogênico , Antagonistas de Receptores de Andrógenos/efeitos adversos , Antagonistas de Receptores de Andrógenos/uso terapêutico , Androgênios/efeitos adversos , Androgênios/uso terapêutico , Inibidores da Aromatase/efeitos adversos , Inibidores da Aromatase/uso terapêutico , Humanos , Hipogonadismo/tratamento farmacológico , Masculino , Qualidade de Vida , Receptores Androgênicos , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Testosterona
18.
Fertil Steril ; 118(2): 371-381, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35710599

RESUMO

OBJECTIVE: To study the efficacy and safety of the GLP-1 analog liraglutide 3 mg (LIRA 3 mg) vs. placebo (PL) for reduction of body weight (BW) and hyperandrogenism in women with obesity and polycystic ovary syndrome (PCOS). DESIGN: Randomized, double-blind, placebo-controlled trial. SETTING: Hospital-based outpatient endocrine and metabolic center. PATIENT(S): Women diagnosed with PCOS (NIH criteria) were randomly assigned to LIRA 3 mg (n = 55) or PL (n = 27) once daily for 32 weeks with lifestyle intervention. INTERVENTION(S): Study visits at baseline and 32 weeks included BW and body composition by dual-energy x-ray absorptiometry. Oral glucose tolerance tests were done with sex steroids, free androgen index (FAI), and lipids measured in the fasting sample. MAIN OUTCOME MEASURE(S): The primary end points were changes in BW and FAI. Safety was assessed in all patients who received at least one dose of the study drug. RESULT(S): Change in BW from baseline to week 32 was -5.7% (±0.75) with LIRA 3 mg vs. -1.4% (±1.09) with PL. At week 32, more participants on LIRA 3 mg than on PL achieved at least 5% weight reductions (25 of 44 vs. 5 of 23). Free androgen index significantly reduced with LIRA 3 mg compared with the PL where the mean FAI slightly increased. Gastrointestinal events, which were mostly mild to moderate, were reported in 58.2% of the LIRA 3 mg-subjects and 18.5% of PL subjects. CONCLUSION(S): LIRA 3 mg once daily appears superior to PL in reducing BW and androgenicity and improving cardiometabolic parameters in women with PCOS and obesity. CLINICAL TRIAL REGISTRATION NUMBER: NCT03480022.


Assuntos
Resistência à Insulina , Síndrome do Ovário Policístico , Androgênios/efeitos adversos , Composição Corporal , Feminino , Humanos , Liraglutida/efeitos adversos , Obesidade/complicações , Obesidade/diagnóstico , Obesidade/tratamento farmacológico , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/diagnóstico , Síndrome do Ovário Policístico/tratamento farmacológico
19.
Best Pract Res Clin Endocrinol Metab ; 36(5): 101653, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35414435

RESUMO

In this review, we discuss the effects on androgens on the haemopoietic system, focussing largely on the effects of testosterone on erythropoiesis. Stimulation of erythropoiesis is one of the most consistent effects of testosterone treatment observed in clinical trials. In men with anaemia this effect can be beneficial. Conversely, erythrocytosis is one of the most common adverse effects of testosterone treatment with a relative risk of 8.14 (95% CI: 1.87-35.40) estimated by a recent meta-analysis of randomised placebo controlled clinical trials. A reduction in haemoglobin is commonly seen in men receiving androgen deprivation therapy for prostate cancer, and in transwomen receiving gender affirming therapy to reduce serum testosterone. While mechanisms by which androgens regulate erythropoiesis are not fully understood, it is likely that effects on erythropoietic progenitor cells and erythropoietin are involved, with secondary effects on iron metabolism. In contrast, whether androgens exert clinically relevant effects on white blood cells and on platelets requires further study.


Assuntos
Eritropoetina , Neoplasias da Próstata , Antagonistas de Androgênios/uso terapêutico , Androgênios/efeitos adversos , Eritropoetina/uso terapêutico , Hemoglobinas/metabolismo , Hemoglobinas/uso terapêutico , Humanos , Ferro/uso terapêutico , Masculino , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/tratamento farmacológico , Testosterona/efeitos adversos , Testosterona/uso terapêutico
20.
Best Pract Res Clin Endocrinol Metab ; 36(5): 101628, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35248487

RESUMO

Prostate cancer is one of the most frequently diagnosed malignancies in men worldwide and the life expectancy for men with prostate cancer is improving due to advancements in diagnostics and treatment. Male hypogonadism is associated with obesity, diabetes, and other comorbidities and also has been linked with increasing age; the primary therapy modality for this condition is testosterone replacement therapy (TRT). There are concerns that testosterone therapy may cause prostate cancer disease progression. However, contemporary evidence suggests that testosterone replacement therapy may be safe in specific groups of patients with prostate cancer. This chapter will summarise the contemporary literature regarding TRT use in hypogonadal men with prostate cancer, including limitations and future research goals.


Assuntos
Hipogonadismo , Neoplasias da Próstata , Androgênios/efeitos adversos , Terapia de Reposição Hormonal/efeitos adversos , Humanos , Hipogonadismo/complicações , Hipogonadismo/tratamento farmacológico , Masculino , Neoplasias da Próstata/complicações , Neoplasias da Próstata/tratamento farmacológico , Testosterona/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA