Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Haematol ; 105(3): 247-254, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32311143

RESUMO

BACKGROUND: Androgens function through DNA and non-DNA binding-dependent signalling of the androgen receptor (AR). How androgens promote erythropoiesis is not fully understood. DESIGN AND METHODS: To identify the androgen signalling pathway, we treated male mice lacking the second zinc finger of the DNA-binding domain of the AR (ARΔZF2 ) with non-aromatizable 5α-dihydrotestosterone (5α-DHT) or aromatizable testosterone. To distinguish direct hematopoietic and non-hematopoietic mechanisms, we performed bone marrow reconstitution experiments. RESULTS: In wild-type mice, 5α-DHT had greater erythroid activity than testosterone, which can be aromatized to estradiol. The erythroid response in wild-type mice following 5α-DHT treatment was associated with increased serum erythropoietin (EPO) and its downstream target erythroferrone, and hepcidin suppression. 5α-DHT had no erythroid activity in ARΔZF2 mice, proving the importance of DNA binding by the AR. Paradoxically, testosterone, but not 5α-DHT, suppressed EPO levels in ARΔZF2 mice, suggesting testosterone following aromatization may oppose the erythroid-stimulating effects of androgens. Female wild-type mice reconstituted with ARΔZF2 bone marrow cells remained responsive to 5α-DHT. In contrast, ARΔZF2 mice reconstituted with female wild-type bone marrow cells showed no response to 5α-DHT. CONCLUSION: Erythroid promoting effects of androgens are mediated through DNA binding-dependent actions of the AR in non-hematopoietic cells, including stimulating EPO expression.


Assuntos
Androgênios/metabolismo , Proteínas de Ligação a DNA/metabolismo , Eritropoese , Receptores Androgênicos/metabolismo , Androgênios/farmacologia , Animais , Biomarcadores , Eritropoese/efeitos dos fármacos , Eritropoetina/sangue , Feminino , Regulação da Expressão Gênica , Ferro/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Ligação Proteica , Receptores Androgênicos/genética , Transdução de Sinais
2.
Endocr Res ; 39(3): 130-5, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24467187

RESUMO

Androgens (testosterone and dihydrotestosterone) acting via the androgen receptor (AR) are required for male sexual differentiation, and also regulate the development of many other tissues including muscle, fat and bone. We previously generated an AR(lox) mouse line with exon 3 of the AR gene targeted by loxP sites. The deletion of exon 3 is in-frame, so only the DNA binding-dependent actions of the AR are deleted, but non-DNA binding-dependent actions are retained. This line also contained an antibiotic resistance selection cassette, neomycin (neo) in intron 3, which was also flanked by loxP sites. Hemizygous AR(lox) male mice demonstrated a phenotype of hyperandrogenization, with increased mass of androgen-dependent tissues. We hypothesized that this hyperandrogenization was likely to be due to the presence of the neo cassette. In this study, we have generated an AR(lox) neo-negative mouse line, using the EIIa-cre deleter mouse line to remove the neo cassette. Hemizygous AR(lox) neo-negative male mice have a normal phenotype, with normal body mass and normal mass of androgen-dependent tissues including the testis, seminal vesicles, kidney, spleen, heart and retroperitoneal fat. This neo-negative exon 3-targeted mouse line is the only floxed AR mouse line available to study the DNA binding-dependent actions of the AR in a tissue-specific manner, and is suitable for investigation in all tissues. This study demonstrates the importance of removing the selection cassette, which can potentially alter the phenotype of floxed mouse lines even in the absence of detectable effects on target gene expression.


Assuntos
Técnicas de Inativação de Genes/métodos , Receptores Androgênicos/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Feminino , Deleção de Genes , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neomicina/metabolismo , Fenótipo , Receptores Androgênicos/metabolismo
3.
Gen Comp Endocrinol ; 193: 1-9, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23871650

RESUMO

Jawed vertebrates (Gnasthostomes) are broadly separated into cartilaginous fishes (Chondricthyes) and bony vertebrates (Osteichthyes). Cartilaginous fishes are divided into chimaeras (e.g. ratfish, rabbit fish and elephant shark) and elasmobranchs (e.g. sharks, rays and skates). Both cartilaginous fish and bony vertebrates are believed to have a common armoured bony ancestor (Class Placodermi), however cartilaginous fish are believed to have lost bone. This study has identified and investigated genes involved in skeletal development in vertebrates, in the cartilaginous fish, elephant shark (Callorhinchus milii). Ctnnb1 (ß-catenin), Sfrp (secreted frizzled protein) and a single Sost or Sostdc1 gene (sclerostin or sclerostin domain-containing protein 1) were identified in the elephant shark genome and found to be expressed in a number of tissues, including cartilage. ß-catenin was also localized in several elephant shark tissues. The expression of these genes, which belong to the Wnt/ß-catenin pathway, is required for normal bone formation in mammals. These findings in the cartilaginous skeleton of elephant shark support the hypothesis that the common ancestor of cartilaginous fishes and bony vertebrates had the potential for making bone.


Assuntos
Proteínas de Peixes/genética , Regulação da Expressão Gênica no Desenvolvimento , Tubarões/crescimento & desenvolvimento , Tubarões/genética , Via de Sinalização Wnt/fisiologia , Animais , Cartilagem/metabolismo , Feminino , Proteínas de Peixes/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Via de Sinalização Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
4.
J Cell Physiol ; 226(6): 1453-60, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21413019

RESUMO

The naturally occurring polyamines, spermidine, spermine, and their precursor putrescine, play indispensible roles in both prokaryotic and eukaryotic cells, from basic DNA synthesis to regulation of cell proliferation and differentiation. The rate-limiting polyamine biosynthetic enzymes, ornithine decarboxylase (ODC) and S-adenosylmethionine decarboxylase, are essential for mammalian development, with knockout of the genes encoding these enzymes, Odc1 and Amd1, causing early embryonic lethality in mice. In muscle, the involvement of polyamines in muscle hypertrophy is suggested by the concomitant increase in cardiac and skeletal muscle mass and polyamine levels in response to anabolic agents including ß-agonists. In addition to ß-agonists, androgens, which increase skeletal mass and strength, have also been shown to stimulate polyamine accumulation in a number of tissues. In muscle, androgens act via the androgen receptor to regulate expression of polyamine biosynthetic enzyme genes, including Odc1 and Amd1, which may be one mechanism via which androgens promote muscle growth. This review outlines the role of polyamines in proliferation and hypertrophy, and explores their possible actions in mediating the anabolic actions of androgens in muscle.


Assuntos
Androgênios/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Poliaminas/metabolismo , Animais , Proliferação de Células , Humanos , Hipertrofia , Poliaminas/química
5.
Am J Physiol Endocrinol Metab ; 301(1): E172-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21505150

RESUMO

The aim of this study is to determine if the Odc1 gene, which encodes ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine biosynthesis, is directly regulated by the androgen receptor (AR) in skeletal muscle myoblasts and if Odc1 regulates myoblast proliferation and differentiation. We previously showed that expression of Odc1 is decreased in muscle from AR knockout male mice. In this study, we show in vivo that Odc1 expression is also decreased >60% in muscle from male muscle-specific AR knockout mice. In normal muscle homeostasis, Odc1 expression is regulated by age and sex, reflecting testosterone levels, as muscle of adult male mice expresses high levels of Odc1 compared with age-matched females and younger males. In vitro, expression of Odc1 is 10- and 1.5-fold higher in proliferating mouse C(2)C(12) and human skeletal muscle myoblasts, respectively, than in differentiated myotubes. Dihydrotestosterone increases Odc1 levels 2.7- and 1.6-fold in skeletal muscle cell myoblasts after 12 and 24 h of treatment, respectively. Inhibition of ODC activity in C(2)C(12) myoblasts by α-difluoromethylornithine decreases myoblast number by 40% and 66% following 48 and 72 h of treatment, respectively. In contrast, overexpression of Odc1 in C(2)C(12) myoblasts results in a 27% increase in cell number vs. control when cells are grown under differentiation conditions for 96 h. This prolonged proliferation is associated with delayed differentiation, with reduced expression of the differentiation markers myogenin and Myf6 in Odc1-overexpressing cells. In conclusion, androgens act via the AR to upregulate Odc1 in skeletal muscle myoblasts, and Odc1 promotes myoblast proliferation and delays differentiation.


Assuntos
Proliferação de Células , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/fisiologia , Ornitina Descarboxilase/genética , Receptores Androgênicos/fisiologia , Androgênios/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Embrião de Mamíferos , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/metabolismo , Ornitina Descarboxilase/metabolismo , Gravidez , Receptores Androgênicos/metabolismo , Regulação para Cima/efeitos dos fármacos
6.
Am J Physiol Endocrinol Metab ; 301(5): E767-78, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21712531

RESUMO

In men, as testosterone levels decrease, fat mass increases and muscle mass decreases. Increased fat mass in men, in particular central obesity, is a major risk factor for type 2 diabetes, cardiovascular disease, and all-cause mortality. Testosterone treatment has been shown to decrease fat mass and increase fat-free mass. We hypothesize that androgens act directly via the DNA binding-dependent actions of the androgen receptor (AR) to regulate genes controlling fat mass and metabolism. The aim of this study was to determine the effect of a global DNA binding-dependent (DBD) AR knockout (DBD-ARKO) on the metabolic phenotype in male mice by measuring body mass, fat mass, food intake, voluntary physical activity, resting energy expenditure, substrate oxidation rates, serum glucose, insulin, lipid, and hormone levels, and metabolic gene expression levels and second messenger protein levels. DBD-ARKO males have increased adiposity despite a decreased total body mass compared with wild-type (WT) males. DBD-ARKO males showed reduced voluntary activity, decreased food intake, increased serum leptin and adiponectin levels, an altered lipid metabolism gene profile, and increased phosphorylated CREB levels compared with WT males. This study demonstrates that androgens acting via the DNA binding-dependent actions of the AR regulate fat mass and metabolism in males and that the increased adiposity in DBD-ARKO male mice is associated with decreased voluntary activity, hyperleptinemia and hyperadiponectinemia and not with insulin resistance, increased food intake, or decreased resting energy expenditure.


Assuntos
Adiposidade/genética , Resistência à Insulina/genética , Motivação/genética , Atividade Motora/genética , Receptores Androgênicos/genética , Adiponectina/sangue , Animais , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Resistência à Insulina/fisiologia , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Motivação/fisiologia , Domínios e Motivos de Interação entre Proteínas/genética , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Regulação para Cima/genética
7.
FASEB J ; 22(8): 2676-89, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18390925

RESUMO

To identify mechanisms of anabolic androgen action in muscle, we generated male and female genomic androgen receptor (AR) knockout (ARKO) mice, and characterized muscle mass, contractile function, and gene expression. Muscle mass is decreased in ARKO males, but normal in ARKO females. The levator ani muscle, which fails to develop in normal females, is also absent in ARKO males. Force production is decreased from fast-twitch ARKO male muscle, and slow-twitch muscle has increased fatigue resistance. Microarray analysis shows up-regulation of genes encoding slow-twitch muscle contractile proteins. Real-time PCR confirms that expression of genes encoding polyamine biosynthetic enzymes, ornithine decarboxylase (Odc1), and S-adenosylmethionine decarboxylase (Amd1), is reduced in ARKO muscle, suggesting androgens act through regulation of polyamine biosynthesis. Altered expression of regulators of myoblast progression from proliferation to terminal differentiation suggests androgens also promote muscle growth by maintaining myoblasts in the proliferate state and delaying differentiation (increased Cdkn1c and Igf2, decreased Itg1bp3). A similar pattern of gene expression is observed in orchidectomized male mice, during androgen withdrawal-dependent muscle atrophy. In conclusion, androgens are not required for peak muscle mass in females. In males, androgens act through the AR to regulate multiple gene pathways that control muscle mass, strength, and fatigue resistance.


Assuntos
Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/fisiopatologia , Receptores Androgênicos/deficiência , Androgênios/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Feminino , Expressão Gênica , Redes Reguladoras de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular/fisiologia , Fibras Musculares de Contração Rápida/fisiologia , Fibras Musculares de Contração Lenta/fisiologia , Músculo Esquelético/patologia , Mioblastos Esqueléticos/patologia , Mioblastos Esqueléticos/fisiologia , Orquiectomia , Tamanho do Órgão , Receptores Androgênicos/genética , Receptores Androgênicos/fisiologia , Caracteres Sexuais , Testículo/fisiologia
8.
Physiol Genomics ; 33(1): 133-7, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18171720

RESUMO

We previously generated a conditional floxed mouse line to study androgen action, in which exon 3 of the androgen receptor (AR) gene is flanked by loxP sites, with the neomycin resistance gene present in intron 3. Deletion of exon 3 in global AR knockout mice causes androgen insensitivity syndrome, characterized by genotypic males lacking normal masculinization. We now report that male mice carrying the floxed allele (AR(lox)) have the reverse phenotype, termed hyperandrogenization. AR(lox) mice have increased mass of androgen-dependent tissues, including kidney, (P < 0.001), seminal vesicle (P < 0.001), levator ani muscle (P = 0.001), and heart (P < 0.05). Serum testosterone is not significantly different. Testis mass is normal, histology shows normal spermatogenesis, and AR(lox) males are fertile. AR(lox) males also have normal AR mRNA levels in kidney, brain, levator ani, liver, and testis. This study reaffirms the need to investigate the potential phenotypic effects of floxed alleles in the absence of cre in tissue-specific knockout studies. In addition, this androgen hypersensitivity model may be useful to further investigate the effects of subtle perturbations of androgen action in a range of androgen-responsive systems in the male.


Assuntos
Hiperandrogenismo/genética , Perda de Heterozigosidade/fisiologia , Receptores Androgênicos/genética , Animais , Peso Corporal/genética , Cruzamentos Genéticos , Feminino , Regulação da Expressão Gênica/fisiologia , Coração/anatomia & histologia , Integrases/genética , Integrases/metabolismo , Rim/anatomia & histologia , Fígado/anatomia & histologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tamanho do Órgão/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos/metabolismo , Testículo/anatomia & histologia , Testículo/citologia , Testosterona/sangue
9.
Bone ; 42(6): 1164-74, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18387351

RESUMO

Human parathyroid hormone (hPTH) is currently the only treatment for osteoporosis that forms new bone. Previously we described a fish equivalent, Fugu parathyroid hormone 1 (fPth1) which has hPTH-like biological activity in vitro despite fPth1(1-34) sharing only 53% identity with hPTH(1-34). Here we demonstrate the in vivo actions of fPth1(1-34) on bone. In study 1, young male rats were injected intermittently for 30 days with fPth1 [30 microg-1,000 microg/kg body weight (b.w.), (30fPth1-1,000fPth1)] or hPTH [30 microg-100 microg/kg b.w. (30hPTH-100hPTH)]. In proximal tibiae at low doses, the fPth1 was positively correlated with trabecular bone volume/total volume (TbBV/TV) while hPTH increased TbBV/TV, trabecular thickness (TbTh) and trabecular number (TbN). 500fPth1 and 1000fPth1 increased TbBV/TV, TbTh, TbN, mineral apposition rate (MAR) and bone formation rate/bone surface (BFR/BS) with a concomitant decrease in osteoclast surface and number. In study 2 ovariectomized (OVX), osteopenic rats and sham operated (SHAM) rats were injected intermittently with 500 microg/kg b.w. of fPth1 (500fPth1) for 11 weeks. 500fPth1 treatment resulted in increased TbBV/TV (151%) and TbTh (96%) in the proximal tibiae due to increased bone formation as assessed by BFR/BS (490%) and MAR (131%). The effect was restoration of TbBV/TV to SHAM levels without any effect on bone resorption. 500fPth1 also increased TbBV/TV and TbTh in the vertebrae (L6) and cortical thickness in the mid-femora increasing bone strength at these sites. fPth1 was similarly effective in SHAM rats. Notwithstanding the low amino acid sequence homology with hPTH (1-34), we have clearly established the efficacy of fPth1 (1-34) as an anabolic bone agent.


Assuntos
Anabolizantes/farmacologia , Doenças Ósseas Metabólicas/metabolismo , Osso e Ossos/efeitos dos fármacos , Ovariectomia , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Takifugu , Anabolizantes/uso terapêutico , Animais , Biomarcadores/metabolismo , Doenças Ósseas Metabólicas/tratamento farmacológico , Osso e Ossos/anatomia & histologia , Osso e Ossos/fisiologia , Feminino , Humanos , Masculino , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Hormônio Paratireóideo/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Estresse Mecânico
10.
Reprod Fertil Dev ; 20(8): 861-70, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19007549

RESUMO

Spermatogenesis requires androgen but, paradoxically, oestradiol (E2) treatment stimulates spermatogenic development in gonadotrophin- and androgen-deficient hypogonadal (hpg) mice. The mechanisms of E2-induced spermatogenesis were investigated by determining intratesticular E2 levels and testis cell populations in E2-treated hpg male mice, and E2 spermatogenic actions were determined in androgen receptor-knockout (ARKO) mice. Despite increased serum E2 concentrations (150-300 pmol L(-1)), intratesticular E2 concentrations declined fivefold (P < 0.001) in E2-treated v. untreated hpg male mice. Serum FSH reached 40% of normal and total testicular numbers of known FSH-responsive Sertoli, spermatogonia and meiotic spermatocyte populations were significantly (P < 0.001) elevated 1.7-, 4- and 13-fold, respectively. However, E2 administration also increased androgen-dependent pachytene spermatocytes and post-meiotic spermatids to levels comparable with testosterone-treated hpg testes. Selective investigation of androgen receptor involvement used E2-treated ARKO mice, which were found to exhibit increased (1.6-fold; P < 0.05) intratesticular E2 concentrations and suppression of the elevated serum gonadotrophins, although FSH remained twofold higher than normal. However, testis size and total Sertoli, spermatogonia and spermatocyte numbers were not increased in E2-treated ARKO male mice. Therefore, E2-stimulated murine spermatogenic development occurs with markedly suppressed and not elevated intratesticular E2 levels and displays an absolute requirement for functional androgen receptors. We propose that this paradoxical E2 spermatogenic response is explained by predominantly extratesticular E2 actions, increasing FSH to combine with residual androgen activity in hpg testes to stimulate pre- to post-meiotic development.


