Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
2.
Clin Endocrinol (Oxf) ; 87(1): 59-67, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28370068

ABSTRACT

OBJECTIVE: Serum sex steroid concentrations may alter body composition and glucose homoeostasis in men in a dose-response manner. We evaluated these end-points in healthy men rendered medically castrate through use of a gonadotrophin-releasing hormone antagonist (acyline) with incremental doses of exogenous testosterone (T) gel. DESIGN: Subjects (n=6-9 per group) were randomly assigned to injections of acyline every 2 weeks plus transdermal T gel (1.25 g, 2.5 g, 5.0 g, 10 g or 15 g) daily or double placebo (injections and gel) for 12 weeks. PATIENTS: Healthy men, ages 25-55 years, with normal serum total T concentrations. MEASUREMENTS: Serum T, dihydrotestosterone (DHT) and oestradiol (E2) were measured at baseline and every 2 weeks. Body composition was analysed by dual-energy X-ray absorptiometry at baseline and week 12. Fasting serum adiponectin, leptin, glucose and insulin concentrations were measured at baseline and week 10. RESULTS: Forty-eight men completed the study. A significant treatment effect was observed for change in lean mass (ANOVAP=.01) but not fat mass (P=.14). Lean mass increased in the 15 g T group relative to all lower dose groups, except the 10 g T group. When all subjects were analysed together, changes in lean mass correlated directly and changes in fat mass correlated inversely with serum T, E2 and DHT. No changes were noted in serum glucose, insulin or adipokine levels. CONCLUSIONS: In healthy men, higher serum concentrations of T, DHT and E2 were associated with greater increases in lean mass and decreases in fat mass but not with changes in serum glucose, insulin or adipokines.


Subject(s)
Adipokines/blood , Body Composition/drug effects , Gonadal Steroid Hormones/administration & dosage , Testosterone/administration & dosage , Adult , Blood Glucose , Dihydrotestosterone/blood , Dose-Response Relationship, Drug , Estradiol/blood , Gonadal Steroid Hormones/blood , Healthy Volunteers , Hormone Antagonists , Humans , Insulin/blood , Male , Middle Aged , Oligopeptides/administration & dosage , Testosterone/blood
3.
J Clin Endocrinol Metab ; 101(7): 2937-44, 2016 07.
Article in English | MEDLINE | ID: mdl-27172434

ABSTRACT

CONTEXT: Concern exists that T replacement therapy (TRT) might increase the risk of prostate disease. There are limited data regarding the impact of TRT on prostate androgen concentrations. OBJECTIVE: Determine the dose-dependent effects of exogenous T administration on intraprostatic androgen concentrations. DESIGN: Twelve-week, double-blinded, randomized, placebo-controlled trial. SETTING: Academic medical center. PARTICIPANTS: Sixty-two healthy eugonadal men, aged 25-55 years. INTERVENTIONS: Subjects were randomly assigned to receive injections of acyline, a GnRH antagonist (used to achieve medical castration), every 2 weeks plus transdermal T gel (1.25 g, 2.5 g, 5.0 g, 10 g, or 15 g daily), or placebo injections and transdermal gel for 12 weeks. MAIN OUTCOMES: Serum T and dihydrotestosterone (DHT) were measured at baseline and every 2 weeks during treatment. Intraprostatic T and DHT concentrations were assessed from tissue obtained through ultrasound-guided prostate needle biopsies at week 12. Androgens were quantified by liquid chromatography-tandem mass spectrometry. RESULTS: 51 men completed the study and were included in the analysis. There were no significant adverse events. Exogenous T resulted in a dose-dependent increase in serum T and DHT concentrations (190-770 and 60-180 ng/dL, respectively). Although intraprostatic T differed among dose groups (P = .01), intraprostatic DHT was comparable regardless of T dose (P = .11) and was 10- to 20-fold greater than intraprostatic T. CONCLUSIONS: In healthy, medically castrate men receiving exogenous T, the total intraprostatic androgen concentration (predominantly DHT) remained stable across serum T concentrations within the physiological range. These findings further our knowledge of the relationship between serum and intraprostatic androgens and suggest that physiological serum T achieved by TRT is unlikely to alter the prostate hormonal milieu.


