Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.807
Filtrar
1.
Nutrients ; 16(12)2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38931170

RESUMEN

Androgen production primarily occurs in Leydig cells located in the interstitial compartment of the testis. In aging males, testosterone is crucial for maintaining muscle mass and strength, bone density, sexual function, metabolic health, energy levels, cognitive function, as well as overall well-being. As men age, testosterone production by Leydig cells of the testes begins to decline at a rate of approximately 1% per year starting from their 30s. This review highlights recent findings concerning the use of natural polyphenolics compounds, such as flavonoids, resveratrol, and phenolic acids, to enhance testosterone production, thereby preventing age-related degenerative conditions associated with testosterone insufficiency. Interestingly, most of the natural polyphenolic antioxidants having beneficial effects on testosterone production tend to enhance the expression of the steroidogenic acute regulatory protein (Star) gene in Leydig cells. The STAR protein facilitates the entry of the steroid precursor cholesterol inside mitochondria, a rate-limiting step for androgen biosynthesis. Natural polyphenolic compounds can also improve the activities of steroidogenic enzymes, hypothalamus-pituitary gland axis signaling, and testosterone bioavailability. Thus, many polyphenolic compounds such as luteolin, quercetin, resveratrol, ferulic acid phenethyl ester or gigantol may be promising in delaying the initiation of late-onset hypogonadism accompanying aging in males.


Asunto(s)
Antioxidantes , Hipogonadismo , Polifenoles , Testosterona , Masculino , Humanos , Hipogonadismo/tratamiento farmacológico , Antioxidantes/farmacología , Polifenoles/farmacología , Testosterona/metabolismo , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Animales , Envejecimiento/efectos de los fármacos , Fosfoproteínas/metabolismo , Resveratrol/farmacología
2.
JBJS Rev ; 12(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38889232

RESUMEN

¼ Testosterone replacement treatment (TRT) and anabolic androgenic steroid (AAS) use is common and possibly increasing.¼ Diagnosing and treating hypogonadism in men is controversial.¼ Hypogonadism and the use of AASs seem to have a detrimental effect on the musculoskeletal system. The current literature on TRT and the musculoskeletal system shows an increased risk of tendon injury.¼ There may be a role for testosterone supplementation in the postoperative period.


Asunto(s)
Terapia de Reemplazo de Hormonas , Hipogonadismo , Testosterona , Humanos , Testosterona/uso terapéutico , Testosterona/efectos adversos , Masculino , Terapia de Reemplazo de Hormonas/efectos adversos , Hipogonadismo/tratamiento farmacológico , Cirujanos Ortopédicos , Andrógenos/efectos adversos , Andrógenos/uso terapéutico
3.
Am Fam Physician ; 109(6): 543-549, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38905552

RESUMEN

Testosterone deficiency, or male hypogonadism, is a clinical syndrome that can be defined as persistently low serum testosterone levels in the setting of symptoms consistent with testosterone deficiency. Studies suggest that testosterone replacement therapy may improve sexual function, depressive symptoms, bone density, and lean body mass. Evidence is conflicting regarding its effect on cardiovascular events and mortality. Although prior studies suggested that testosterone replacement therapy increased the risk of cardiovascular disease, a large, randomized trial showed that it does not increase the risk of myocardial infarction or stroke, even in patients at high risk. After a detailed discussion of the potential benefits and risks through shared decision-making, testosterone replacement therapy should be considered for men with testosterone deficiency to correct selected symptoms and induce and maintain secondary sex characteristics. Treatment method should take into consideration patient preference, pharmacokinetics, potential for medication interactions, formulation-specific adverse effects, treatment burden, and cost. Clinicians should monitor men receiving testosterone replacement therapy for symptom improvement, potential adverse effects, and adherence. Serum testosterone, hematocrit, and prostate-specific antigen levels should be measured at baseline and at least annually in men 40 years or older receiving testosterone replacement therapy. (Am Fam Physician. 2024;109(6):543-549.


Asunto(s)
Terapia de Reemplazo de Hormonas , Hipogonadismo , Testosterona , Humanos , Masculino , Testosterona/uso terapéutico , Testosterona/sangre , Testosterona/efectos adversos , Terapia de Reemplazo de Hormonas/métodos , Hipogonadismo/tratamiento farmacológico , Persona de Mediana Edad , Adulto
4.
Arch Endocrinol Metab ; 68: e230101, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38739523

RESUMEN

Objective: Both pulsatile gonadotropin-releasing hormone (GnRH) and combined gonadotropin therapy are effective to induce spermatogenesis in men with congenital hypogonadotropic hypogonadism (CHH). This study aimed to evaluate the effect of pulsatile GnRH therapy on spermatogenesis in male patients with CHH who had poor response to combined gonadotropin therapy. Materials and methods: Patients who had poor response to combined gonadotropin therapy ≥ 6 months were recruited and shifted to pulsatile GnRH therapy. The rate of successful spermatogenesis, the median time to achieve spermatogenesis, serum gonadotropins, testosterone, and testicular volume were used for data analysis. Results: A total of 28 CHH patients who had poor response to combined gonadotropin (HCG/HMG) therapy for 12.5 (6.0, 17.75) months were recruited and switched to pulsatile GnRH therapy for 10.0 (7.25, 16.0) months. Sperm was detected in 17/28 patients (60.7%). The mean time for the appearance of sperm in semen was 12.0 (7.5, 17.5) months. Compared to those who could not achieve spermatogenesis during pulsatile GnRH therapy, the successful group had a higher level of LH60min (4.32 vs. 1.10 IU/L, P = 0.043) and FSH60min (4.28 vs. 1.90 IU/L, P = 0.021). Testicular size increased during pulsatile GnRH therapy, compared to previous HCG/ HMG therapy (P < 0.05). Conclusion: For CHH patients with prior poor response to one year of HCG/ HMG therapy, switching to pulsatile GnRH therapy may induce spermatogenesis.


Asunto(s)
Hormona Liberadora de Gonadotropina , Hipogonadismo , Espermatogénesis , Testosterona , Humanos , Masculino , Espermatogénesis/efectos de los fármacos , Hormona Liberadora de Gonadotropina/administración & dosificación , Hipogonadismo/tratamiento farmacológico , Adulto , Testosterona/administración & dosificación , Testosterona/sangre , Testosterona/uso terapéutico , Adulto Joven , Resultado del Tratamiento , Gonadotropina Coriónica/administración & dosificación , Gonadotropina Coriónica/uso terapéutico , Menotropinas/administración & dosificación , Menotropinas/uso terapéutico , Testículo/efectos de los fármacos , Quimioterapia Combinada , Quimioterapia por Pulso , Adolescente
5.
South Med J ; 117(5): 245-253, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38701845

RESUMEN

Androgen deprivation therapy is the cornerstone of systemic management for prostate cancer but is associated with multiple adverse effects that must be considered during treatment. These effects occur because of the profound hypogonadism that is induced from lack of testosterone or due to the medications used in the treatment or in combination with androgen receptor signaling inhibitors. This article critically reviews the associations between androgen deprivation therapy, androgen receptor signaling inhibitors, and cardiovascular complications such as prolonged QT interval, atrial fibrillation, heart failure, atherosclerosis, coronary heart disease, venous thromboembolism, and peripheral arterial occlusive disease. These unfavorable outcomes reinforce the need for regular cardiovascular screening of patients undergoing androgen deprivation for the management of prostate cancer.


Asunto(s)
Antagonistas de Andrógenos , Enfermedades Cardiovasculares , Neoplasias de la Próstata , Humanos , Masculino , Antagonistas de Andrógenos/efectos adversos , Antagonistas de Andrógenos/uso terapéutico , Antagonistas de Receptores Androgénicos/uso terapéutico , Antagonistas de Receptores Androgénicos/efectos adversos , Enfermedades Cardiovasculares/prevención & control , Enfermedades Cardiovasculares/etiología , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/fisiopatología , Neoplasias de la Próstata/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos
6.
Biomed Pharmacother ; 175: 116700, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38703505

RESUMEN

Late-onset hypogonadism (LOH) is an age-related disease in men characterized by decreased testosterone levels with symptoms such as decreased libido, erectile dysfunction, and depression. Thymus quinquecostatus Celakovski (TQC) is a plant used as a volatile oil in traditional medicine, and its bioactive compounds have anti-inflammatory potential. Based on this knowledge, the present study aimed to investigate the effects of TQC extract (TE) on LOH in TM3 Leydig cells and in an in vivo aging mouse model. The aqueous extract of T. quinquecostatus Celakovski (12.5, 25, and 50 µg/mL concentrations) was used to measure parameters such as cell viability, testosterone level, body weight, and gene expression, via in vivo studies. Interestingly, TE increased testosterone levels in TM3 cells in a dose-dependent manner without affecting cell viability. Furthermore, TE significantly increased the expression of genes involved in the cytochrome P450 family (Cyp11a1, Cyp17a1, Cyp19a1, and Srd5a2), which regulate testosterone biosynthesis. In aging mouse models, TE increased testosterone levels without affecting body weight and testicular tissue weight tissue of an aging animal group. In addition, the high-dose TE-treated group (50 mg/kg) showed significantly increased expression of the cytochrome p450 enzymes, similar to the in vitro results. Furthermore, HPLC-MS analysis confirmed the presence of caffeic acid and rosmarinic acid as bioactive compounds in TE. Thus, the results obtained in the present study confirmed that TQC and its bioactive compounds can be used for LOH treatment to enhance testosterone production.


Asunto(s)
Envejecimiento , Extractos Vegetales , Testículo , Testosterona , Thymus (Planta) , Animales , Testosterona/sangre , Masculino , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Ratones , Extractos Vegetales/farmacología , Testículo/efectos de los fármacos , Testículo/metabolismo , Thymus (Planta)/química , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Supervivencia Celular/efectos de los fármacos , Línea Celular , Hipogonadismo/tratamiento farmacológico , Modelos Animales de Enfermedad
7.
Aging Male ; 27(1): 2357548, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38812251

RESUMEN

OBJECTIVE: We evaluated change (Δ) in AMSS in men with adult-onset testosterone deficiency (TD) on/not on testosterone undecanoate (TU) by analysing a registry of men with adult-onset TD. METHODS: Analyses were performed using non-parametric statistics to determine ΔAMSS at 6-12 monthly intervals in men on/not on TU and movement in AMSS. Factors predicting ΔAMSS were established via linear/multiple regression. RESULTS: TU was significantly associated with lower AMSS values compared with that at baseline/prior assessment during the initial 42 months treatment; 259 of the 260 men showed improvement. In the 361 men not on TU, AMSS values increased during 60 months of follow-up compared with that at baseline/prior assessment; improvement after 60 months was evident in 1 man, whilst AMSS remained the same or worsened in 213 and 147 men, respectively. In men on TU, baseline AMSS was inversely associated with ΔAMSS (R2 = 0.97), with no other factors reaching significance. Baseline AMSS, age, serum total testosterone (TT), waist circumference (WC), and diastolic blood pressure (BP) were associated with ΔAMSS in men not on TU. DISCUSSION: We show that TU was associated with lower AMSS in men with adult-onset TD whilst non-treatment led to increased values. Baseline AMSS values inversely predicted ΔAMSS in both groups.


Asunto(s)
Testosterona , Humanos , Masculino , Testosterona/deficiencia , Testosterona/sangre , Testosterona/análogos & derivados , Testosterona/uso terapéutico , Testosterona/administración & dosificación , Persona de Mediana Edad , Anciano , Terapia de Reemplazo de Hormonas/métodos , Adulto , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/sangre , Sistema de Registros , Envejecimiento/fisiología
8.
Growth Horm IGF Res ; 76: 101593, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38669801

RESUMEN

Prader-Willi syndrome (PWS) is a rare genetic disorder typically characterized by body composition abnormalities, hyperphagia, behavioral challenges, cognitive dysfunction, and hormone deficiencies. Hypogonadism is common but knowledge on potential side effects of testosterone replacement is limited, in particular, the long-term effects on behavior and PSA. PATIENTS AND METHODS: Retrospective case studies of seven men, median age 46 years, with genetically verified PWS, testosterone treated hypogonadism and available PSA values were included. Long-term follow-up of PSA was accessible in four patients. Medical records were reviewed for adverse effects. RESULTS: Five men were treated with intramuscular testosterone undecanoate, two had no hypogonadism. Median PSA was 0.68 µg/L (0.23-1.3), median testosterone 15 nmol/L. After a median time of 17 years of testosterone replacement median PSA was 0.75 µg/L (range 0.46-1.4). Testosterone replacement was well tolerated, and no major behavioral changes were reported. Five were treated with growth hormone for >20 years. CONCLUSION: Levels of PSA were low. Long-term treatment with testosterone was working well and did not result in any clinically meaningful increase in PSA. Our results indicate that testosterone replacement is neither associated with serious adverse events regarding changes in behavior or effect on PSA. However, larger studies are needed to confirm our results.


Asunto(s)
Terapia de Reemplazo de Hormonas , Hipogonadismo , Síndrome de Prader-Willi , Antígeno Prostático Específico , Testosterona , Humanos , Masculino , Síndrome de Prader-Willi/tratamiento farmacológico , Síndrome de Prader-Willi/sangre , Antígeno Prostático Específico/sangre , Testosterona/análogos & derivados , Testosterona/administración & dosificación , Testosterona/efectos adversos , Testosterona/uso terapéutico , Estudios Retrospectivos , Persona de Mediana Edad , Adulto , Hipogonadismo/tratamiento farmacológico , Estudios de Seguimiento
9.
J Appl Physiol (1985) ; 136(6): 1460-1467, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38634506

RESUMEN

Chronic testosterone (T) substitution and short-term T administration positively affect protein metabolism, however, data on acute effects in humans are sparse. This study aimed to investigate T's acute effects on whole body protein metabolism in hypogonadal and eugonadal conditions. We designed a randomized, double-blind, placebo-controlled, crossover study, including 12 healthy young males. Whole body protein metabolism was evaluated during 1) eugonadism, and after medically induced hypogonadism, with application of a gel on each trial day containing either 2) placebo, 3) T 50 mg, or 4) T 150 mg; under basal (5-h basal period) and insulin-stimulated conditions (3-h clamp). The main outcome measure was a change in net protein balance. The net protein loss was 62% larger in the placebo-treated hypogonadal state compared with the eugonadal state during the basal period (-5.5 ± 3.5 µmol/kg/h vs. -3.4 ± 1.2 µmol/kg/h, P = 0.038), but not during the clamp (P = 0.06). Also, hypogonadism resulted in a 25% increase in whole body urea flux (P = 0.006). However, T did not result in any significant changes in protein breakdown, synthesis, or net balance during either the basal period or clamp (all P > 0.05). Protein breakdown was reduced during clamp compared with the basal period regardless of gonadal status or T exposure (all P ≤ 0.001). In conclusion, the application of transdermal T did not counteract the negative effects of hypogonadism with no effects on protein metabolism within 5 h of administration. Insulin (during clamp) mitigated the effects of hypogonadism. This study is the first to investigate acute protein metabolic effects of T in hypogonadal men.NEW & NOTEWORTHY In a model of medically induced hypogonadism in male volunteers, we found increased whole body urea flux and net protein loss as an expected consequence of hypogonadism. Our study demonstrates the novel finding that the application of transdermal testosterone had no acute effects on whole body protein metabolism under eugonadal conditions, nor could it mitigate the hypogonadism-induced changes in protein metabolism. In contrast, insulin (during clamp) mitigated the effects of hypogonadism on protein metabolism.


Asunto(s)
Estudios Cruzados , Hipogonadismo , Testosterona , Humanos , Masculino , Testosterona/administración & dosificación , Testosterona/metabolismo , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/metabolismo , Método Doble Ciego , Adulto , Adulto Joven , Proteínas/metabolismo , Insulina/metabolismo
10.
J Investig Med ; 72(5): 487-491, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38644376

RESUMEN

Oral calorie intake causes an acute and transient decline in serum testosterone concentrations. It is not known whether this decline occurs in men on testosterone therapy. In this study, we evaluated the change in testosterone concentrations following oral glucose ingestion in hypogonadal men before and after treatment with testosterone therapy. This is a secondary analysis of samples previously collected from a study of hypogonadal men with type 2 diabetes who received testosterone therapy. Study participants (n = 14) ingested 75 grams of oral glucose, and blood samples were collected over 2 h. The test was repeated after 23 weeks of intramuscular testosterone therapy. The mean age and body mass index of study volunteers were 53 ± 8 years and 38 ± 7 kg/m2, respectively. Following glucose intake, testosterone concentrations fell significantly prior to testosterone therapy (week 0, p = 0.04). The nadir of testosterone concentration was at 1 h, followed by recovery to baseline by 2 h. In contrast, there was no change in testosterone concentrations at week 23. The change in serum testosterone concentrations at 60 min was significantly more at week 0 than week 23 (-11 ± 10% vs 0 ± 16%, p = 0.05). We conclude that oral glucose intake has no impact on testosterone concentrations in men on testosterone therapy. Endocrinology societies should consider clarifying in their recommendations that fasting testosterone concentrations are required for the diagnosis of hypogonadism, but not for monitoring testosterone therapy.


Asunto(s)
Glucosa , Testosterona , Humanos , Testosterona/sangre , Masculino , Persona de Mediana Edad , Glucosa/metabolismo , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/sangre , Administración Oral , Adulto
11.
Endocrinol Metab Clin North Am ; 53(2): 307-320, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677872

RESUMEN

Managing patients unable to produce sex steroids using gonadotropins to mimic minipuberty in hypogonadotropic hypogonadism, or sex steroids in patients with Klinefelter or Turner syndrome, is promising. There is a need to pursue research in this area, with large prospective cohorts and long-term data before these treatments can be routinely considered.


Asunto(s)
Hipogonadismo , Síndrome de Klinefelter , Síndrome de Turner , Humanos , Síndrome de Turner/tratamiento farmacológico , Síndrome de Turner/complicaciones , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/etiología , Síndrome de Klinefelter/complicaciones , Síndrome de Klinefelter/tratamiento farmacológico , Lactante , Masculino , Preescolar , Femenino , Terapia de Reemplazo de Hormonas/métodos , Niño , Gonadotropinas/uso terapéutico
14.
J Paediatr Child Health ; 60(2-3): 53-57, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38572627

RESUMEN

AIM: Hormone replacement therapy with testosterone for pubertal induction in boys with congenital hypogonadotropic hypogonadism (CHH) achieves virilization but not spermatogenesis. By contrast, human chorionic gonadotropin (hCG) and recombinant follicle stimulating hormone (rFSH) provides both virilization and spermatogenesis. Fertility outcomes of boys treated with recombinant therapy during adolescence have been infrequently described. We report fertility induction and pregnancy outcomes in CHH patients treated with recombinant gonadotropins during puberty. METHODS: Data of six subjects with CHH (n = 3 Kallmann syndrome & n = 3 Isolated hypogonadotropic hypogonadism) treated with hCG and FSH for pubertal induction were reviewed. Of these, five underwent subsequent fertility induction while one desired fertility at the end of pubertal induction. RESULTS: Partners of all subjects achieved pregnancies using hCG and rFSH, all with full term live births. All infants were clinically normal. CONCLUSION: This study provides early evidence of proof of concept of use of gonadotropin induction of puberty being beneficial in subsequent fertility outcome.


Asunto(s)
Gonadotropina Coriónica , Hipogonadismo , Adulto , Embarazo , Lactante , Femenino , Adolescente , Humanos , Masculino , Gonadotropina Coriónica/uso terapéutico , Hipogonadismo/tratamiento farmacológico , Hormona Folículo Estimulante , Testosterona/uso terapéutico , Fertilidad , Proteínas Recombinantes/uso terapéutico , Pubertad , Virilismo
17.
Acta Med Indones ; 56(1): 1-2, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38561883

RESUMEN

Hypogonadism is a condition characterized by diminished or absent production of sex hormones by the testicles in men and the ovaries in women. Hypogonadism is classified into primary and secondary hypogonadism. Each type of hypogonadism can be caused by congenital and acquired factors. There are many factors that contribute to the occurrence of hypogonadism, including genetic and developmental disorders, infection, kidney disease, liver disease, autoimmune disorders, chemotherapy, radiation, surgery, and trauma. This represents the considerable challenge in diagnosing hypogonadism.The goals of treatment include restore sexual functionality and well-being, initiating and sustaining virilization, osteoporosis prevention, normalize growth hormone levels in elderly men if possible, and restoring fertility in instances of hypogonadotropic hypogonadism. The main approach to treating hypogonadism is hormone replacement therapy. Male with prostate cancer, breast cancer, and untreated prolactinoma are contraindicated for hormone replacement therapy. When selecting a type of testosterone therapy for male with hypogonadism, several factors need to be considered, such as the diversity of treatment response and the  type of testosterone formulation. The duration of therapy depends on individual response, therapeutic goals, signs and symptoms, and hormonal levels. The response to testosterone therapy is evaluated based on symptoms and signs as well as improvements in hormone profiles in the blood. Endocrine Society Clinical Practice Guideline recommend therapeutic goals based on the alleviation of symptoms and signs, as well as reaching testosterone levels between 400 - 700 ng/dL (one week after administering testosterone enanthate or cypionate) and maintaining baseline hematocrit.Hormone therapy is the primary modality in the management of hypogonadism. The variety of signs and symptoms makes early diagnosis of this condition challenging. Moreover, administering hypogonadism therapy involves numerous considerations influenced by various patient factors and the potential for adverse effects. This poses a challenge for physicians to provide targeted hypogonadism therapy with minimal complications.


Asunto(s)
Hipogonadismo , Humanos , Masculino , Femenino , Anciano , Hipogonadismo/diagnóstico , Hipogonadismo/tratamiento farmacológico , Testosterona/uso terapéutico , Testículo , Terapia de Reemplazo de Hormonas/efectos adversos
18.
Lancet Diabetes Endocrinol ; 12(4): 257-266, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38437850

RESUMEN

BACKGROUND: Although some male patients with congenital hypogonadotropic hypogonadism (CHH) undergo spontaneous reversal following treatment, predictors of reversal remain elusive. We aimed to assemble the largest cohort of male patients with CHH reversal to date and identify distinct classes of reversal. METHODS: This multicentre cross-sectional study was conducted in six international CHH referral centres in Brazil, Finland, France, Italy, the UK, and the USA. Adult men with CHH (ie, absent or incomplete spontaneous puberty by age 18 years, low serum testosterone concentrations, and no identifiable cause of hypothalamic-pituitary-gonadal [HPG] axis dysfunction) were eligible for inclusion. CHH reversal was defined as spontaneous recovery of HPG axis function off treatment. Centres provided common data elements on patient phenotype, clinical assessment, and genetics using a structured, harmonised data collection form developed by COST Action BM1105. Latent class mixture modelling (LCMM) was applied to establish whether at least two distinct classes of reversal could be identified and differentially predicted, and results were compared with a cohort of patients without CHH reversal to identify potential predictors of reversal. The primary outcome was the presence of at least two distinct classes of reversal. FINDINGS: A total of 87 male patients with CHH reversal and 108 without CHH reversal were included in the analyses. LCMM identified two distinct reversal classes (75 [86%] in class 1 and 12 [14%] in class 2) on the basis of mean testicular volume, micropenis, and serum follicle-stimulating hormone (FSH) concentration. Classification probabilities were robust (0·998 for class 1 and 0·838 for class 2) and modelling uncertainty was low (entropy 0·90). Compared with class 1, patients in class 2 had significantly larger testicular volume (p<0·0001), no micropenis, and higher serum FSH concentrations (p=0·041), consistent with the Pasqualini syndrome (fertile eunuch) subtype of CHH. Patients without CHH reversal were more likely to have anosmia (p=0·016), cryptorchidism (p=0·0012), complete absence of puberty (testicular volume <4 cm³; p=0·0016), and two or more rare genetic variants (ie, oligogenicity; p=0·0001). Among patients who underwent genetic testing, no patients (of 75) with CHH reversal had a rare pathogenic ANOS1 variant compared with ten (11%) of 95 patients without CHH reversal. Individuals with CHH reversal had a significantly higher rate of rare variants in GNRHR than did those without reversal (nine [12%] of 75 vs three [3%] of 95; p=0·025). INTERPRETATION: Applying LCMM to a large cohort of male patients with CHH reversal uncovered two distinct classes of reversal. Genetic investigation combined with careful clinical phenotyping could help surveillance of reversal after withdrawing treatment, representing the first tailored management approach for male patients with this rare endocrine disorder. FUNDING: National Institutes of Health National Center for Advancing Translational Sciences; Ministry of Health, Rome, Italy; Ministry of University, Rome, Italy; National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development; and the Josiah Macy Jr Foundation. TRANSLATION: For the Italian translation of the abstract see Supplementary Materials section.


Asunto(s)
Enfermedades de los Genitales Masculinos , Hipogonadismo , Pene/anomalías , Estados Unidos , Niño , Adulto , Humanos , Masculino , Adolescente , Estudios Transversales , Hipogonadismo/genética , Hipogonadismo/tratamiento farmacológico , Hormona Folículo Estimulante/uso terapéutico
19.
J Nutr Health Aging ; 28(5): 100217, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38552276

RESUMEN

OBJECTIVE: To test whether transdermal testosterone at a dose of 75 mg per day and/or monthly 24'000 IU Vitamin D reduces the fall risk in pre-frail hypogonadal men aged 65 and older. DESIGN: 2 × 2 factorial design randomized controlled trial, follow up of 12 months. METHODS: Hypogonadism was defined as total testosterone <11.3 nmol/L and pre-frailty as ≥1 Fried- frailty criteria and/or being at risk for falling at the time of screening. The primary outcomes were number of fallers and the rate of falls, assessed prospectively. Secondary outcomes were appendicular lean mass (ALM), sit-to-stand, gait speed, and the short physical performance test battery. Analyses were adjusted for age, BMI, fall history and the respective baseline measurement. RESULTS: We aimed to recruit 168 men and stopped at 91 due to unexpected low recruitment rate (1266 men were pre-screened). Mean age was 72.2 years, serum total testosterone was 10.8 ± 3.0 nmol/l, and 20.9% had 25(OH)D levels below 20 ng/mL. Over 12 months, 37 participants had 72 falls. Neither the odds of falling nor the rate of falls were reduced by testosterone or by vitamin D. Testosterone improved ALM compared to no testosterone (0.21 kg/m2 [0.06, 0.37]), and improved gait speed (0.11 m/s, [0.03, 0.20]) compared to placebo. CONCLUSION: Transdermal testosterone did not reduce fall risk but improved ALM and gait speed in pre-frail older men. Monthly vitamin D supplementation had no benefit.


Asunto(s)
Accidentes por Caídas , Hipogonadismo , Testosterona , Vitamina D , Humanos , Accidentes por Caídas/prevención & control , Masculino , Testosterona/sangre , Anciano , Vitamina D/sangre , Vitamina D/administración & dosificación , Hipogonadismo/tratamiento farmacológico , Anciano de 80 o más Años , Fragilidad/prevención & control , Anciano Frágil
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...