Assuntos
Estradiol/farmacologia , Receptores Androgênicos/fisiologia , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Estradiol/metabolismo , Hormônio Foliculoestimulante/sangue , Hipogonadismo/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Receptores Androgênicos/genética , Células de Sertoli/citologia , Células de Sertoli/efeitos dos fármacos , Transdução de Sinais/fisiologia , Espermatogênese/fisiologia , Espermatozoides/citologia , Espermatozoides/efeitos dos fármacos , Testículo/citologia , Testículo/metabolismo , Testosterona/metabolismo
11.
J Clin Invest ; 113(9): 1334-43, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15124025

RESUMO

Parathyroid hormone-related peptide (PTHrP) is a positive regulator of chondrocyte proliferation during bone development. In embryonic mice lacking PTHrP, chondrocytes stop proliferating prematurely, with accelerated differentiation. Because the bone phenotype of mice lacking the cyclin-dependent kinase inhibitor p57(Kip2) is the opposite of the PTHrP-null phenotype, we hypothesized that PTHrP's proliferative actions in chondrocytes might be mediated by opposing p57. We generated p57/PTHrP-null embryos, which showed partial rescue of the PTHrP-null phenotype. There was reversal of the loss of proliferative chondrocytes in most bones, with reversal of the accelerated differentiation that occurs in the PTHrP-null phenotype. p57 mRNA and protein were upregulated in proliferative chondrocytes in the absence of PTHrP. Metatarsal culture studies confirmed the action of PTHrP to decrease p57 mRNA and protein levels in a model in which parathyroid hormone (PTH), used as an analog of PTHrP, increased chondrocyte proliferation rate and the length of the proliferative domain. PTH treatment of p57-null metatarsals had no effect on proliferation rate in round proliferative chondrocytes but still stimulated proliferation in columnar chondrocytes. These studies suggest that the effects of PTHrP on both the rate and extent of chondrocyte proliferation are mediated, at least in part, through suppression of p57 expression.


Assuntos
Desenvolvimento Ósseo/fisiologia , Divisão Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Hormônio Paratireóideo/farmacologia , Animais , Diferenciação Celular , Condrócitos/citologia , Colágeno Tipo X/metabolismo , Inibidor de Quinase Dependente de Ciclina p57 , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Ossos do Metatarso/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Hormônio Paratireóideo/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/análise , RNA Mensageiro/análise , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Ulna/embriologia
12.
Hum Mutat ; 27(5): 483-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16619235

RESUMO

We have characterized an unusual family with two different androgen receptor (AR) gene deletions, in which we propose a novel mechanism of deletion formation has occurred. Affected individuals have the X-linked disorder androgen insensitivity syndrome, and we previously showed that different family members have deletions of different exons of the AR gene. We have now fully sequenced the deletions from affected individuals, and confirmed the presence of different deletions in different affected family members. Most affected and heterozygote individuals have a 4,430-bp deletion of exon 5 that occurred between repeated GTGGCAT motifs in introns 4 and 5. One affected hemizygous individual has a 4,033-bp deletion of exons 6 and 7 that occurred between repeated CCTC motifs in introns 5 and 7. The intron 5 breakpoint junctions of the two deletions are only 11 bp apart. Surprisingly, the maternal grandmother of the original index case was found to be mosaic for both deletional events, as well as having the normal AR gene. Karyotyping ruled out 47,XXX trisomy, indicating triple mosaicism for the two different deleted AR alleles and a normal AR allele. This triple mosaicism must have occurred early in embryonic development, as both deletions were passed on to different children. Based on these findings, we propose a novel mechanism of deletion formation. We suggest that during AR gene replication, a double strand DNA break occurred in intron 5, and that a variant of replication slippage occurred on both newly synthesized strands between the repeat motifs of microhomology, leading to the formation of the two different AR gene deletions.


Assuntos
Reparo do DNA , Replicação do DNA/fisiologia , Deleção de Genes , Receptores Androgênicos/genética , Síndrome de Resistência a Andrógenos/genética , Cromossomos Humanos X , Análise Mutacional de DNA , Éxons , Feminino , Ligação Genética , Heterozigoto , Humanos , Cariotipagem , Masculino , Dados de Sequência Molecular , Mosaicismo , Linhagem
13.
J Mol Endocrinol ; 57(2): 125-38, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27402875

RESUMO

The aim of this study was to investigate the direct muscle cell-mediated actions of androgens by comparing two different mouse lines. The cre-loxP system was used to delete the DNA-binding activity of the androgen receptor (AR) in mature myofibers (MCK mAR(ΔZF2)) in one model and the DNA-binding activity of the AR in both proliferating myoblasts and myofibers (α-actin mAR(ΔZF2)) in another model. We found that hind-limb muscle mass was normal in MCK mAR(ΔZF2) mice and that relative mass of only some hind-limb muscles was reduced in α-actin mAR(ΔZF2) mice. This suggests that myoblasts and myofibers are not the major cellular targets mediating the anabolic actions of androgens on male muscle during growth and development. Levator ani muscle mass was decreased in both mouse lines, demonstrating that there is a myofiber-specific effect in this unique androgen-dependent muscle. We found that the pattern of expression of genes including c-myc, Fzd4 and Igf2 is associated with androgen-dependent changes in muscle mass; therefore, these genes are likely to be mediators of anabolic actions of androgens. Further research is required to identify the major targets of androgen actions in muscle, which are likely to include indirect actions via other tissues.


Assuntos
Deleção de Genes , Músculos/metabolismo , Mioblastos/metabolismo , Miofibrilas/metabolismo , Receptores Androgênicos/genética , Animais , Biomarcadores , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Knockout , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Tamanho do Órgão , Especificidade de Órgãos/genética , Condicionamento Físico Animal , Receptores Androgênicos/metabolismo
14.
J Endocrinol ; 186(1): 21-31, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16002532

RESUMO

Androgen treatment can enhance the size and strength of muscle. However, the mechanisms of androgen action in skeletal muscle are poorly understood. This review discusses potential mechanisms by which androgens regulate satellite cell activation and function. Studies have demonstrated that androgen administration increases satellite cell numbers in animals and humans in a dose-dependent manner. Moreover, androgens increase androgen receptor levels in satellite cells. In vitro, the results are contradictory as to whether androgens regulate satellite cell proliferation or differentiation. IGF-I is one major target of androgen action in satellite cells. In addition, the possibility of non-genomic actions of androgens on satellite cells is discussed. In summary, this review focuses on exploring potential mechanisms through which androgens regulate satellite cells, by analyzing developments from research in this area.


Assuntos
Androgênios/fisiologia , Células Satélites de Músculo Esquelético/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Hipogonadismo/tratamento farmacológico , Hipogonadismo/metabolismo , Hipogonadismo/patologia , Fator de Crescimento Insulin-Like I/metabolismo , Mioblastos/citologia , Receptores Androgênicos/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/efeitos dos fármacos
15.
Neurol Res ; 27(5): 548-51, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15978183

RESUMO

OBJECTIVES: To investigate androgen receptor (AR) function in spinal and bulbar muscular atrophy (SBMA). METHODS: A kindred was identified with five individuals carrying the AR gene CAG repeat expansion that causes SBMA. Androgen binding was measured in cultured genital skin fibroblasts from three affected individuals. One newborn, pre-symptomatic, individual showed normal androgen binding, but two older, symptomatic individuals showed a decrease in androgen binding affinity. This difference was not related to AR CAG repeat size, as all affected individuals in this kindred had 49 repeats (normal range 6-35). Post-mortem analysis in one subject confirmed the signs of androgen insufficiency in the testis, with marked seminiferous tubule atrophy, and the absence of germinal cells. The characteristic neuronal depletion in the anterior horn gray matter was also observed. CONCLUSION: This report raises the possibility that age- or puberty-related changes in androgen binding could occur, which could potentially contribute to the progressive development of androgen resistance in affected men.


Assuntos
Envelhecimento/fisiologia , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Receptores Androgênicos/metabolismo , Repetições de Trinucleotídeos/genética , Adulto , Idoso , Southern Blotting , Saúde da Família , Feminino , Humanos , Masculino , Metribolona/farmacocinética , Pessoa de Meia-Idade , Atrofia Muscular Espinal/classificação , Atrofia Muscular Espinal/patologia , Reação em Cadeia da Polimerase/métodos , Mudanças Depois da Morte , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ensaio Radioligante/métodos , Receptores Androgênicos/genética , Testículo/fisiologia , Trítio/farmacocinética
16.
J Bone Miner Res ; 19(6): 882-92, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15125787

RESUMO

Genetic modification of mice is a powerful tool for the study of bone development and metabolism. This review discusses the advantages and disadvantages of various approaches used in bone-related research and the contributions these studies have made to bone biology. Genetic modification of mice is a powerful tool for the study of bone development and metabolism. This review discusses the advantages and disadvantages of various approaches used in bone-related research and the contributions these studies have made to bone biology. The approaches to genetic modification included in this review are (1) overexpression of genes, (2) global gene knockouts, (3) tissue-specific gene deletion, and (4) gene knock-in models. This review also highlights issues that should be considered when using genetically modified animal models, including the rigorous control of genetic background, use of appropriate control lines, and confirmation of tissue specificity of gene expression where appropriate. This technology provides a unique and powerful way to probe the function of genes and is already revolutionizing our approach to understanding the physiology of bone development and metabolism.


Assuntos
Animais Geneticamente Modificados , Osso e Ossos/metabolismo , Minerais/metabolismo , Modelos Animais , Animais , Camundongos , Camundongos Knockout , Fenótipo
17.
Hum Mutat ; 23(3): 287, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14974091

RESUMO

We have identified androgen receptor (AR) gene mutations in eight Australian subjects with complete androgen insensitivity syndrome (AIS). Four individuals, from three families, have novel mutations that introduce premature termination codons. Two siblings have the nonsense mutation Glu681X, and another subject has the nonsense mutation p.Ser884X. The other subject has a CA insertion at codon 829 (c.2847_2848insCA), causing a frameshift mutation that introduces four nonsense amino acids prior to a Stop codon. All the termination codons occur in the ligand binding domain, and cause reduced androgen binding in patient genital skin fibroblasts. Four further patients have missense mutations. One subject has two different mutations, p.Ala645Asp in the hinge region of the receptor, and p.Arg752Gln in the ligand binding domain. Both these mutations have previously been reported in patients with AIS, but the combination of these two mutations in one subject is unique. Another subject has a novel c.2533G>C transversion at the first nucleotide in exon 5, introducing the amino acid change p.Gly724Ala at a highly conserved residue in the ligand binding domain. Androgen binding is normal in fibroblasts from this subject, although other point mutations at this amino acid totally abolish binding. Two other subjects have mutations previously described as causing AIS, namely p.Arg779Trp and p.Val889Met substitutions in the ligand binding domain of the receptor. The p.Arg779Trp mutation is associated with the detection of a truncated AR protein in this patient's fibroblasts, suggesting the mutation renders the receptor susceptible to proteolysis.


Assuntos
Síndrome de Resistência a Andrógenos/genética , Mutação/genética , Receptores Androgênicos/genética , Adulto , Austrália , Criança , Pré-Escolar , Códon sem Sentido/genética , Códon de Terminação/genética , Feminino , Mutação da Fase de Leitura/genética , Disgenesia Gonadal 46 XY/genética , Humanos , Masculino
18.
Gene Expr Patterns ; 4(6): 633-6, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15465485

RESUMO

We have examined the expression of the basic helix-loop-helix factor Stra13 (DEC1/Sharp2) during endochondral bone development in the mouse. Stra13 expression was examined by in situ hybridization in the tibia from E14.5-E18.5, and at post-natal day 24. At E14.5, expression of Stra13 mRNA was very low, with expression limited to scattered hypertrophic chondrocytes. At E15.5 Stra13 mRNA was present in post-mitotic hypertrophic chondrocytes, co-localizing with collagen X expression. At E16.5-E18.5, Stra13 was expressed in both the proliferating chondrocytes and in the late hypertrophic chondrocytes. At E15.5-E18.5, Stra13 expression was also observed in the primary spongiosa. Stra13 expression was also maintained in the 24-day post-natal tibia, with expression detectable only in the late hypertrophic chondrocytes. Because Stra13 has been shown to be induced by hypoxia, and the growth plate is hypoxic during embryonic development, we compared the expression pattern of Stra13 and the HIF1-alpha target gene VEGF. VEGF is expressed predominantly in the late hypertrophic chondrocytes, with lower expression in the proliferating chondrocytes. Thus, there was a large degree of overlap in the expression patterns of Stra13 and VEGF in chondrocytes during embryonic development.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Cartilagem/citologia , Condrócitos/metabolismo , Colágeno Tipo X/biossíntese , Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese
19.
Asian J Androl ; 16(5): 675-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24713826