Subject(s)
Dihydrotestosterone/metabolism , Oligopeptides/administration & dosage , Orchiectomy , Prostate/metabolism , Testosterone/therapeutic use , Adult , Gonadotropin-Releasing Hormone/agonists , Health , Hormone Replacement Therapy/adverse effects , Humans , Male , Middle Aged , Orchiectomy/methods , Prostate/drug effects , Testosterone/pharmacology , Treatment Outcome
4.
Clin Endocrinol (Oxf) ; 83(6): 828-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25777143

ABSTRACT

OBJECTIVE: Total testosterone concentrations are influenced by sex hormone-binding globulin (SHBG) concentrations, which are decreased by obesity and increased with ageing. Therefore, we sought to understand and compare the associations of ageing and obesity with SHBG. DESIGN: We performed a retrospective, cross-sectional analysis of the associations of obesity and age on SHBG and testosterone measurements in men being evaluated for hypogonadism. PATIENTS, MEASUREMENTS AND ANALYSIS: A total of 3671 men who underwent laboratory testing for testosterone deficiency from the Veterans Administration Puget Sound Health Care System from 1997 through 2007 was included. Univariate and multivariate linear regression modelling of the associations between age and body mass index (BMI) and SHBG was performed. RESULTS: Obesity was associated with a significantly lower SHBG [ß = -1·26 (95% CI -1·14, -1·38) nmol/l] per unit increase in BMI. In contrast, ageing was associated with a significantly increased SHBG [ß = 0·46 (95% CI 0·39, 0·53) nmol/l per year] (P < 0·001 for both effects). The association of obesity with lower SHBG was two to three times larger than the association of ageing with increased SHBG in both univariate and multivariate modelling. On average, obese men (BMI >30 kg/m(2)) had significantly lower SHBG and total testosterone concentrations than nonobese men [(mean ± SD) SHBG: 36 ± 22 vs 50 ± 27 nmol/l and total testosterone: 10·5 ± 5·4 nmol/l vs 14·1 ± 7·4 nmol/l; (P < 0·001 for both comparisons)], but calculated free testosterone concentrations did not differ between obese and nonobese men. CONCLUSIONS: We found that the association between obesity and lowered SHBG is greater than the association of ageing with increased SHBG. These competing effects may impact total testosterone measurements for the diagnosis of low testosterone, particularly in obese men.


Subject(s)
Aging/metabolism , Obesity/metabolism , Sex Hormone-Binding Globulin/metabolism , Adult , Aged , Aged, 80 and over , Aging/blood , Aging/physiology , Body Mass Index , Cross-Sectional Studies , Humans , Male , Middle Aged , Obesity/blood , Retrospective Studies , Testosterone/blood , Young Adult
5.
Asian J Androl ; 16(2): 248-55, 2014.
Article in English | MEDLINE | ID: mdl-24407178

ABSTRACT

A growing body of literature has established the anabolic benefi ts of testosterone (T) therapy in hypogonadal men. However, there remains a paucity of data regarding the risks of exogenous androgen use in older men and the potential for adverse effects on the prostate gland. Whether T therapy in older, hypogonadal men might worsen lower urinary tract symptoms or exacerbate, unmask, or even incite prostate cancer development has tempered enthusiasm for T therapy, while known prostatic disease has served as a relative contraindication to T therapy. Androgens are necessary for the development and maintenance of the prostate gland. However, epidemiologic studies do not consistently fi nd a positive relationship between endogenous serum androgen concentrations and the risk of prostate disease. Recent data demonstrate that 5α-reductase inhibitors decrease the risk of low-grade prostate cancer, suggesting that modifying androgen metabolism may have beneficial effects on prostate health, yet similar reductions in high-grade disease have not been observed, thereby questioning the true clinical benefits of these agents for chemoprevention. Knowing how to best investigate the relationship between androgens and the development of prostate disease given the lack of large, randomized trials is difficult. Accumulating data challenges the assumption that alterations in serum androgens have parallel effects within the prostate hormonal environment or change androgen-regulated processes within the gland. Long-term intervention studies are needed to truly ascertain the effects of androgen manipulation on prostate tissue and disease risk. However, available data do not support the notion that restoring serum androgens to normal physiologic ranges drives prostate disease.


Subject(s)
Androgens/physiology , Prostatic Diseases/physiopathology , Prostatic Neoplasms/physiopathology , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...