Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 7(1): 16406, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29180785

RESUMO

HSD17B1 is a steroid metabolising enzyme. We have previously generated knockout mice that had the entire coding region of Hsd17b1 replaced with lacZ-neo cassette (Hsd17b1-LacZ/Neo mice). This resulted in a 90% reduction of HSD17B1 activity, associated with severe subfertility in the knockout females. The present study indicates that Hsd17b1-LacZ/Neo male mice have a metabolic phenotype, including reduced adipose mass, increased lean mass and lipid accumulation in the liver. During the characterisation of this metabolic phenotype, it became evident that the expression of the Naglu gene, located closely upstream of Hsd17b1, was severely reduced in all tissues analysed. Similar results were obtained from Hsd17b1-LacZ mice after removing the neo cassette from the locus or by crossing the Hsd17b1-LacZ/Neo mice with transgenic mice constitutively expressing human HSD17B1. The deficiency of Naglu caused the accumulation of glycosaminoglycans in all studied mouse models lacking the Hsd17b1 gene. The metabolic phenotypes of the Hsd17b1 knockout mouse models were recapitulated in Naglu knockout mice. Based on the data we propose that the Hsd17b1 gene includes a regulatory element controlling Naglu expression and the metabolic phenotype in mice lacking the Hsd17b1 genomic region is caused by the reduced expression of Naglu rather than the lack of Hsd17b1.


Assuntos
17-Hidroxiesteroide Desidrogenases/genética , Alelos , Deleção de Genes , Estudos de Associação Genética , Doenças por Armazenamento dos Lisossomos/genética , Fenótipo , Animais , Modelos Animais de Doenças , Expressão Gênica , Loci Gênicos , Glicosaminoglicanos/metabolismo , Doenças por Armazenamento dos Lisossomos/diagnóstico , Doenças por Armazenamento dos Lisossomos/metabolismo , Lisossomos/metabolismo , Masculino , Camundongos , Mucopolissacaridose III/diagnóstico , Mucopolissacaridose III/genética , Mucopolissacaridose III/metabolismo
2.
FASEB J ; 29(9): 3806-16, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26018678

RESUMO

Hydroxysteroid (17ß)-dehydrogenase type 1 (HSD17B1) catalyzes the conversion of low active 17-ketosteroids, androstenedione (A-dione) and estrone (E1) to highly active 17-hydroxysteroids, testosterone (T) and E2, respectively. In this study, the importance of HSD17B1 in ovarian estrogen production was determined using Hsd17b1 knockout (HSD17B1KO) mice. In these mice, the ovarian HSD17B enzyme activity was markedly reduced, indicating a central role of HSD17B1 in ovarian physiology. The lack of Hsd17b activity resulted in increased ovarian E1:E2 and A-dione:T ratios, but we also observed reduced progesterone concentration in HSD17B1KO ovaries. Accordingly with the altered steroid production, altered expression of Star, Cyp11a1, Lhcgr, Hsd17b7, and especially Cyp17a1 was observed. The ovaries of HSD17B1KO mice presented with all stages of folliculogenesis, while the corpus luteum structure was less defined and number reduced. Surprisingly, bundles of large granular cells of unknown origin appeared in the stroma of the KO ovaries. The HSD17B1KO mice presented with severe subfertility and failed to initiate pseudopregnancy. However, the HSD17B1KO females presented with normal estrous cycle defined by vaginal smears and normal puberty appearance. This study indicates that HSD17B1 is a key enzyme in ovarian steroidogenesis and has a novel function in initiation and stabilization of pregnancy.


Assuntos
17-Hidroxiesteroide Desidrogenases/deficiência , Ciclo Estral , Infertilidade Feminina/enzimologia , Luteinização , Ovário/metabolismo , Progesterona/biossíntese , 17-Hidroxiesteroide Desidrogenases/biossíntese , 17-Hidroxiesteroide Desidrogenases/genética , 17-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Enzima de Clivagem da Cadeia Lateral do Colesterol/biossíntese , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Feminino , Infertilidade Feminina/genética , Masculino , Camundongos , Camundongos Knockout , Ovário/patologia , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Gravidez , Progesterona/genética , Maturidade Sexual/genética , Esteroide 17-alfa-Hidroxilase/biossíntese , Esteroide 17-alfa-Hidroxilase/genética
3.
Lab Anim ; 47(3): 134-45, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23479772

RESUMO

The use of genetically-modified (GM) animals as research models continues to grow. The completion of the mouse genome sequence, together with the high-throughput international effort to introduce mutations across the mouse genome in the embryonic stem (ES) cells (www.knockoutmouse.org) facilitates an efficient way to obtain mutated mouse strains as research models. The increasing number of available mutated mouses trains and their combinations, together with the increasing complexity in the targeting approaches used,reinforces the need for guidelines that will provide information about the mouse strains and the robust and reliable methods used for their genotyping. This information, however, should be obtained with a method causing minimal discomfort to the experimental animals. We have, therefore, compiled the present document which summarizes the currently available methods for obtaining genotype information. It provides updated guidelines concerning animal identification, DNA sampling and genotyping, and the information to be kept and distributed for any mutated rodent strain.


Assuntos
Animais Geneticamente Modificados/genética , Técnicas de Genotipagem/métodos , Camundongos/genética , Ratos/genética , Animais , Europa (Continente)
4.
FASEB J ; 27(4): 1342-50, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23239821

RESUMO

The human CYP19A1 gene is expressed in various tissues by the use of tissue-specific promoters, whereas the rodent cyp19a1 gene is expressed mainly in the gonads and brain. We generated a transgenic mouse model containing a >100-kb 5' region of human CYP19A1 gene connected to a luciferase reporter gene. The luciferase activity in mouse tissues mimicked the CYP19A1 gene expression pattern in humans. Interestingly, the reporter gene activity was 16 and 160 times higher in the urinary bladder and seminal vesicles, respectively, as compared with the activity in the testis. Accordingly, CYP19A1 gene and P450arom protein expression was detected in those human tissues. Moreover, the data revealed that the expression of CYP19A1 gene is driven by promoters PII, I.4, and I.3 in the seminal vesicles, and by promoters PII and I.4 in the urinary bladder. Furthermore, the reporter gene expression in the seminal vesicles was androgen dependent: Castration decreased the expression ∼20 times, and testosterone treatment restored it to the level of an intact mouse. This reporter mouse model facilitates studies of tissue-specific regulation of the human CYP19A1 gene, and our data provide evidence for seminal vesicles as important sites for estrogen production in males.


Assuntos
Androgênios/metabolismo , Aromatase/metabolismo , Glândulas Seminais/metabolismo , Bexiga Urinária/metabolismo , Androgênios/genética , Animais , Aromatase/genética , Regulação Enzimológica da Expressão Gênica/genética , Genes Reporter/genética , Humanos , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Sequências Reguladoras de Ácido Nucleico/genética , Testículo/metabolismo
5.
PLoS One ; 7(12): e53257, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300903

RESUMO

It is currently admitted that Follicle-Stimulating Hormone (FSH) is physiologically involved in the development and function of fetal/neonatal Sertoli cells in the rat but not the mouse. However, FSH is produced by both species from late fetal life onwards. We thus reinvestigated the role of FSH in mouse testis development at day 0 (birth) 6, 8 and 10 post-partum (dpp) by using mice that lack functional FSH receptors (FSH-R(-/-)). At birth, the number and proliferative index of Sertoli cells were significantly lower in FSH-R(-/-) mice than in wild type neonates. Claudin 11 mRNA expression also was significantly reduced in FSH-R(-/-) testes at 0 and 8 dpp, whereas the mRNA levels of other Sertoli cell markers (Transferrin and Desert hedgehog) were comparable in FSH-R(-/-) and wild type testes. Conversely, AMH mRNA and protein levels were higher at birth, comparable at 6 dpp and then significantly lower in FSH-R(-/-) testes at 8-10 dpp in FSH-R(-/-) mice than in controls. Although the plasma concentration of LH and the number of Leydig cells were similar in FSH-R(-/-) and control (wild type), testosterone concentration and P450c17 mRNA expression were significantly increased in FSH-R(-/-) testes at birth. Conversely, at 10 dpp when adult Leydig cells appear, expression of the steroidogenic genes P450scc, P450c17 and StAR was lower in FSH-R(-/-) testes than in controls. In conclusion, our results show that 1) like in the rat, signaling via FSH-R controls Sertoli cell development and function during late fetal life in the mouse as well; 2) paracrine factors produced by Sertoli cells are involved in the FSH-R-dependent regulation of the functions of fetal Leydig cells in late fetal life; and 3) the role of FSH-R signaling changes during the prepubertal period.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Receptores do FSH/metabolismo , Transdução de Sinais/fisiologia , Testículo/fisiologia , Animais , Células Intersticiais do Testículo/fisiologia , Masculino , Camundongos , Camundongos Knockout , Receptores do FSH/genética , Células de Sertoli/fisiologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Testosterona/sangue
6.
J Endocrinol ; 212(1): 27-40, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22045753

RESUMO

Disturbed action of sex steroid hormones, i.e. androgens and estrogens, is involved in the pathogenesis of various severe diseases in humans. Interestingly, recent studies have provided data further supporting the hypothesis that the circulating hormone concentrations do not explain all physiological and pathological processes observed in hormone-dependent tissues, while the intratissue sex steroid concentrations are determined by the expression of steroid metabolising enzymes in the neighbouring cells (paracrine action) and/or by target cells themselves (intracrine action). This local sex steroid production is also a valuable treatment option for developing novel therapies against hormonal diseases. Hydroxysteroid (17ß) dehydrogenases (HSD17Bs) compose a family of 14 enzymes that catalyse the conversion between the low-active 17-keto steroids and the highly active 17ß-hydroxy steroids. The enzymes frequently expressed in sex steroid target tissues are, thus, potential drug targets in order to lower the local sex steroid concentrations. The present review summarises the recent data obtained for the role of HSD17B1, HSD17B2, HSD17B7 and HSD17B12 enzymes in various metabolic pathways and their physiological and pathophysiological roles as revealed by the recently generated genetically modified mouse models. Our data, together with that provided by others, show that, in addition to having a role in sex steroid metabolism, several of these HSD17B enzymes possess key roles in other metabolic processes: for example, HD17B7 is essential for cholesterol biosynthesis and HSD17B12 is involved in elongation of fatty acids. Additional studies in vitro and in vivo are to be carried out in order to fully define the metabolic role of the HSD17B enzymes and to evaluate their value as drug targets.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Animais , Redes e Vias Metabólicas , Camundongos , Camundongos Transgênicos , Modelos Animais , Fenótipo
7.
Biol Reprod ; 85(4): 823-33, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21653891

RESUMO

Despite numerous observations of the effects of estrogens on spermatogenesis, identification of estrogen-regulated genes in the testis is limited. Using rats in which irradiation had completely blocked spermatogonial differentiation, we previously showed that testosterone suppression with gonadotropin-releasing hormone-antagonist acyline and the antiandrogen flutamide stimulated spermatogenic recovery and that addition of estradiol (E2) to this regimen accelerated this recovery. We report here the global changes in testicular cell gene expression induced by the E2 treatment. By minimizing the changes in other hormones and using concurrent data on regulation of the genes by these hormones, we were able to dissect the effects of estrogen on gene expression, independent of gonadotropin or testosterone changes. Expression of 20 genes, largely in somatic cells, was up- or downregulated between 2- and 5-fold by E2. The unexpected and striking enrichment of transcripts not corresponding to known genes among the E2-downregulated probes suggested that these might represent noncoding mRNAs; indeed, we have identified several as miRNAs and their potential target genes in this system. We propose that genes for which expression levels are altered in one direction by irradiation and in the opposite direction by both testosterone suppression and E2 treatment are candidates for controlling the block in differentiation. Several genes, including insulin-like 3 (Insl3), satisfied those criteria. If they are indeed involved in the inhibition of spermatogonial differentiation, they may be candidate targets for treatments to enhance recovery of spermatogenesis following gonadotoxic exposures, such as those resulting from cancer therapy.


Assuntos
Estradiol/uso terapêutico , Estrogênios/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Espermatogênese/efeitos da radiação , Testículo/efeitos dos fármacos , Testículo/metabolismo , Antagonistas de Androgênios/uso terapêutico , Animais , Cruzamentos Genéticos , Quimioterapia Combinada , Flutamida/uso terapêutico , Raios gama , Regulação da Expressão Gênica/efeitos da radiação , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/uso terapêutico , Insulina/genética , Insulina/metabolismo , Masculino , MicroRNAs/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligopeptídeos/uso terapêutico , Proteínas/genética , Proteínas/metabolismo , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Testículo/patologia , Testículo/efeitos da radiação , Testosterona/antagonistas & inibidores
8.
Endocrinology ; 151(4): 1893-901, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20130115

RESUMO

Hydroxysteroid (17beta) dehydrogenases (HSD17Bs) have a significant role in steroid metabolism by catalyzing the conversion between 17-keto and 17beta-hydroxysteroids. However, several studies in vitro have shown that some of these enzymes may also be involved in other metabolic pathways. Among these enzymes, HSD17B12 has been shown to be involved in both the biosynthesis of estradiol and the elongation of the essential very long fatty acids in vitro and in vivo. To investigate the function of mammalian HSD17B12 in vivo, we generated mice with a null mutation of the Hsd17b12 gene (HSD17B12KO mice) by using a gene-trap vector, resulting in the expression of the lacZ gene of the trapped allele. The beta-galactosidase staining of the heterozygous HSD17B12KO mice revealed that Hsd17b12 is expressed widely in the embryonic day (E) 7.5-E9.5 embryos, with the highest expression in the neural tissue. The HSD17B12KO mice die at E9.5 at latest and present severe developmental defects. Analysis of the knockout embryos revealed that the embryos initiate gastrulation, but organogenesis is severely disrupted. As a result, the E8.5-E9.5 embryos were void of all normal morphological structures. In addition, the inner cell mass of knockout blastocysts showed decreased proliferation capacity in vitro, and the amount of arachidonic acid was significantly decreased in heterozygous HSD17B12 ES cells. This, together with the expression pattern, suggests that in mouse, the HSD17B12 is involved in the synthesis of arachidonic acid and is essential for normal neuronal development during embryogenesis.


Assuntos
17-Hidroxiesteroide Desidrogenases/genética , Ácido Araquidônico/biossíntese , Gastrulação/genética , Organogênese/genética , Alelos , Animais , Morte Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Camundongos , Camundongos Knockout
9.
Endocrinology ; 151(4): 1884-92, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20185768

RESUMO

Hydroxysteroid (17beta) dehydrogenase 7 (HSD17B7) has been shown to catalyze the conversion of both estrone to estradiol (17-ketosteroid reductase activity) and zymosterone to zymosterol (3-ketosteroid reductase activity involved in cholesterol biosynthesis) in vitro. To define the metabolic role of the enzyme in vivo, we generated knockout mice deficient in the enzyme activity (HSD17B7KO). The data showed that the lack of HSD17B7 results in a blockage in the de novo cholesterol biosynthesis in mouse embryos in vivo, and HSD17BKO embryos die at embryonic day (E) 10.5. Analysis of neural structures revealed a defect in the development of hemispheres of the front brain with an increased apoptosis in the neuronal tissues. Morphological defects in the cardiovascular system were also observed from E9.5 onward. Mesodermal, endodermal, and hematopoietic cells were all detected by the histological analysis of the visceral yolk sac, whereas no organized vessels were observed in the knockout yolk sac. Immunohistological staining for platelet endothelial cell adhesion molecule-1 indicated that the complexity of the vasculature also was reduced in the HSD17B7KO embryos, particularly in the head capillary plexus and branchial arches. At E8.5-9.5, the heart development and the looping of the heart appeared to be normal in the HSD17B7KO embryos. However, at E10.5 the heart was dilated, and the thickness of the cardiac muscle and pericardium in the HSD17B7KO embryos was markedly reduced, and immunohistochemical staining for GATA-4 revealed that HSD17B7KO embryos had a reduced number of myocardial cells. The septum of the atrium was also defected in the knockout mice.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Diferenciação Celular/genética , Colesterol/biossíntese , Coração/embriologia , Placa Neural/embriologia , 17-Hidroxiesteroide Desidrogenases/genética , Animais , Apoptose/genética , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Miocárdio/enzimologia , Placa Neural/enzimologia , Saco Vitelino/irrigação sanguínea , Saco Vitelino/embriologia
10.
Biol Reprod ; 82(1): 54-65, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19684331

RESUMO

Although gonadotropins and androgen are required for normal spermatogenesis and both testosterone and follicle-stimulating hormone (FSH) are responsible for the inhibition of spermatogonial differentiation that occurs in irradiated rats, it has been difficult to identify the specific genes involved. To study specific hormonally regulated changes in somatic cell gene expression in the testis that may be involved in these processes, without the complication of changing populations of germ cells, we used irradiated LBNF(1) rats, the testes of which contain almost exclusively somatic cells except for a few type A spermatogonia. Three different groups of these rats were treated with various combinations of gonadotropin-releasing hormone antagonist, an androgen receptor antagonist (flutamide), testosterone, and FSH, and we compared the gene expression levels 2 wk later to those of irradiated-only rats by microarray analysis. By dividing the gene expression patterns into three major patterns and 11 subpatterns, we successfully distinguished, in a single study, the genes that were specifically regulated by testosterone, by luteinizing hormone (LH), and by FSH from the large number of genes that were not hormonally regulated in the testis. We found that hormones produced more dramatic upregulation than downregulation of gene expression: Testosterone had the strongest upregulatory effect, LH had a modest but appreciable upregulatory effect, and FSH had a minor upregulatory effect. We also separately identified the somatic cell genes that were chronically upregulated by irradiation. Thus, the present study identified gene expression changes that may be responsible for hormonal action on somatic cells to support normal spermatogenesis and the hormone-mediated block in spermatogonial development after irradiation.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica , Hormônio Luteinizante/metabolismo , Testículo/metabolismo , Testosterona/farmacologia , Animais , Flutamida/farmacologia , Raios gama , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Células Germinativas/efeitos dos fármacos , Células Germinativas/metabolismo , Células Germinativas/efeitos da radiação , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Oligopeptídeos/farmacologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/efeitos dos fármacos , Testículo/efeitos da radiação , Testosterona/sangue
11.
Endocrinology ; 150(11): 4941-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19797119

RESUMO

We have recently generated transgenic (TG) mice overexpressing human hydroxysteroid (17beta) dehydrogenase 2 enzyme (HSD17B2TG mice) under the ubiquitous chicken beta-actin promoter. As shown in the present study, the HSD17B2TG female mice presented with slower gain of body weight as compared with the wild-type (WT) littermates and suffered from ovarian dysfunction and mammary gland hyperplasia associated with increased expression of multiple pregnancy-associated genes. The macroscopic phenotype observed in the mammary gland was likely to be dependent on the increased progesterone and prolactin secretion, and a normal histological appearance was observed in HSD17B2TG mammary gland transplanted into a WT host. However, a significant suppression of several known estrogen target genes in the HSD17B2TG mammary transplants in WT females was observed, suggesting that HSD17B2 modulates estrogen action in vivo. Interestingly, the growth retardation of HSD17B2TG females was not efficiently rescued in the bi-TG mice expressing both HSD17B2 and HSD17B1 enzymes, and the bi-TG mice presented with certain masculinized phenotypes, including lack of nipples and closed vagina, recently reported for HSD17B1TG females. The present data suggest that HSD17B2 expression affects both sex steroid-independent and steroid-dependent pathways.


Assuntos
Estradiol Desidrogenases/genética , Estradiol Desidrogenases/metabolismo , Estrogênios/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Animais , Feminino , Expressão Gênica , Humanos , Masculino , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Camundongos , Camundongos Transgênicos , Ovário/crescimento & desenvolvimento , Ovário/patologia , Ovário/fisiopatologia
12.
Endocrinology ; 150(6): 2865-72, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19196801

RESUMO

Deteriorated male reproductive health has been connected to overexposure to estrogens or to imbalanced androgen-estrogen ratio. Transgenic male mice expressing human aromatase (AROM(+) mice) serve as an apt model for the study of the consequences of an altered androgen-estrogen ratio. Our previous studies with AROM(+) mice showed that low androgen levels together with high estrogen levels result in cryptorchidism and infertility. In the present study, the AROM(+) mice were shown to have severe abnormalities in the structure and function of Leydig cells before the appearance of spermatogenic failure. Decreased expression of adult-type Leydig cell markers (Ptgds, Vcam1, Insl3, Klk21, -24 and -27, Star, Cyp17a1, and Hsd17b3) indicated an immature developmental stage of the Leydig cells, which appears to be the first estrogen-dependent alteration. Genes involved in steroidogenesis (Star, Cyp17a1, and Hsd17b3) were suppressed despite normal LH levels. The low expression level of kallikreins 21, 24, and 27 potentially further inhibited Leydig cell function via remodeling extracellular matrix composition. In connection with disrupted steroidogenesis, Leydig cells showed enlarged mitochondria, a reduced amount of smooth endoplasmic reticulum, and an accumulation of cholesterol and precursors for cholesterol synthesis. The results of studies with AROM(+) mice crossed with estrogen receptor alpha or beta (ERalpha and ERbeta, respectively) knockout mice lead to the conclusion that the structural and functional disorders caused by estrogen exposure were mediated via ERalpha, whereas ERbeta was not involved.


Assuntos
Colesterol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Homeostase/fisiologia , Células Intersticiais do Testículo/metabolismo , Maturidade Sexual/fisiologia , Esteroides/metabolismo , Animais , Aromatase/genética , Aromatase/metabolismo , Estradiol Desidrogenases/metabolismo , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Estrogênios/farmacologia , Hormônio Foliculoestimulante/sangue , Homeostase/efeitos dos fármacos , Células Intersticiais do Testículo/efeitos dos fármacos , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Fosfoproteínas/metabolismo , Maturidade Sexual/efeitos dos fármacos , Esteroide 17-alfa-Hidroxilase/metabolismo , Testosterona/metabolismo
13.
J Androl ; 30(4): 440-51, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19136390

RESUMO

Irradiation of LBNF(1) rat testes induces spermatogonial differentiation arrest, which can be reversed by gonadotropin-releasing hormone (GnRH) antagonist-induced suppression of intratesticular testosterone (ITT) and follicle-stimulating hormone (FSH). Although exogenous estrogen treatment also enhanced spermatogenic recovery, as measured by the tubule differentiation index (TDI), it was not clear whether estrogen stimulated spermatogonial differentiation only by further suppressing ITT or by an additional independent mechanism as well. To resolve this question, we performed the following experiments. At 15 weeks after irradiation, rats were treated with GnRH antagonist; some also received 17beta-estradiol (E2) and were killed 4 weeks later. GnRH antagonist treatment increased the TDI from 0% to 8%, and addition of E2 further increased the TDI to 39%. However, E2 addition further reduced ITT from 7 ng/g testis, observed with GnRH antagonist to 3 ng/g testis, so decreased ITT levels might have contributed to recovery. Next GnRH antagonist-treated rats were given exogenous testosterone and flutamide to stabilize ITT levels and block its action. This increased TDI slightly from 8% to 13%, but the further addition of E2 significantly raised the TDI to 27%, indicating it acted by a mechanism independent of ITT levels. Plots of TDI for all treatment groups compared with ITT, FSH, or a linear combination of ITT and FSH showed that treatments including E2 produced higher TDI values than did treatments without E2. These results indicate that there was an effect of E2 on spermatogonial differentiation because of an additional direct action on the testis that is unrelated to its suppression of testosterone or gonadotropins.


Assuntos
Estradiol/farmacologia , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/efeitos da radiação , Antagonistas de Androgênios/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Estradiol/metabolismo , Flutamida/farmacologia , Hormônio Foliculoestimulante/sangue , Masculino , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Espermatogênese/efeitos da radiação , Espermatogônias/efeitos dos fármacos , Testosterona/antagonistas & inibidores , Testosterona/farmacologia
14.
Biol Reprod ; 80(2): 320-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18987330

RESUMO

Advances in treatment for testicular cancer that include the coadministration of bleomycin, etoposide, and cisplatin (BEP) have brought the cure rate to higher than 90%%. The goal of this study was to elucidate the impact of BEP treatment on gene expression in male germ cells. Brown-Norway rats were treated for 9 wk with vehicle (0x) or BEP at doses equivalent to 0.3x and 0.6x the human dose. At the end of treatment, spermatogenesis was affected, showing altered histology and a decreased sperm count; spermatozoa had a higher number of DNA breaks. After 9 wk of treatment, round spermatids were isolated, and RNA was extracted and probed on Rat230-2.0 Affymetrix arrays. Of the 31 099 probe sets present on the array, 59%% were expressed in control round spermatids. BEP treatment significantly altered the expression of 221 probe sets, with at least a 1.5-fold change compared with controls; 80% were upregulated. We observed a dose-dependent increase in the expression of oxidative stress response genes and no change in the expression of genes involved in DNA repair. BEP upregulated genes were implicated in pathways related to Jun and Junb protooncogenes. Increased mRNA levels of Jun and Junb were confirmed by quantitative RT-PCR; furthermore, JUN protein was increased in elongating spermatids. Thus, BEP exposure triggers an oxidative stress response in round spermatids and induces many pathways that may lead to the survival of damaged cells and production of abnormal sperm.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Células Germinativas/efeitos dos fármacos , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Testiculares/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Bleomicina/efeitos adversos , Bleomicina/uso terapêutico , Cisplatino/efeitos adversos , Cisplatino/uso terapêutico , Análise por Conglomerados , Etoposídeo/efeitos adversos , Etoposídeo/uso terapêutico , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Células Germinativas/metabolismo , Masculino , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Endogâmicos BN , Espermátides/efeitos dos fármacos , Espermátides/metabolismo , Espermatogênese/efeitos dos fármacos , Espermatogênese/genética , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Sistema Urogenital/efeitos dos fármacos , Sistema Urogenital/metabolismo
15.
Gen Comp Endocrinol ; 156(3): 595-602, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18377904

RESUMO

The biological activity and spatio-temporal expression patterns of the gonadotropin receptors FSH-R and LH-R were examined in the repetitive spawner Atlantic halibut to elucidate the gonadotropic regulation of the asynchronous follicle development. The cloned receptors were expressed in mammalian COS-7 cells, and stimulation with sea bass FSH and LH increased the cAMP production. The halibut FSH-R and LH-R genes were shown to be highly expressed in the gonads of sexually mature fish, but the transcripts were also found in extra-gonadal tissues such as pituitary and brain. Different expression patterns of FSH-R and LH-R in the developing follicles were documented by semi-quantitative RT-PCR. Abundant FSH-R mRNA was found in the small follicles during primary growth and vitellogenesis, and the signals were localized to the granulosa cells by in situ hybridization. In contrast, follicular LH-R mRNA was hardly detectable during the early stages. Conversely, in follicles during final maturation FSH-R mRNA levels tended to decrease, while the expression of LH-R was highly upregulated. Whereas the pituitary FSH and LH are asynchronously expressed in annual spawners, both gonadotropins were expressed in the female halibut pituitary throughout the reproductive cycle, except in the prespawning females. Hence, the sequential gonadotropic activation of ovarian follicle growth and maturation in repetitive spawners is probably regulated by modulating the temporal expression of FSH-R and LH-R in the follicle membrane.


Assuntos
Linguado/fisiologia , Oogênese/fisiologia , Folículo Ovariano/metabolismo , Receptores do FSH/metabolismo , Receptores do LH/metabolismo , Actinas/biossíntese , Actinas/genética , Animais , Células COS , Chlorocebus aethiops , AMP Cíclico/fisiologia , Feminino , Masculino , Folículo Ovariano/fisiologia , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Hipófise/crescimento & desenvolvimento , Hipófise/metabolismo , Receptores do FSH/biossíntese , Receptores do FSH/genética , Receptores do LH/biossíntese , Receptores do LH/genética , Reprodução , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual
16.
Mol Endocrinol ; 22(3): 665-75, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18048640

RESUMO

Hydroxysteroid (17-beta) dehydrogenase 2 (HSD17B2) is a member of aldo-keto reductase superfamily, known to catalyze the inactivation of 17beta-hydroxysteroids to less active 17-keto forms and catalyze the conversion of 20alpha-hydroxyprogesterone to progesterone in vitro. To examine the role of HSD17B2 in vivo, we generated mice deficient in Hsd17b2 [HSD17B2 knockout (KO)] by a targeted gene disruption in embryonic stem cells. From the homozygous mice carrying the disrupted Hsd17b2, 70% showed embryonic lethality appearing at the age of embryonic d 11.5 onward. The embryonic lethality was associated with reduced placental size measured at embryonic d 17.5. The HSD17B2KO mice placentas presented with structural abnormalities in all three major layers: the decidua, spongiotrophoblast, and labyrinth. Most notable was the disruption of the spongiotrophoblast and labyrinthine layers, together with liquid-filled cysts in the junctional region and the basal layer. Treatments with an antiestrogen or progesterone did not rescue the embryonic lethality or the placenta defect in the homozygous mice. In hybrid background used, 24% of HSD17B2KO mice survived through the fetal period but were born growth retarded and displayed a phenotype in the brain with enlargement of ventricles, abnormal laminar organization, and increased cellular density in the cortex. Furthermore, the HSD17B2KO mice had unilateral renal degeneration, the affected kidney frequently appearing as a fluid-filled sac. Our results provide evidence for a role for HSD17B2 enzyme in the cellular organization of the mouse placenta.


Assuntos
Estradiol Desidrogenases/genética , Placenta/anormalidades , Placenta/enzimologia , 17-Hidroxiesteroide Desidrogenases , Animais , Encéfalo/anormalidades , Estradiol/análogos & derivados , Estradiol/análise , Estradiol/farmacologia , Estradiol Desidrogenases/metabolismo , Antagonistas de Estrogênios/farmacologia , Feminino , Morte Fetal/enzimologia , Fulvestranto , Histocitoquímica , Rim/anormalidades , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Testosterona/análise
17.
Endocrinology ; 147(7): 3563-70, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16627582

RESUMO

The jsd mice experience a single wave of spermatogenesis, followed by an arrest of spermatogenesis because of a block in spermatogonial differentiation. Previous pharmacological and surgical studies have indicated that testosterone (T) and low scrotal temperatures but not FSH block spermatogonial differentiation in jsd mice. We sought to test these observations by genetic approaches by producing male jsd mutant mice with either defective androgen receptor (AR, Tfm mutation) or a deficiency of FSH (fshb(-/-)). In adult jsd-Tfm double-mutant mice, the tubule differentiation index was 95% compared with 14% in jsd littermates, suggesting that general ablation of AR function restored spermatogonial differentiation in jsd mice. The results indicated that this enhancement of differentiation was primarily a result of elevation of temperature caused by the cryptorchid position of the testis in jsd-Tfm double-mutant mice, which resulted from the lack of AR in the gubernaculum. The low levels of T were not a factor in the release of the spermatogonial differentiation block in the jsd-Tfm mice, but we were unable to determine whether inactivation of AR in the adult jsd testis had a direct effect on the restoration of spermatogonial differentiation because the elevated temperature bypassed the T-induced block in spermatogonial differentiation. Although spermatogonia were indeed present in adult jsd-fshb double-mutant mice and were capable of differentiation after androgen deprivation, these mice had a tubule differentiation index of 0%, ruling out the possibility that endogenous FSH inhibited spermatogonial differentiation in jsd mice. The results are consistent in support of the hypothesis that inhibition of spermatogonial differentiation in jsd mice is a result of T acting through the AR only at scrotal temperatures.


Assuntos
Hormônio Foliculoestimulante/genética , Mutação , Receptores Androgênicos/genética , Ribonucleoproteínas Nucleolares Pequenas/genética , Espermatogônias/citologia , Animais , Diferenciação Celular , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ribonucleoproteínas Nucleolares Pequenas/fisiologia , Espermatogônias/metabolismo , Testículo/metabolismo
18.
Endocrinology ; 147(1): 472-82, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16210366

RESUMO

Simultaneous suppression of both testosterone and FSH with GnRH antagonists (GnRH-ant) reverses the radiation-induced block in spermatogonial differentiation in F1 hybrids of Lewis and Brown-Norway rats. Although addition of exogenous testosterone restores the block, it also raises FSH, and hence it had not been possible to conclusively determine which hormone was inhibiting spermatogonial differentiation. In the present study, we establish the relative roles of testosterone and FSH in this inhibition using three different approaches. The first approach involved the treatment of irradiated rats, in which differentiation was stimulated by GnRH-ant plus flutamide, with FSH for 2 wk; the FSH reduced the percentage of tubules that were differentiated (TDI) by about 2-fold, indicating that FSH does have an inhibitory role. The second approach involved treatment of irradiated, hypophysectomized rats with exogenous testosterone for 10 wk; testosterone also reduced the TDI, demonstrating that testosterone had a definite inhibitory effect, independent of pituitary hormones. Furthermore, in this protocol we showed that TDI in the hypophysectomized testosterone-treated group, which had higher intratesticular testosterone levels but lacked FSH, was slightly higher than the TDI in a GnRH-antagonist-testosterone-treated group of irradiated rats, which had normal physiological levels of FSH; this result supports a role for endogenous FSH in suppressing spermatogonial differentiation in the latter group. The third approach involved injection of an active anti-FSH antibody for 10 d in untreated, GnRH-ant plus flutamide-treated, or GnRH-ant plus testosterone-treated irradiated rats. This was not sufficient to increase the TDI. However, flutamide given in a similar treatment schedule did increase the TDI in GnRH-ant plus testosterone-treated rats. We conclude that both testosterone and FSH individually inhibit spermatogonial differentiation after irradiation, but testosterone is a more highly potent inhibitor than is FSH.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Espermatogônias/citologia , Espermatogônias/efeitos da radiação , Testosterona/farmacologia , Animais , Flutamida/farmacologia , Humanos , Hipofisectomia , Masculino , Ratos , Proteínas Recombinantes/farmacologia , Espermatogônias/efeitos dos fármacos
19.
Mol Hum Reprod ; 11(8): 601-5, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16100240

RESUMO

FSH is a pituitary gonadotropin that along with LH plays a key role in the regulation of gonadal function. The gonadotropic hormones are composed of two subunits, the common alpha subunit and the hormone-specific beta subunit, which determines the binding to specific receptors and induction of biological response. Unlike the LHbeta gene, shown in earlier studies to harbour several amino acid-altering polymorphisms and mutations, information about the eventual sequence variation of the human FSHbeta subunit is not available. In this study, we made sequence analysis and comparison of polymorphisms found in FSHbeta in two Caucasian populations, the Finns and the Danes. It was found that FSHbeta subunit is highly conserved in these populations. Compared with the published sequences, only three silent polymorphisms were detected in the coding regions of the gene, and the promoter sequence was completely identical with the reported sequence. Two of the polymorphisms found were novel, one in the Finnish and one in the Danish population. The results of the sequence analysis show that the human FSHbeta gene is highly conserved and amino acid changing mutations are apparently extremely rare, at least in the samples collected randomly from control populations. This may be due to the crucial role of normal FSH function in the regulation of fertility.


Assuntos
Sequência Conservada , Subunidade beta do Hormônio Folículoestimulante/genética , Sequência de Aminoácidos , Sequência de Bases , Genótipo , Humanos , Mutação , Polimorfismo Genético , Regiões Promotoras Genéticas
20.
Endocrinology ; 146(5): 2454-61, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15661855

RESUMO

It is now accepted that estrogens play a role in male fertility and that exposure to exogenous estrogens during fetal/neonatal life can lead to reproductive disorders in the male. However, the estrogen receptor (ER)-mediated processes involved in the regulation of male reproduction during fetal and neonatal development are still largely unclear. We previously reported that ER beta deficiency affects gametogenesis in mice but changes neither the number nor the differentiated functions of fetal Leydig cells. We show here that ER alpha-deficient mice (ER alpha-/-) display higher levels of testicular testosterone secretion than wild-type mice from fetal d 13.5 onwards. This results from higher levels of steroidogenic activity per fetal Leydig cell, as indicated by the hypertrophy of these cells and the higher levels of mRNA for StAR, P450c17 and P450scc in the testis, for a similar number of Leydig cells. Because LH is not produced on fetal d 13.5 and because no change in plasma LH concentration was observed in 2-d-old ER alpha-deficient mice, LH is probably not involved in the effects of estrogens on testicular steroidogenesis in fetal and early neonatal Leydig cells. Furthermore, inactivation of ER beta did not change the effect of ER alpha inactivation on steroidogenesis. Lastly, in an organ culture system, 1 mum diethylstilbestrol decreased the testosterone secretion of wild-type fetal and neonatal testes but not of ER alpha-/- testes. Thus, this study shows that endogenous estrogens physiologically inhibit steroidogenesis via ER alpha by acting directly on the testis early in fetal and neonatal development.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Estrogênios/fisiologia , Feto/citologia , Células Intersticiais do Testículo/fisiologia , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Dietilestilbestrol/farmacologia , Receptor alfa de Estrogênio/deficiência , Feminino , Células Intersticiais do Testículo/química , Hormônio Luteinizante/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Fosfoproteínas/genética , Gravidez , RNA Mensageiro/análise , Esteroide 17-alfa-Hidroxilase/genética , Testículo/embriologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Testosterona/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA