Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroendocrinology ; 106(4): 389-400, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29635226

RESUMO

The androgen receptor (AR) is expressed throughout the hypothalamic-pituitary-gonadal (HPG) axis, and findings from female global AR knockout mice confirm that AR-mediated androgen actions play important roles in regulating female reproductive function. We generated neuron-specific AR knockout mice (NeurARKO) to investigate the functional role of neuronal AR-mediated androgen action in regulating the female HPG axis and fertility. Relative to control females, NeurARKO females exhibited elevated luteinizing hormone (LH) levels at diestrus (p < 0.05) and a compromised serum LH response to ovariectomy and E2 priming (p < 0.01). Furthermore, NeurARKO females displayed reduced Kiss1 mRNA expression in the anteroventral periventricular nucleus at diestrus (p < 0.05) and proestrus (p < 0.05), but elevated Kiss1 (p < 0.05) and neurokinin B (Tac2, p < 0.05) mRNA expression in the arcuate nucleus at proestrus compared to WT controls. Ovarian follicle dynamics were also altered in NeurARKO ovaries at 3 months of age, with a significant reduction in large antral follicle numbers at the proestrus stage compared to control WT ovaries (p < 0.05). Increased follicular atresia was evident in NeurARKO ovaries with a 4-fold increase in unhealthy large preantral follicles (p < 0.01). Despite the findings of aberrant neuroendocrine and ovarian characteristics in the NeurARKO females, estrous cyclicity and overall fertility were comparable between NeurARKO and WT females. In conclusion, our findings revealed that selective loss of neuronal AR actions impacts the kisspeptin/GnRH/LH cascade leading to compromised ovarian follicle dynamics.


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Ovário/metabolismo , Receptores Androgênicos/metabolismo , Animais , Ciclo Estral/metabolismo , Feminino , Camundongos , Camundongos Knockout , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Ovário/patologia
2.
Proc Natl Acad Sci U S A ; 114(16): E3334-E3343, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28320971

RESUMO

Polycystic ovary syndrome (PCOS) is a complex hormonal disorder characterized by reproductive, endocrine, and metabolic abnormalities. As the origins of PCOS remain unknown, mechanism-based treatments are not feasible and current management relies on treatment of symptoms. Hyperandrogenism is the most consistent PCOS characteristic; however, it is unclear whether androgen excess, which is treatable, is a cause or a consequence of PCOS. As androgens mediate their actions via the androgen receptor (AR), we combined a mouse model of dihydrotestosterone (DHT)-induced PCOS with global and cell-specific AR-resistant (ARKO) mice to investigate the locus of androgen actions that mediate the development of the PCOS phenotype. Global loss of the AR reveals that AR signaling is required for all DHT-induced features of PCOS. Neuron-specific AR signaling was required for the development of dysfunctional ovulation, classic polycystic ovaries, reduced large antral follicle health, and several metabolic traits including obesity and dyslipidemia. In addition, ovariectomized ARKO hosts with wild-type ovary transplants displayed normal estrous cycles and corpora lutea, despite DHT treatment, implying extraovarian and not intraovarian AR actions are key loci of androgen action in generating the PCOS phenotype. These findings provide strong evidence that neuroendocrine genomic AR signaling is an important extraovarian mediator in the development of PCOS traits. Thus, targeting AR-driven mechanisms that initiate PCOS is a promising strategy for the development of novel treatments for PCOS.


Assuntos
Androgênios/farmacologia , Modelos Animais de Doenças , Células da Granulosa/patologia , Neurônios/patologia , Sistemas Neurossecretores/efeitos dos fármacos , Síndrome do Ovário Policístico/patologia , Receptores Androgênicos/fisiologia , Animais , Células Cultivadas , Ciclo Estral/efeitos dos fármacos , Feminino , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo
3.
Horm Cancer ; 7(5-6): 316-326, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27506975

RESUMO

Phosphatase and tensin homologue (PTEN) is a known tumour suppressor. To explore the role of Pten in ovarian tumorigenesis, we used transgenic (Tg) SOX2. Cre and AMH. Cre mouse models to direct global Pten haploinsufficiency (Pten +/-) or ovary-specific granulosa cell (GC) Pten disruption (Pten GC ). Pten mutant models were combined with progressively rising Tg-follicle-stimulating hormone (TgFSH) levels to study the tumorigenic potential of combined genetic/endocrine modification in vivo. Global Pten +/- mice exhibited grossly detectable tumours in multiple organs including uterine and mammary tissue and displayed reduced survival. Despite extra-ovarian tumorigenesis, Pten +/- females had no detectable ovarian tumours, although elevated corpus luteum numbers increased ovary size and estrous cycling was altered. Combined TgFSH/Pten +/- mice also had no ovarian tumours, but early survival was reduced in the presence of TgFSH. Ovary-specific Pten GC  ± TgFSH females exhibited no detectable ovarian or uterine tumours, and corpus luteum numbers and estrous cycling remained unchanged. The non-tumorigenic ovarian phenotypes in Pten +/- and Pten GC  ± TgFSH mice support the proposal that multi-hit genetic mutations (including ovarian and extra-ovarian tissue) initiate ovarian tumours. Our findings suggest that elevated FSH may reduce early cancer survival; however, the ovary remains remarkably resistant to Pten-induced tumorigenic changes even in the presence of uterine and reproductive cancers.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Células da Granulosa/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/veterinária , PTEN Fosfo-Hidrolase/genética , Fatores de Transcrição SOXB1/genética , Animais , Feminino , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos Transgênicos , Mutação , Tamanho do Órgão , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Análise de Sobrevida , Neoplasias Uterinas/genética
4.
Am J Physiol Endocrinol Metab ; 311(2): E396-404, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27354237

RESUMO

Recently, we created a unique gain-of-function mouse model with Sertoli cell-specific transgenic androgen receptor expression (TgSCAR) showing that SCAR activity controls the synchronized postnatal development of somatic Sertoli and Leydig cells and meiotic-postmeiotic germ cells. Moderate TgSCAR (TgSCAR(m)) expression reduced testis size but had no effect on male fertility. Here, we reveal that higher TgSCAR expression (TgSCAR(H)) causes male infertility. Higher SCAR activity, shown by upregulated AR-dependent transcripts (Rhox5, Spinw1), resulted in smaller adult TgSCAR(H) testes (50% of normal) despite normal or elevated circulating and intratesticular testosterone levels. Unlike fertile TgSCAR(m) males, testes of adult TgSCAR(H) males exhibited focal regions of interstitial hypertrophy featuring immature adult Leydig cells and higher intratesticular dihydrotestosterone and 5α-androstane 3α,17ß-diol levels that are normally associated with pubertal development. Mature TgSCAR(H) testes also exhibited markedly reduced Sertoli cell numbers (70%), although meiotic and postmeiotic germ cell/Sertoli cell ratios were twofold higher than normal, suggesting that elevated TgSCAR activity supports excessive spermatogenic development. Concurrent with the higher germ cell load of TgSCAR(H) Sertoli cells were increased levels of apoptotic germ cells in TgSCAR(H) relative to TgSCAR(m) testes. In addition, TgSCAR(H) testes displayed unique morphological degeneration that featured accumulated cellular and spermatozoa clusters in dilated channels of rete testes, consistent with reduced epididymal sperm numbers. Our findings reveal for the first time that excessive Sertoli cell AR activity in mature testes can reach a level that disturbs Sertoli/germ cell homeostasis, impacts focal Leydig cell function, reduces sperm output, and disrupts male fertility.


Assuntos
Benzamidas/metabolismo , Fertilidade/genética , Infertilidade Masculina/genética , Piperidinas/metabolismo , Receptores Androgênicos/genética , Células de Sertoli/metabolismo , Androstano-3,17-diol/metabolismo , Animais , Di-Hidrotestosterona/metabolismo , Epididimo , Proteínas de Homeodomínio/genética , Masculino , Meiose , Camundongos Transgênicos , Proteínas Secretadas Inibidoras de Proteinases/genética , Proteínas/genética , Reação em Cadeia da Polimerase em Tempo Real , Rede do Testículo/patologia , Espermatogênese , Espermatozoides , Testículo , Fatores de Transcrição/genética
5.
Anal Chem ; 87(14): 7180-6, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26090565

RESUMO

A liquid chromatography-tandem mass spectrometry (LC-MS/MS) method is described that employs a novel derivatization reagent for the measurement of serum estradiol (E2), with simultaneous analysis of underivatized testosterone (T) and dihydrotestosterone (DHT). The main advantage of the new derivatization reagent 1,2-dimethylimidazole-5-sulfonyl chloride is its analyte-specific fragmentation that enables monitoring of confirmatory mass transitions with high sensitivity. The reaction mixture can be analyzed without additional purification steps using a 9.5 min gradient run, and sensitive detection is achieved with a triple quadrupole mass spectrometer using atmospheric pressure photoionization. Method validation was performed with human serum samples, including a comparison with a standard LC-MS/MS method using 120 samples from a clinical study, and analysis of certified E2 serum reference materials BCR-576, BCR-577, and BCR-578. The lower limits of quantification for E2, T, and DHT were 0.5 pg/mL, 25 pg/mL, and 0.10 ng/mL, respectively, from a 200-µL sample. Validation results indicated good accuracy and agreement with established, conventional LC-MS/MS assays, demonstrating suitability for analysis of samples containing E2 in the low pg/mL range, such as serum from men, children, and postmenopausal women.


Assuntos
Estradiol/sangue , Estrogênios/sangue , Indicadores e Reagentes/química , Ácidos Sulfínicos/química , Cromatografia Líquida de Alta Pressão , Estradiol/química , Humanos , Estrutura Molecular , Espectrometria de Massas em Tandem
6.
Horm Cancer ; 6(4): 142-52, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25943777

RESUMO

BRCA1 mutations are associated with ovarian cancer. Previous studies reported that murine granulosa cell (GC) Brca1 loss caused ovarian-uterine tumors resembling serous cystadenomas, but the pathogenesis of these tumors may have been confounded by ectopic Brca1 expression and altered estrous cycling. We have used Tg.AMH.Cre conferring proven ovarian and GC-specific Cre activity to selectively target Brca1 disruption, denoted Brca1(GC-/-). Furthermore, ovary-specific Brca1(GC-/-) was combined with global Trp53 haploinsufficiency (Trp53(+/-)) and transgenic follicle-stimulating hormone (Tg.FSH) overexpression as a multi-hit strategy to investigate additional genetic and hormonal ovarian tumorigenesis mechanisms. However, 12-month-old Brca1(GC-/-) mice had no detectable ovarian or uterine tumors. Brca1(GC-/-) mice had significantly increased ovary weights, follicles exhibiting more pyknotic granulosa cells, and fewer corpora lutea with regular estrous cycling compared to controls. Isolated Brca1(GC-/-) mutation lengthened the estrous cycle and proestrus stage; however, ovarian cystadenomas were not observed, even when Brca1(GC-/-) was combined with Trp53(+/-) and overexpressed Tg.FSH. Our Brca1(GC-/-) models reveal that specific intra-follicular Brca1 loss alone, or combined with cancer-promoting genetic (Trp53 loss) and endocrine (high serum FSH) changes, was not sufficient to cause ovarian tumors. Our findings show that the ovary is remarkably resistant to oncogenesis, and support the emerging view of an extragonadal, multi-hit origin for ovarian tumorigenesis.


Assuntos
Proteína BRCA1/genética , Hormônio Foliculoestimulante/genética , Haploinsuficiência , Neoplasias Ovarianas/patologia , Proteína Supressora de Tumor p53/genética , Animais , Cistadenoma/genética , Cistadenoma/patologia , Estradiol/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Células da Granulosa/metabolismo , Camundongos , Camundongos Transgênicos , Neoplasias Ovarianas/genética , Ovário/patologia , Útero/patologia
7.
Ann Clin Biochem ; 52(Pt 6): 665-71, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25780247

RESUMO

OBJECTIVES: Urinary hormone concentrations are often adjusted to correct for hydration status. We aimed to determine whether first morning void urine hormones in growing adolescents require adjustments and, if so, whether urinary creatinine or specific gravity are better adjustments. DESIGN AND METHODS: The study population was adolescents aged 10.1 to 14.3 years initially who provided fasting morning blood samples at 0 and 12 months (n = 343) and first morning urine every three months (n = 644). Unadjusted, creatinine and specific gravity-adjusted hormonal concentrations were compared by Deming regression and Bland-Altman analysis and grouped according to self-rated Tanner stage or chronological age. F-ratios for self-rated Tanner stages and age groups were used to compare unadjusted and adjusted hormonal changes in growing young adolescents. Correlations of paired serum and urinary hormonal concentration of unadjusted and creatinine and specific gravity-adjusted were also compared. RESULTS: Fasting first morning void hormone concentrations correlated well and were unbiased between unadjusted or adjusted by either creatinine or specific gravity. Urine creatinine concentration increases with Tanner stages, age and male gender whereas urine specific gravity was not influenced by Tanner stage, age or gender. Adjustment by creatinine or specific gravity of urinary luteinizing hormone, estradiol, testosterone, dihydrotestosterone and dehydroepiandrosterone concentrations did not improve correlation with paired serum concentrations. CONCLUSIONS: Urine steroid and luteinizing hormone concentrations in first morning void samples of adolescents are not significantly influenced by hydration status and may not require adjustments; however, if desired, both creatinine and specific gravity adjustments are equally suitable.


Assuntos
Creatinina/urina , Hormônio Luteinizante/urina , Esteroides/urina , Urinálise/métodos , Adolescente , Criança , Feminino , Humanos , Masculino , Puberdade , Gravidade Específica , Micção
8.
Endocrinology ; 156(1): 400-5, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25365769

RESUMO

Accurate measurement of testosterone is important for reproductive endocrinology research, but the validity of direct (nonextraction) testosterone immunoassays, developed and validated for human serum, has not been appraised for application to mouse serum or steroidogenic tissue extracts. Testosterone was measured in serum and extracts of testis or ovary from male and female wild-type mice by 2 commercial direct testosterone immunoassays, with and without preassay extraction, and by the liquid chromatography, tandem mass spectrometry reference method. Results were compared hierarchically by correlation (Kendall's τ), regression (Passing-Bablok), and deviance (Bland-Altman) analysis, under the null hypothesis of perfect agreement between assays (slope = 1, intercept and deviation = 0). For mouse serum, immunoassays displayed an upward bias with performance better for male vs female sera and, within gender, improved by preassay extraction relative to liquid chromatography, tandem mass spectrometry. Testosterone was detectable in all serum samples, but few (male 54%, female 9%) were accurate (within 20% of the reference measurement). For mouse testis extracts, immunoassays were biased upwards, and preassay extraction improved immunoassay performance. Although testosterone was detectable in all extracts, a minority (45%) was accurate. For mouse ovary extracts, all correlations were poor with severe, upward bias, and while testosterone was detectable in all samples, virtually none were accurate. We conclude that these direct testosterone immunoassay kits provide relatively, but not absolutely, accurate results with male mouse serum and testis extracts but not with female mouse serum and ovary extracts, with performance improved by preassay extraction. Whether relative accuracy is fit for purpose depends on the experimental aims, design, and interpretation.


Assuntos
Imunoensaio/métodos , Espectrometria de Massas/métodos , Ovário/metabolismo , Testículo/metabolismo , Testosterona/sangue , Testosterona/química , Animais , Feminino , Masculino , Camundongos , Testosterona/metabolismo
9.
J Clin Endocrinol Metab ; 99(7): 2592-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24684468

RESUMO

CONTEXT: Testosterone (T) and nandrolone (N) esters require deep im injections by medical personnel but these often deposit injectate into sc fat so that more convenient sc self-administration may be feasible. OBJECTIVE: To investigate the feasibility and pharmacology of sc injection of N decanoate in healthy men using dried blood spot (DBS) for frequent blood sampling without clinic visits. SETTING AND DESIGN: Healthy male volunteers received 100 mg N decanoate by a single sc injection. Finger-prick capillary blood was spotted onto filter paper before injection daily at home for 21 d and stored at room temperature. Venous whole blood was also spotted onto filter paper before and weekly for 3 wk after injection. DBS were extracted for assay of N and T by liquid chromatography tandem mass spectrometry in a single batch with serum concentrations estimated with adjustment for capillary blood sample volume and hematocrit to define peak (N) or nadir (T) time and concentration from individual daily measurements. RESULTS: Daily serum N peaked 2.50 ± 0.25 (SEM) ng/mL at a median (range) of 6 (4-13) days causing a reduction in serum T from 3.50 ± 0.57 ng/mL at baseline to a nadir of 0.38 ± 0.13 (SEM) ng/mL (89 ± 3% suppression) at a median (range) of 8 (5-16) days. Simultaneously sampled capillary, venous whole blood, and serum gave almost identical results for serum T and N. Finger-pricks and sc injections were well tolerated. CONCLUSIONS: This study demonstrates that A) DBS sampling with liquid chromatography mass spectrometry steroid analysis achieves frequent time sampling in the community without requiring clinic visits, venesection, or frozen serum storage, and B) androgen esters in an oil vehicle can be delivered effectively by sc injection, thus avoiding the need for medically supervised deep-im injections.


Assuntos
Preparações de Ação Retardada/farmacocinética , Teste em Amostras de Sangue Seco/métodos , Nandrolona/análogos & derivados , Espectrometria de Massas em Tandem/métodos , Adulto , Coleta de Amostras Sanguíneas/métodos , Cromatografia Líquida/métodos , Preparações de Ação Retardada/administração & dosagem , Hormônio Foliculoestimulante/sangue , Humanos , Injeções Subcutâneas , Hormônio Luteinizante/sangue , Masculino , Nandrolona/administração & dosagem , Nandrolona/sangue , Nandrolona/farmacocinética , Decanoato de Nandrolona , Testosterona/sangue , Adulto Jovem
10.
Endocrinology ; 155(3): 1120-30, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24424066

RESUMO

We determined the functional role of the Sertoli cell glucocorticoid receptor (GR) in vivo using a transgenic Cre-loxP approach to conditionally disrupt GR expression. Sertoli cell GR knockout (SCGRKO) was shown by absent Sertoli cell-specific GR immunolocalization and reduced levels of glucocorticoid-responsive Stc1 and Tsc22d3 mRNA in SCGRKO relative to control testes. Adult SCGRKO testes exhibited distinct morphological changes, including reduced seminiferous tubular lumen formation, decreased total Sertoli cell numbers, and parallel reductions in meiotic spermatocyte and postmeiotic spermatid numbers. Conversely, tubular diameter was increased and testis size was normal in SCGRKO males. Decreased serum FSH and testicular Fshr mRNA levels were consistent with reduced Sertoli cell number. Adult SCGRKO testes also displayed atypical germ cells and interstitial focal accumulations of hypertrophic lipid-laden, immature-like Leydig cells. Circulating LH, and testicular Lhr mRNA, testosterone, dihydrotestosterone, and 3α/3ß-diol levels were all reduced in mature SCGRKO mice, whereas serum testosterone and dihydrotestosterone levels remained normal. Moreover, Sertoli cell GR disruption caused differential changes to steroidogenic enzyme transcripts, with down-regulated testicular Cyp11a1 contrasting with up-regulated Hsd17b3 expression. Reduced SCGRKO testicular expression of Hsd11b2, encoding an enzyme for corticosterone inactivation, supports a dynamic coupling between Hsd11b and androgen production. Our novel SCGRKO model has revealed that Sertoli cell-mediated GR actions support normal testicular function. Sertoli cell GR is required to maintain normal testicular Sertoli/germ cell numbers and circulating gonadotropin levels, as well as optimal Leydig cell maturation and steroidogenesis, providing new insight into gluocorticoid-mediated impact on male reproduction.


Assuntos
Regulação da Expressão Gênica , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiologia , Células de Sertoli/metabolismo , Testículo/crescimento & desenvolvimento , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , 17-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Regulação Enzimológica da Expressão Gênica , Glucocorticoides/metabolismo , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Knockout , Células de Sertoli/citologia , Espermatogênese , Testículo/metabolismo , Transgenes , Regulação para Cima
11.
Clin Chem ; 60(3): 510-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24334824

RESUMO

BACKGROUND: Steroid immunoassays originally required solvent extraction, chromatography, and structurally authentic tracers to avoid interference from steroid cross-reactivity and matrix effects. The demand for steroid assays has driven assay simplification, bypassing this triplet of validity criteria to allow use of unextracted serum, which has introduced bias and nonspecificity at low steroid concentrations. We aimed to evaluate the performance of commercial direct estradiol (E2) immunoassays relative to the reference method of LC-MS and compared serum E2 measurements from each assay with biomarkers of estrogen action. METHODS: We measured serum E2 in duplicate using 5 commercial direct immunoassays and LC-MS in a nested cohort of 101 healthy, asymptomatic men >40 years old from the Healthy Man Study. For each immunoassay, we evaluated the detectability and distribution of serum E2 measurements, CV, and bias (relative to LC-MS) by Passing-Bablok regression and deviance plots. RESULTS: Three assays detected E2 in all samples, whereas E2 was detected in only 53% and 72% of samples by 2 other assays. All 5 assays had positive biases, ranging from 6% to 74%, throughout their ranges. CVs were lower with 4 immunoassays than with LC-MS. LC-MS, but none of the direct immunoassays, correlated with serum testosterone and sex steroid-binding globulin. CONCLUSIONS: The positive bias of direct E2 immunoassays throughout their working range reflects the nonspecific effects of steroid cross-reactivity and/or matrix interference arising from the violation of the triplet validity criteria for steroid immunoassay.


Assuntos
Estradiol/sangue , Cromatografia Líquida/normas , Humanos , Imunoensaio/normas , Masculino , Espectrometria de Massas/normas , Pessoa de Meia-Idade , Valores de Referência
12.
Drug Test Anal ; 6(4): 336-41, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23606665

RESUMO

Urine provides a convenient non-invasive alternative to blood sampling for measurement of certain hormones. Urinary luteinizing hormone (LH) measurements have been used for endocrinology research and anti-doping testing. However, the commercially available LH immunoassays are developed and validated for human blood samples but not urine so that LH assays intended for use with urine samples need thorough validation. Therefore, the present study evaluated the measurement of urinary LH immunoreactivity using previously validated immunofluorometric (IF) and immunochemiluminometric (ICL) LH assays after prolonged frozen storage. LH was measured in serial urine samples following administration of a single injection of one of two doses of recombinant human chorionic hormone (rhCG) with assays run at the end of study (2008) and again after four years of frozen (-20 °C) storage where samples were stored without adding preservatives. The ICL assay showed quantitatively reproducible LH measurements after prolonged -20 °C storage. However, the IF immunoassay gave consistently lower LH levels relative to ICL (2008) with a further proportionate reduction after four years of sample storage (2012). Yet, both the assays displayed similar patterns of the time-course of urine LH measurement both before and after four years of frozen storage. In conclusion, we found that both immunoassays are suitable for urinary LH measurements with ICL assay being more robust for quantitative urinary LH measurement such as for anti-doping purposes, whereas the IF could be applicable for research studies where urine LH levels are compared within-study but not in absolute terms.


Assuntos
Hormônio Luteinizante/urina , Coleta de Amostras Sanguíneas , Dopagem Esportivo , Fluorimunoensaio , Congelamento , Humanos , Medições Luminescentes , Detecção do Abuso de Substâncias
13.
Am J Physiol Endocrinol Metab ; 305(6): E717-26, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23880317

RESUMO

Homozygous androgen receptor (AR)-knockout (ARKO) female mice are subfertile due to both intra- and extraovarian (neuroendocrine) defects as defined by ovary transplantation. Using ARKO mice, this study set out to reveal the precise AR-regulated pathways required for optimal androgen-regulated ovulation and fertility. ARKO females exhibit deficient neuroendocrine negative feedback, with a reduced serum luteinizing hormone (LH) response to ovariectomy (OVX) (P < 0.01). Positive feedback is also altered as intact ARKO females, at late proestrus, exhibit an often mistimed endogenous ovulatory LH surge. Furthermore, at late proestrus, intact ARKO females display diminished preovulatory serum estradiol (E2; P < 0.01) and LH (P < 0.05) surge levels and reduced Kiss1 mRNA expression in the anteroventral periventricular nucleus (P < 0.01) compared with controls. However, this reduced ovulatory LH response in intact ARKO females can be rescued by OVX and E2 priming or treatment with endogenous GnRH. These findings reveal that AR regulates the negative feedback response to E2, E2-positive feedback is compromised in ARKO mice, and AR-regulated negative and positive steroidal feedback pathways impact on intrahypothalamic control of the kisspeptin/GnRH/LH cascade. In addition, intraovarian AR-regulated pathways controlling antral to preovulatory follicle dynamics are disrupted because adult ARKO ovaries collected at proestrus have small antral follicles with reduced oocyte/follicle diameter ratios (P < 0.01) and increased proportions of unhealthy large antral follicles (P < 0.05) compared with controls. As a consequence of aberrant follicular growth patterns, proestrus ARKO ovaries also exhibit fewer preovulatory follicle (P < 0.05) and corpora lutea numbers (P < 0.01). However, embryo development to the blastocyst stage is unchanged in ARKO females, and hence, the subfertility is a consequence of reduced ovulations and not altered embryo quality. These findings reveal that the AR has a functional role in neuroendocrine regulation and timing of the ovulatory LH surge as well as antral/preovulatory follicle development.


Assuntos
Hipotálamo/metabolismo , Infertilidade Feminina/metabolismo , Ovário/metabolismo , Ovulação/metabolismo , Receptores Androgênicos/metabolismo , Animais , Corpo Lúteo/metabolismo , Estradiol/sangue , Ciclo Estral/sangue , Ciclo Estral/genética , Ciclo Estral/metabolismo , Feminino , Hipotálamo/fisiopatologia , Infertilidade Feminina/genética , Infertilidade Feminina/fisiopatologia , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Camundongos , Camundongos Knockout , Folículo Ovariano/metabolismo , Ovário/fisiopatologia , Ovulação/sangue , Ovulação/genética , Receptores Androgênicos/genética
14.
Endocrinology ; 154(9): 3410-22, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23766127

RESUMO

We recently created a mouse model displaying precocious Sertoli cell (SC) and spermatogenic development induced by SC-specific transgenic androgen receptor expression (TgSCAR). Here we reveal that TgSCAR regulates the development, function, and absolute number of Leydig cells (LCs). Total fetal and adult type LC numbers were reduced in postnatal and adult TgSCAR vs control testes, despite normal circulating LH levels. Normal LC to SC ratios found in TgSCAR testes indicate that SC androgen receptor (SCAR)-mediated activity confers a quorum-dependent relationship between total SC and LC numbers. TgSCAR enhanced LC differentiation, shown by elevated ratios of advanced to immature LC types, and reduced LC proliferation in postnatal TgSCAR vs control testes. Postnatal TgSCAR testes displayed up-regulated expression of coupled ligand-receptor transcripts (Amh-Amhr2, Dhh-Ptch1, Pdgfa-Pdgfra) for potential SCAR-stimulated paracrine pathways, which may coordinate LC differentiation. Neonatal TgSCAR testes displayed normal T and dihydrotestosterone levels despite differential changes to steroidogenic gene expression, with down-regulated Star, Cyp11a1, and Cyp17a1 expression contrasting with up-regulated Hsd3b1, Hsd17b3, and Srd5a1 expression. TgSCAR males also displayed elevated postnatal and normal adult serum testosterone levels, despite reduced LC numbers. Enhanced adult-type LC steroidogenic output was revealed by increased pubertal testicular T, dihydrotestosterone, 3α-diol and 3ß-diol levels per LC and up-regulated steroidogenic gene (Nr5a1, Lhr, Cyp11a1, Cyp17a1, Hsd3b6, Srd5a1) expression in pubertal or adult TgSCAR vs control males, suggesting regulatory mechanisms maintain androgen levels independently of absolute LC numbers. Our unique gain-of-function TgSCAR model has revealed that SCAR activity controls temporal LC differentiation, steroidogenic function, and population size.


Assuntos
Diferenciação Celular , Células Intersticiais do Testículo/citologia , Receptores Androgênicos/metabolismo , Células de Sertoli/metabolismo , Testículo/citologia , Congêneres da Testosterona/metabolismo , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Contagem de Células , Hemizigoto , Isoenzimas/biossíntese , Isoenzimas/genética , Isoenzimas/metabolismo , Células Intersticiais do Testículo/metabolismo , Ligantes , Masculino , Camundongos , Camundongos Transgênicos , Receptores Patched , Receptor Patched-1 , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores Androgênicos/biossíntese , Receptores Androgênicos/genética , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Peptídeos/biossíntese , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Células de Sertoli/citologia , Desenvolvimento Sexual , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Congêneres da Testosterona/sangue , Regulação para Cima
15.
J Endocrinol ; 218(2): 151-63, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23678134

RESUMO

Neurturin (NTN) is a member of the glial cell line-derived neurotrophic factor (GDNF) family and signals through GDNF family receptor alpha 2 (GFRα2). We hypothesised that epithelial atrophy reported in the reproductive organs of Ntn (Nrtn)- and Gfrα2 (Gfra2)-deficient mice could be due to NTN affecting the hormonal environment. To investigate this, we compared the reproductive organs of Ntn- and Gfrα2-deficient male mice in parallel with an analysis of their circulating reproductive hormone levels. There were no significant structural changes within the organs of the knockout mice; however, serum and intratesticular testosterone and serum LH levels were very low. To reconcile these observations, we tested androgen sensitivity by creating a dihydrotestosterone (DHT) clamp (castration plus DHT implant) to create fixed circulating levels of androgens, allowing the evaluation of androgen-sensitive endpoints. At the same serum DHT levels, serum LH levels were lower and prostate and seminal vesicle weights were higher in the Ntn knockout (NTNKO) mice than in the wild-type mice, suggesting an increased response to androgens in the accessory glands and hypothalamus and pituitary of the NTNKO mice. Testicular and pituitary responsiveness was unaffected in the NTNKO males, as determined by the response to the human chorionic gonadotrophin or GNRH analogue, leuprolide, respectively. In conclusion, our results suggest that NTN inactivation enhances androgen sensitivity in reproductive and neuroendocrine tissues, revealing a novel mechanism to influence reproductive function and the activity of other androgen-dependent tissues.


Assuntos
Androgênios/metabolismo , Genitália Masculina/metabolismo , Sistemas Neurossecretores/metabolismo , Neurturina/deficiência , Animais , Genitália Masculina/crescimento & desenvolvimento , Humanos , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Knockout , Sistemas Neurossecretores/crescimento & desenvolvimento , Neurturina/genética , Tamanho do Órgão , Próstata/crescimento & desenvolvimento , Próstata/metabolismo , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Testosterona/sangue
16.
Biol Reprod ; 87(6): 151, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23115271

RESUMO

Ovarian granulosa cells display strong androgen receptor (AR) expression, suggesting a functional role for direct AR-mediated actions within developing mammalian follicles. By crossing AR-floxed and anti-Müllerian hormone (AMH)-Cre recombinase mice, we generated granulosa cell-specific androgen receptor knockout mice (GCARKO). Cre expression, assessed by lacZ activity, localized to 70%-100% of granulosa cells in most preantral to antral follicles, allowing for selected evaluation of granulosa cell AR-dependent actions during follicle development. Relative to wild-type (WT) females, GCARKO females were subfertile, producing a 24% reduction in the number of litters (P < 0.05) over 6 mo and an age-dependent decrease in total number of pups born, evident from 6 mo of age (P < 0.05). Follicle dynamics were altered in GCARKO ovaries at 3 mo of age, with a significant reduction in large preantral and small antral follicle numbers compared to WT ovaries (P < 0.05). Global premature follicle depletion was not observed, but increased follicular atresia was evident in GCARKO ovaries at 6 mo of age, with an 81% increase in unhealthy follicles and zona pellucida remnants (P < 0.01). Cumulus cell expansion was decreased (P < 0.01) and oocyte viability was diminished in GCARKO females, with a significant reduction in the percentage of oocytes fertilized after natural mating and, thus, in the rate of progression to the two-cell embryo stage (P < 0.05). In addition, compared with age-matched WT females, 6-mo-old GCARKO females exhibited significantly prolonged estrous cycles (P ≤ 0.05), suggesting altered hypothalamic-pituitary-gonadal feedback signaling. In conclusion, our findings revealed that selective loss of granulosa cell AR actions during preantral and antral stages of development leads to a premature reduction in female fecundity through reduced follicle health and oocyte viability.


Assuntos
Células da Granulosa/metabolismo , Infertilidade Feminina/metabolismo , Oogênese , Receptores Androgênicos/metabolismo , Transdução de Sinais , Animais , Hormônio Antimülleriano/genética , Hormônio Antimülleriano/metabolismo , Sobrevivência Celular , Cruzamentos Genéticos , Células do Cúmulo/metabolismo , Células do Cúmulo/patologia , Ciclo Estral/metabolismo , Feminino , Fertilização , Atresia Folicular/metabolismo , Células da Granulosa/patologia , Heterozigoto , Infertilidade Feminina/etiologia , Infertilidade Feminina/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Receptores Androgênicos/deficiência , Receptores Androgênicos/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Recombinases/genética , Recombinases/metabolismo , Zona Pelúcida/metabolismo , Zona Pelúcida/patologia
17.
Asian J Androl ; 14(3): 476-80, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22522506

RESUMO

Objective studies of men's reproductive function are hindered by their reliance on: (i) self-reporting to quantify sexual activity and (ii) masturbation to quantify sperm output rendering both types of estimate vulnerable to unverifiable subjective factors. We therefore examined whether detection of spermatozoa and measurement of prostate-specific antigen (PSA) in urine could provide objective semiquantitative estimates of sperm output and recent ejaculation, respectively, using widely available laboratory techniques. Of 11 healthy volunteers who provided urine samples before and at intervals for 5 days after ejaculation, sperm was present in 2/11 men before, and in all 11/11 samples immediately after ejaculation, but by the second and subsequent void, spermatozoa were present in ∼10%. PSA was detectable at high levels in all urine samples, peaking at the first post-ejaculatory sample but returning to baseline levels by the second post-ejaculatory void. We conclude that urinary spermatozoa and PSA are objective biomarkers for sperm production and sexual activity, but only for a short-time window until the first post-ejaculatory urine void. Hence, for a single urine specimen, the presence of spermatozoa and PSA are valid biomarkers, reflecting sperm production and recent ejaculation only until the next micturition, so their measurement should be restricted to the first morning urine void.


Assuntos
Abstinência Sexual/fisiologia , Comportamento Sexual/fisiologia , Espermatogênese/fisiologia , Espermatozoides/citologia , Urina/citologia , Adulto , Biomarcadores/urina , Ejaculação/fisiologia , Humanos , Masculino , Antígeno Prostático Específico/urina , Contagem de Espermatozoides , Espermatozoides/fisiologia , Fatores de Tempo , Adulto Jovem
18.
Horm Cancer ; 3(3): 113-24, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22370991

RESUMO

Hormones, notably estrogens, are pivotal in the origins of breast cancer but androgenic effects, while supported by persistence of AR expression in breast cancers, remain controversial. This study determined the role of the androgen actions via androgen receptor (AR) in experimental mammary cancer. Androgen-resistant female and male mice (ARKO) were generated using Cre/loxP technique and featured a global AR inactivation. The effect of AR inactivation and influence of genetic background on 7,12-dimethylbenz[a]anthracene (DMBA)-induced tumorigenesis was confirmed using two separate ARKO models with different genetic backgrounds. The onset of palpable mammary tumors was significantly faster in ARKO females (median time 22 vs 34 weeks, respectively; (p = 0.0024; multivariate Cox regression) compared to WT and independent of the mouse genetic background. The cumulative incidence at 9 months was 81 ± 10% [mean ± SE] for ARKO compared to 50 ± 13% in WT females. The increased DMBA susceptibility of ARKO females was associated with a higher epithelial proliferation index but not with major structural or receptor (estrogen or progesterone) expression differences between the virgin WT or ARKO female mammary glands. AR inactivation allowed substantial ductal extension in ARKO males while WT males displayed only rudimentary epithelial branches or complete regression of epithelial structures. Yet, DMBA did not induce epithelial mammary tumors in WT or ARKO males, demonstrating that AR inactivation alone is insufficient to promote mammary tumors. These results demonstrate that AR inactivation accelerates mammary carcinogenesis in female mice exposed to the chemical carcinogen DMBA regardless of mouse genetic background but require prior exposure to endogenous ovarian hormones.


Assuntos
Neoplasias Mamárias Experimentais/etiologia , Receptores Androgênicos/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Androgênios/metabolismo , Animais , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Predisposição Genética para Doença , Gonadotropinas Hipofisárias/sangue , Masculino , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Androgênicos/genética , Testosterona/sangue
19.
Am J Physiol Endocrinol Metab ; 301(4): E727-35, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21750267

RESUMO

Androgens influence prostate growth and development, so androgen withdrawal can control progression of prostate diseases. Although estrogen treatment was originally used to induce androgen withdrawal, more recently direct estrogen effects on the prostate have been recognized, but the nature of androgen-estrogen interactions within the prostate remain poorly understood. To characterize androgen effects on estrogen sensitivity in the mouse prostate, we contrasted models of castration-induced androgen withdrawal in the prostate stromal and epithelial compartments with a prostate epithelial androgen receptor (AR) knockout (PEARKO) mouse model of selective epithelial AR inactivation. Castration markedly increased prostate epithelial estrogen receptor (ER)α immunoreactivity compared with very low ERα expression in intact males. Similarly, strong basal and luminal ERα expression was detected in PEARKO prostate of intact males, suggesting that epithelial AR activity regulated epithelial ERα expression. ERß was strongly expressed in intact, castrated, and PEARKO prostate. However, strong clusters of epithelial ERß positivity coincided with epithelial stratification in PEARKO prostate. In vivo estrogen sensitivity was increased in PEARKO males, with greater estradiol-induced prostate growth and epithelial proliferation leading to squamous metaplasia, featuring markedly increased epithelial proliferation, thickening, and keratinization compared with littermate controls. Our results suggest that ERα expression in the prostate epithelial cells is regulated by local, epithelia-specific, androgen-dependent mechanisms, and this imbalance in the AR- and ER-mediated signaling sensitizes the mature prostate to exogenous estrogens.


Assuntos
Células Epiteliais/metabolismo , Estradiol/farmacologia , Estrogênios/farmacologia , Próstata/metabolismo , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Células Epiteliais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Próstata/efeitos dos fármacos , Receptores Androgênicos/genética , Receptores de Estrogênio/genética
20.
Mol Cell Endocrinol ; 342(1-2): 81-6, 2011 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-21664242

RESUMO

A well established functional polymorphism of the human androgen receptor (hAR) is the length of AR's N-terminal glutamine tract (Q-tract). This tract is encoded by a CAG trinucleotide repeat and varies from 8 to 33 codons in the healthy population. Q-tract length is inversely correlated with AR transcriptional activity in vitro, but whether endogenous androgen action is affected is not consistently supported by results of clinical and epidemiological studies. To test whether Q-tract length influences androgen sensitivity in vivo, we examined effects of controlled androgen exposure in "humanized" mice with hAR knock-in alleles bearing 12, 21 or 48 CAGs. Mature male mice were analyzed before or 2weeks after orchidectomy, with or without a subdermal dihydrotestosterone (DHT) implant to attain stable levels of this non-aromatizable androgen. The validity of this DHT clamp was demonstrated by similar serum levels of DHT and its two primary 3αDiol and 3ßDiol metabolites, regardless of AR Q-tract length. Q-tract length was inversely related to DHT-induced suppression of castrate serum LH (p=0.005), as well as seminal vesicle (SV) weight (p=0.005) and prostate lobe weights (p<0.006). This confirms that the hAR Q-tract polymorphism mediates in vivo tissue androgen sensitivity by impacting negative hypothalamic feedback and trophic androgen effects on target organs. In this manner, AR Q-tract length variation may influence numerous aspects of male health, from virilization to fertility, as well as androgen-dependent diseases, such as prostate cancer.


Assuntos
Androgênios/metabolismo , Di-Hidrotestosterona/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Di-Hidrotestosterona/sangue , Di-Hidrotestosterona/farmacologia , Técnicas de Introdução de Genes , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Orquiectomia , Polimorfismo de Nucleotídeo Único , Transcrição Gênica , Repetições de Trinucleotídeos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...