RESUMO

We aimed to determine the mechanisms of the anabolic actions of androgens in skeletal muscle by investigating potential androgen receptor (AR)-regulated genes in in vitro and in vivo models. The expression of the myogenic regulatory factor myogenin was significantly decreased in skeletal muscle from testosterone-treated orchidectomized male mice compared to control orchidectomized males, and was increased in muscle from male AR knockout mice that lacked DNA binding activity (AR(ΔZF2)) versus wildtype mice, demonstrating that myogenin is repressed by the androgen/AR pathway. The ubiquitin ligase Fbxo32 was repressed by 12 h dihydrotestosterone treatment in human skeletal muscle cell myoblasts, and c-Myc expression was decreased in testosterone-treated orchidectomized male muscle compared to control orchidectomized male muscle, and increased in AR(∆ZF2) muscle. The expression of a group of genes that regulate the transition from myoblast proliferation to differentiation, Tceal7 , p57(Kip2), Igf2 and calcineurin Aa, was increased in AR(∆ZF2) muscle, and the expression of all but p57(Kip2) was also decreased in testosterone-treated orchidectomized male muscle compared to control orchidectomized male muscle. We conclude that in males, androgens act via the AR in part to promote peak muscle mass by maintaining myoblasts in the proliferative state and delaying the transition to differentiation during muscle growth and development, and by suppressing ubiquitin ligase-mediated atrophy pathways to preserve muscle mass in adult muscle.


Assuntos
Proteínas Musculares/genética , Mioblastos Esqueléticos/metabolismo , Miogenina/genética , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos/genética , Proteínas Ligases SKP Culina F-Box/genética , Animais , Calcineurina/efeitos dos fármacos , Calcineurina/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p57/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p57/genética , Di-Hidrotestosterona/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas In Vitro , Fator de Crescimento Insulin-Like II/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas Musculares/efeitos dos fármacos , Músculo Esquelético , Mioblastos Esqueléticos/efeitos dos fármacos , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/genética , Orquiectomia , Proteínas Proto-Oncogênicas c-myc/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Proteínas Ligases SKP Culina F-Box/efeitos dos fármacos , Testosterona/farmacologia
20.
Mol Cell Endocrinol ; 348(1): 189-97, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-21872641

RESUMO

We tested the hypothesis that androgens have physiological actions via non-DNA binding-dependent androgen receptor (AR) signaling pathways in males, using our genetically modified mice that express a mutant AR with deletion of the 2nd zinc finger of the DNA binding domain (AR(ΔZF2)) that cannot bind DNA. In cultured genital skin fibroblasts, the mutant AR(ΔZF2) has normal ligand binding ability, phosphorylates ERK-1/2 in response to 1 min DHT treatment (blocked by the AR antagonist bicalutamide), but has reduced androgen-dependent nuclear localization compared to wildtype (WT). AR(ΔZF2) males have normal baseline ERK-1/2 phosphorylation, with a 1.5-fold increase in Akt phosphorylation in AR(ΔZF2) muscle vs WT. To identify physiological actions of non-DNA binding-dependent AR signaling, AR(ΔZF2) males were treated for 6 weeks with dihydrotestosterone (DHT). Cortical bone growth was suppressed by DHT in AR(ΔZF2) mice (6% decrease in periosteal and 7% decrease in medullary circumference vs untreated AR(ΔZF2) males). In conclusion, these data suggest that non-DNA binding dependent AR actions suppress cortical bone growth, which may provide a mechanism to fine-tune the response to androgens in bone.


Assuntos
Androgênios/fisiologia , DNA/metabolismo , Regulação da Expressão Gênica , Receptores Androgênicos/metabolismo , Androgênios/farmacologia , Animais , Núcleo Celular/metabolismo , Di-Hidrotestosterona/farmacologia , Fêmur/anatomia & histologia , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Expressão Gênica , Rim/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Músculo Esquelético/metabolismo , Especificidade de Órgãos , Ligação Proteica , Transporte Proteico , Receptores Androgênicos/genética , Elementos de Resposta , Deleção de Sequência , Gordura Subcutânea/metabolismo , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA