Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 381
Filtrar
1.
Andrology ; 5(3): 578-588, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28235253

RESUMO

Activin A is an important regulator of testicular and epididymal development and function, as well as inflammation and immunity. In the adult murine reproductive tract, activin A mRNA (Inhba) expression levels are highest in the caput epididymis and decrease progressively towards the distal vas deferens. The activin-binding protein, follistatin (FST), shows the opposite expression pattern, with exceptionally high levels of the Fst288 mRNA variant in the vas deferens. This unique pattern of expression suggests that activin A and follistatin, in particular FST288, play region-specific roles in regulating the epididymis and vas deferens. The cellular distribution of activin and follistatin and structural organization of the male reproductive tract was examined in wild-type and transgenic (TghFST315) mice lacking FST288. Compared to wild-type littermates, TghFST315 mice showed a 50% reduction in serum follistatin and a significant elevation of both activin A and B. Testicular, epididymal and seminal vesicle weights were reduced, but intra-testicular testosterone was normal. A decrease in the epididymal duct diameter in the corpus and thickening of the peritubular smooth muscle in the cauda, together with increased coiling of the proximal vas deferens, were observed in TghFST315 mice. No immune cell infiltrates were detected. Immunohistochemistry indicated that epithelial cells are the main source of activins and follistatin in the epididymis and vas deferens. Activin A, but not activin B, was also localized to sperm heads in the lumen of the epididymis and vas deferens. Expression of Inhba and another immunoregulatory gene, indoleamine-2,3-dioxygenase (Ido-1), was increased approximately twofold in the TghFST315 caput epididymis, but several other genes associated with immunoregulation, inflammation or fibrosis were unaffected. Our novel data indicate that disruption of follistatin expression has significant effects on the testis and epididymis, and suggest an association between activin A and indoleamine-2,3-dioxygenase in the caput epididymis, with implications for the epididymal immunoenvironment.


Assuntos
Ativinas/metabolismo , Folistatina/metabolismo , Genitália Masculina/metabolismo , Animais , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase
2.
Hum Reprod Update ; 22(3): 342-57, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26884470

RESUMO

BACKGROUND: Activins are members of the pleiotrophic family of the transforming growth factor-beta (TGF-ß) superfamily of cytokines, initially isolated for their capacity to induce the release of FSH from pituitary extracts. Subsequent research has demonstrated that activins are involved in multiple biological functions including the control of inflammation, fibrosis, developmental biology and tumourigenesis. This review summarizes the current knowledge on the roles of activin in reproductive and developmental biology. It also discusses interesting advances in the field of modulating the bioactivity of activins as a therapeutic target, which would undoubtedly be beneficial for patients with reproductive pathology. METHODS: A comprehensive literature search was carried out using PUBMED and Google Scholar databases to identify studies in the English language which have contributed to the advancement of the field of activin biology, since its initial isolation in 1987 until July 2015. 'Activin', 'testis', 'ovary', 'embryonic development' and 'therapeutic targets' were used as the keywords in combination with other search phrases relevant to the topic of activin biology. RESULTS: Activins, which are dimers of inhibin ß subunits, act via a classical TGF-ß signalling pathway. The bioactivity of activin is regulated by two endogenous inhibitors, inhibin and follistatin. Activin is a major regulator of testicular and ovarian development. In the ovary, activin A promotes oocyte maturation and regulates granulosa cell steroidogenesis. It is also essential in endometrial repair following menstruation, decidualization and maintaining pregnancy. Dysregulation of the activin-follistatin-inhibin system leads to disorders of female reproduction and pregnancy, including polycystic ovary syndrome, ectopic pregnancy, miscarriage, fetal growth restriction, gestational diabetes, pre-eclampsia and pre-term birth. Moreover, a rise in serum activin A, accompanied by elevated FSH, is characteristic of female reproductive aging. In the male, activin A is an autocrine and paracrine modulator of germ cell development and Sertoli cell proliferation. Disruption of normal activin signalling is characteristic of many tumours affecting reproductive organs, including endometrial carcinoma, cervical cancer, testicular and ovarian cancer as well as prostate cancer. While activin A and B aid the progression of many tumours of the reproductive organs, activin C acts as a tumour suppressor. Activins are important in embryonic induction, morphogenesis of branched glandular organs, development of limbs and nervous system, craniofacial and dental development and morphogenesis of the Wolffian duct. CONCLUSIONS: The field of activin biology has advanced considerably since its initial discovery as an FSH stimulating agent. Now, activin is well known as a growth factor and cytokine that regulates many aspects of reproductive biology, developmental biology and also inflammation and immunological mechanisms. Current research provides evidence for novel roles of activins in maintaining the structure and function of reproductive and other organ systems. The fact that activin A is elevated both locally as well as systemically in major disorders of the reproductive system makes it an important biomarker. Given the established role of activin A as a pro-inflammatory and pro-fibrotic agent, studies of its involvement in disorders of reproduction resulting from these processes should be examined. Follistatin, as a key regulator of the biological actions of activin, should be evaluated as a therapeutic agent in conditions where activin A overexpression is established as a contributing factor.


Assuntos
Ativinas/fisiologia , Ovário/fisiologia , Reprodução/fisiologia , Testículo/fisiologia , Ativinas/química , Animais , Feminino , Folistatina/química , Folistatina/fisiologia , Glicoproteínas , Humanos , Subunidades beta de Inibinas , Inibinas/química , Inibinas/fisiologia , Masculino , Neoplasias Ovarianas , Pré-Eclâmpsia , Gravidez
3.
Reprod Fertil Dev ; 27(7): 985-99, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24630125

RESUMO

Female mice lacking the follistatin gene but expressing a human follistatin-315 transgene (tghFST315) have reproductive abnormalities (reduced follicles, no corpora lutea and ovarian-uterine inflammation). We hypothesised that the absence of follistatin-288 causes the abnormal reproductive tract via both developmental abnormalities and abnormal ovarian activity. We characterised the morphology of oviducts and uteri in wild type (WT), tghFST315 and follistatin-knockout mice expressing human follistatin-288 (tghFST288). The oviducts and uteri were examined in postnatal Day-0 and adult mice (WT and tghFST315 only) using histology and immunohistochemistry. Adult WT and tghFST315 mice were ovariectomised and treated with vehicle, oestradiol-17ß (100ng injection, dissection 24h later) or progesterone (1mg×three daily injections, dissection 24h later). No differences were observed in the oviducts or uteri at birth, but abnormalities developed by adulthood. Oviducts of tghFST315 mice failed to coil, the myometrium was disorganised, endometrial gland number was reduced and oviducts and uteri contained abundant leukocytes. After ovariectomy, tghFST315 mice had altered uterine cell proliferation, and inflammation was maintained and exacerbated by oestrogen. These studies show that follistatin is crucial to postnatal oviductal-uterine development and function. Further studies differentiating the role of ovarian versus oviductal-uterine follistatin in reproductive tract function at different developmental stages are warranted.


Assuntos
Folistatina/genética , Oviductos/crescimento & desenvolvimento , Útero/crescimento & desenvolvimento , Animais , Proliferação de Células/genética , Endométrio/crescimento & desenvolvimento , Endométrio/metabolismo , Estrogênios/farmacologia , Feminino , Folistatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miométrio/crescimento & desenvolvimento , Miométrio/metabolismo , Ovariectomia , Oviductos/diagnóstico por imagem , Oviductos/metabolismo , Útero/efeitos dos fármacos , Útero/metabolismo
4.
Cytokine Growth Factor Rev ; 24(3): 285-95, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23541927

RESUMO

The activins, as members of the transforming growth factor-ß superfamily, are pleiotrophic regulators of cell development and function, including cells of the myeloid and lymphoid lineages. Clinical and animal studies have shown that activin levels increase in both acute and chronic inflammation, and are frequently indicators of disease severity. Moreover, inhibition of activin action can reduce inflammation, damage, fibrosis and morbidity/mortality in various disease models. Consequently, activin A and, more recently, activin B are emerging as important diagnostic tools and therapeutic targets in inflammatory and fibrotic diseases. Activin antagonists such as follistatin, an endogenous activin-binding protein, offer considerable promise as therapies in conditions as diverse as sepsis, liver fibrosis, acute lung injury, asthma, wound healing and ischaemia-reperfusion injury.


Assuntos
Ativinas/fisiologia , Folistatina/metabolismo , Ativinas/biossíntese , Animais , Asma/tratamento farmacológico , Modelos Animais de Doenças , Feminino , Fibrose , Folistatina/uso terapêutico , Humanos , Inflamação/tratamento farmacológico , Inibinas/fisiologia , Masculino , Cicatrização/efeitos dos fármacos
5.
Reprod Biomed Online ; 24(3): 364-74, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22285243

RESUMO

Follistatin, an inhibitor of activin A, has key regulatory roles in the female reproductive tract. Follistatin has two splice variants: FST288, largely associated with cell surfaces, and FST315, the predominant circulating form. The mechanism regulating uterine expression of these variants is unknown. Quantitative RT-PCR was used to measure expression of follistatin splice variants (Fst288, Fst315), the activin bA subunit (Inhba) and the inhibin a subunit (Inha) in uterine tissues during early pregnancy (days 1­4, preimplantation) and in response to exogenous 17b-oestradiol (single s.c. injection) and progesterone (three daily s.c. injections) in ovariectomized mice. Uterine Fst288, Fst315 and Inhba expression increased during early pregnancy, with greater increases in Fst315 relative to Fst288 suggesting differential regulation of these variants. Fst288, Fst315, Inhba and Inha all increased in response to progesterone treatment. Fst288, but not Fst315, mRNA decreased in response to 17b-oestradiol treatment, whereas Inhba increased. A comparison of the absolute concentrations of uterine follistatin mRNA using crossing thresholds indicated that both variants were more highly expressed in early pregnancy in contrast to the hormone treatment models. It is concluded that progesterone regulates uterine expression of both follistatin variants, as well as activin A, during early pregnancy in the mouse uterus


Assuntos
Folistatina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Progesterona/farmacologia , Útero/efeitos dos fármacos , Animais , Estradiol/farmacologia , Feminino , Folistatina/química , Folistatina/genética , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/metabolismo , Inibinas/genética , Inibinas/metabolismo , Camundongos , Gravidez , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Útero/metabolismo
6.
Mamm Genome ; 22(3-4): 235-48, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21347845

RESUMO

An N-ethyl-N-nitrosourea random mutation screen was used to identify recessive modifiers of gene silencing in the mouse using an epigenetically sensitive reporter transgene. One of the mutant lines, MommeR1, was identified as a suppressor of variegation and it showed female-specific age-associated infertility in homozygotes. Linkage analysis identified a region on chromosome 10, containing the Foxo3a gene, previously shown to play a critical role in female gametogenesis. Foxo3a is a transcription factor with roles in cell cycle control, apoptosis, neural and hematopoietic cell differentiation, and DNA repair. Sequencing of the Foxo3a gene in MommeR1 mice revealed a point mutation that causes an amino acid substitution in the highly conserved Forkhead DNA-binding domain. In vitro transcription assays showed that the point mutation causes loss of FOXO3a transactivation activity. Compound heterozygotes made with Foxo3a-null mice (carrying the targeted deletion of exon 2) displayed complementation with respect to both the activation of the reporter transgene and defects in folliculogenesis similar to those seen in MommeR1 homozygotes, supporting the conclusion that this is the causative mutation. Approximately one in six female MommeR1 homozygotes develop teratomas, a phenotype not reported in Foxo3a-null mice. Ovulated oocytes from MommeR1 homozygotes display a number of abnormalities. The MommeR1 mice provide a novel platform to investigate teratocarcinogenesis and link Foxo3a with parthenogenesis and ovarian cancer. The finding of Foxo3a as a modifier of epigenetic reprogramming is discussed.


Assuntos
Fatores de Transcrição Forkhead/genética , Mutação de Sentido Incorreto , Oócitos/citologia , Neoplasias Ovarianas/genética , Teratoma/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Modelos Animais de Doenças , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Inativação Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Oócitos/metabolismo , Neoplasias Ovarianas/metabolismo , Mutação Puntual , Teratoma/metabolismo
7.
Hum Reprod ; 23(9): 2151-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18550510

RESUMO

BACKGROUND: Cysteine-rich secretory protein 2 (CRISP2) is localized to the human sperm acrosome and tail. It can regulate ryanodine receptors Ca(2+) gating and binds to mitogen-activated protein kinase kinase kinase 11 in the acrosome and gametogenetin 1 (GGN1) in the tail. METHODS AND RESULTS: In order to test the hypothesis that CRISP2 variations contribute to male infertility, we screened coding and flanking intronic regions in 92 infertile men with asthenozoo- and/or teratozoospermia and 176 control men using denaturing HPLC and sequencing. There were 21 polymorphisms identified, including 13 unreported variations. Three SNPs resulted in amino acid substitutions: L59V, M176I and C196R. All were only present in a heterozygous state and found in fertile men. However, the C196R polymorphism was of particular interest as it resulted in the loss of a strictly conserved cysteine involved in intramolecular disulphide bonding. Screening of an additional 637 infertile men identified 23 heterozygous C196R men to give an overall frequency of 3.6%, compared with 3.4% in control men. The functional significance of the C196R polymorphism was defined using a yeast two-hybrid assay. The C196R substitution resulted in the loss of CRISP2-GGN1 binding. CONCLUSIONS: Although none of the many polymorphisms identified herein showed a significant association with male infertility, functional studies suggested that the C196R polymorphism may compromise CRISP2 function.


Assuntos
Glicoproteínas/genética , Infertilidade Masculina/genética , Polimorfismo Genético , Substituição de Aminoácidos , Austrália , Estudos de Casos e Controles , Moléculas de Adesão Celular , Cromatografia Líquida de Alta Pressão , Glicoproteínas/química , Glicoproteínas/fisiologia , Heterozigoto , Humanos , Masculino , Análise de Sequência de DNA , Testículo/metabolismo
8.
Hum Reprod ; 22(1): 2-16, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16887924

RESUMO

Testicular biopsy is a crucial assessment in reproductive practice with diagnostic and prognostic importance for assisted reproductive technologies (ARTs) and risk of testicular neoplasia. Endocrine and genetic tests cannot reliably distinguish obstructive azoospermia (OA) from non-obstructive azoospermia (NOA) or predict recovery of mature spermatids by testicular sperm extraction (TESE). Currently, divergent histological reporting systems and the use of imprecise terminology seriously degrade the value of the literature on TESE recovery rates and hamper evaluation of treatments and research on genotype-phenotype relationships. The rising incidence of testis cancer and carcinoma in situ (CIS), especially in infertile populations, requires that every effort be made for its early detection. We provide a systematic approach to the histological classification of spermatogenic disorders and detection of CIS in adult patients. We evaluate a large consecutive series of bilateral biopsies from infertile men and report (i) the frequency of bilateral or discordant patterns that supports the use of bilateral biopsy for comprehensive evaluation and (ii) a high prevalence of mixed patterns, particularly within the hypospermatogenesis classification, that helps account for reported success of TESE. We propose a new diagnosis code for testicular biopsies that addresses the needs of ART clinicians and allows data storage and retrieval of value in clinical practice and research.


Assuntos
Doenças Testiculares/classificação , Testículo/patologia , Azoospermia/patologia , Biópsia/classificação , Biópsia por Agulha Fina , Carcinoma in Situ/diagnóstico , Carcinoma in Situ/patologia , Humanos , Infertilidade Masculina/classificação , Infertilidade Masculina/patologia , Masculino , Manejo de Espécimes/métodos , Recuperação Espermática , Espermatogênese , Terminologia como Assunto , Doenças Testiculares/patologia
9.
Reproduction ; 132(4): 601-5, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17008471

RESUMO

It has been proposed that follistatin can modulate the actions of activins and/or other members of the transforming growth factor-beta superfamily of proteins on testicular function, since mice overexpressing follistatin showed spermatogenic disruption. However, since mice with targeted disruption of the follistatin gene die soon after birth, it is not feasible to determine the effect of the absence of follistatin on testicular function using this model. To further understand the role of follistatin on the development and maintenance of spermatogenesis, fetal testes, collected by Caesarean section at day 18 of gestation from follistatin null mice, were transplanted to the external ear of castrated recombination activating gene 1 immunocompromised male mice. The testicular grafts were then analysed 7-8 weeks after transplantation and showed that full spermatogenesis developed in both the testes of wild-type and follistatin null mice. This study indicates that, if follistatin is required to modulate spermatogenic development, it is not supplied by local testicular production but by circulating follistatin from the host mouse.


Assuntos
Folistatina/metabolismo , Espermatogênese/fisiologia , Testículo/embriologia , Animais , Folistatina/genética , Hospedeiro Imunocomprometido , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Orquiectomia , Testículo/metabolismo , Testículo/transplante
10.
J Endocrinol ; 190(2): 331-40, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16899566

RESUMO

Production and regulation of activin A and inhibin B during the cycle of the seminiferous epithelium were investigated in adult rats. Immunohistochemistry localised the activin beta(A)-subunit to the Sertoli cell cytoplasm, with much weaker expression in spermatocytes and spermatids. Both activin A and inhibin B, measured by ELISA were secreted by, seminiferous tubule fragments over 72 h in culture. Activin A was secreted in a cyclic manner with peak secretion from tubules isolated at stage VIII. Tubules collected during stage VI produced the least activin A. Inhibin B secretion was highest from stage IX-I tubules and lowest from stage VII tubules. Addition of interleukin-1beta (IL-1beta) had relatively little effect on activin A or inhibin B secretion in culture. In contrast, the peak secretion of activin A by stage VIII tubules was blocked by co-incubation with an excess of human recombinant IL-1 receptor antagonist, whereas inhibin B secretion increased slightly. Dibutyryl cAMP stimulated activin A secretion by late stage VII and VIII tubules and stimulated inhibin B across all stages. These data indicate that activin A and inhibin B are cyclically regulated within the seminiferous epithelium, with endogenous IL-1 (presumably IL-1alpha produced by the Sertoli cells), responsible for a peak of activin A production subsequent to sperm release at stage VIII. These data provide direct evidence that production of activin A and inhibin B by the Sertoli cell is locally modulated by IL-1alpha , in addition to FSH/cAMP, under the influence of the developing spermatogenic cells.


Assuntos
Ativinas/biossíntese , Epitélio Seminífero/metabolismo , Espermatogênese/fisiologia , Ativinas/análise , Animais , Bucladesina/farmacologia , Citoplasma/química , Ensaio de Imunoadsorção Enzimática/métodos , Imuno-Histoquímica/métodos , Inibinas/análise , Inibinas/biossíntese , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/química , Células de Sertoli/química , Sialoglicoproteínas/farmacologia , Espermatozoides/química , Estimulação Química , Técnicas de Cultura de Tecidos
11.
Clin Exp Allergy ; 36(7): 941-50, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16839410

RESUMO

BACKGROUND: Activin A is a member of the transforming growth factor-beta superfamily which is directly implicated in airway structural change and inflammation in asthma. In vitro, the biological effects of activin A are neutralized by the soluble binding protein follistatin. OBJECTIVE: To determine the potential of endogenous follistatin to suppress activin A in vivo by analysing their relative tissue and kinetic compartmentalization during the effector phase of subchronic Th2-driven mucosal inflammation in a murine model of allergic asthma. METHODS: Eosinophilic mucosal inflammation was elicited by triggering Th2 recall responses by antigen challenge in ovalbumin-sensitized BALB/c mice. The kinetics and distribution of activin A and follistatin protein were assessed in lung tissue and bronchoalveolar lavage fluid and measured in relation to airway eosinophilia, goblet cell metaplasia and Th2 cytokine production in mediastinal lymph nodes. RESULTS: Follistatin was released concurrently with activin A suggesting it acts as an endogenous regulator: peak BAL concentrations coincided with maximal airway eosinophilia, and frequency of IL-4, IL-5 and IL-13 producing cells in mediastinal lymph nodes but induction lagged behind the onset of inflammation. Follistatin and activin A immunoreactivity were lost in airway epithelial cells in parallel with goblet cell metaplasia. Exogenous follistatin inhibited the allergen-specific Th2 immune response in mediastinal lymph nodes and mucus production in the lung. CONCLUSION: Follistatin is preformed in the normal lung and released in concert with activin A suggesting it serves as an endogenous regulator. Disturbance of the fine balance between activin A and its endogenous inhibitor follistatin may be a determinant of the severity of allergic inflammation or tissue phenotypic shift in asthma.


Assuntos
Ativinas/metabolismo , Asma/metabolismo , Folistatina/fisiologia , Animais , Asma/imunologia , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Feminino , Folistatina/metabolismo , Folistatina/farmacologia , Imunização , Interleucinas/biossíntese , Pulmão/metabolismo , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Muco/metabolismo , Ovalbumina/imunologia , Proteínas Recombinantes/farmacologia , Células Th2/imunologia
12.
J Endocrinol ; 187(1): 125-34, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16214948

RESUMO

The regulation of Sertoli cell activin A and inhibin B secretion during inflammation was investigated in vitro. Adult rat Sertoli cells were incubated with the inflammatory mediators, lipopolysaccharide (LPS), interleukin-1beta (IL-1beta), IL-6 and the IL-1 receptor antagonist (IL-1ra) over 48 h in culture. Activin A, inhibin B and IL-1alpha were measured in the culture medium by specific two-site ELISAs. Both IL-1beta- and LPS-stimulated activin A and inhibited inhibin B secretion. LPS also stimulated the production of IL-1alpha in the cultures. In contrast to IL-1beta, IL-6 had no effect on activin A, although it did have a significant inhibitory effect on inhibin B secretion. Ovine follicle-stimulating hormone (FSH) and the cAMP analogue dibutyryl cAMP opposed the actions of IL-1 and LPS by suppressing activin A and IL-1alpha secretion and by stimulating inhibin B. Blocking IL-1 activity in the cultures by addition of an excess of IL-1ra completely prevented the response of activin A to exogenous IL-1beta, and reduced the response to LPS by 50%. In the presence of IL-1ra, basal secretion of inhibin B was increased, but IL-1ra was unable to reverse the suppression of inhibin B by LPS. These data indicate the importance of both IL-1 isoforms in regulating secretion of activin A and inhibin B by mature Sertoli cells during inflammation. The data also establish that inflammation exerts its effects on activin A and inhibin B secretion via other pathways in addition to those mediated by IL-1, and that hormonal stimulation by FSH and cAMP moderates the Sertoli cell response to inflammation. Interference with the complex interactions between these cytokines and hormones may contribute to the disruption of reproductive function that can accompany infection and illness in men.


Assuntos
Ativinas/metabolismo , Mediadores da Inflamação/farmacologia , Subunidades beta de Inibinas/metabolismo , Inibinas/metabolismo , Células de Sertoli/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática/métodos , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/farmacologia , Interleucina-6/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/imunologia , Sialoglicoproteínas/farmacologia , Estimulação Química
13.
Hum Reprod ; 20(12): 3429-34, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16172145

RESUMO

BACKGROUND: With limited information regarding fertility and sexual activity in the older population, men's behaviour, attitudes and concerns were explored in a representative population of middle-aged and older men using the Men in Australia, Telephone Survey (MATeS). METHODS: A stratified random national sample of 5990 men participated in a standardized computer-assisted telephone interview. Equal numbers in the age strata 40-49, 50-59, 60-69 and >or=70 years were surveyed with findings census-standardized to the national population. Broad aspects of men's health and well-being, including reproductive health, were explored. RESULTS: The majority of men were sexually active in the last 12 months (age-standardized proportion, 78.3%) with approximately 37% of men aged >or=70 years still continuing sexual activity. Overall, 12.2% of men had never fathered children, of whom most (7.7%) had chosen not to have children. Questioning on failed attempts to produce a pregnancy suggested an involuntary infertility rate of 7.6%. The age-standardized vasectomy rate was 25.1%, with 5.6% of vasectomized men having no children. Although 9.2% of vasectomized men regretted sterilization, only 1.4% had undergone vasectomy reversal. CONCLUSIONS: Continuing sexual activity, fertility and contraception needs in middle-aged and older men suggests that education and service delivery must be more appropriately directed to an ageing population.


Assuntos
Anticoncepcionais/uso terapêutico , Fertilidade , Adulto , Idoso , Envelhecimento , Atitude , Austrália , Comportamento , Preservativos , Anticoncepção , Comportamento Contraceptivo , Serviços de Planejamento Familiar , Inquéritos Epidemiológicos , Humanos , Entrevistas como Assunto , Masculino , Pessoa de Meia-Idade , Comportamento Sexual , Parceiros Sexuais , Vasectomia
14.
Mol Hum Reprod ; 11(7): 507-12, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16123079

RESUMO

Men with Y chromosome (Yq) AZFc deletions lack all copies of the DAZ gene and have severe spermatogenic failure. A recently described gr/gr subdeletion of AZFc removes two of four copies of DAZ. To better understand the relative frequencies of AZFc and gr/gr deletions and their associated phenotypes, we analysed two large groups of infertile men. A total of 788 men from the Monash Male Infertility (MMI) database with a range of fertility disorders showed similar overall prevalences of AZFc (2.5%) and gr/gr deletions (3.4%). There was no association of gr/gr deletions with sperm density. In 234 control men of known or presumed fertility, only one gr/gr deletion was found. In a further 599 consecutive men presenting for assisted reproductive technologies, we detected 13 (2.2%) AZFc deletions and 28 (4.7%) gr/gr deletions. All AZFc deletions were seen with sperm densities <5 million/ml but again the gr/gr deletion occurred with similar frequency across all sperm density categories. These data show that gr/gr deletions are significantly associated with infertility in the Australian population (P = 0.0015) but not exclusively with reduced sperm density suggesting a complex interaction with other factors important for male fertility. Vertical transmission of gr/gr deletions from father to son by ICSI was demonstrated in four cases. Analysis of 130 ICSI-conceived sons revealed no de novo gr/gr deletions indicating that ICSI is not a risk factor. The data suggest that testing for gr/gr deletions should be considered in the routine genetic assessment of men with idiopathic infertility.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Y/genética , Infertilidade Masculina/genética , Haplótipos , Humanos , Infertilidade Masculina/epidemiologia , Masculino , Prevalência , Injeções de Esperma Intracitoplásmicas
15.
J Endocrinol ; 185(1): 99-110, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15817831

RESUMO

In several biological systems, the inhibin beta(A) homodimer activin A is stimulated by, and in turn, inhibits the action of interleukin (IL)-1 (both IL-1alpha and IL-1beta) and IL-6. The possibility that a similar regulatory relationship operates within the testis was investigated. Sertoli cells from immature (20-day-old) rats were cultured with human IL-1alpha or IL-1beta, human IL-6 and/or ovine FSH or dibutyryl cAMP. Activin A and the inhibin dimers, inhibin A and inhibin B, were measured by specific ELISA. Immunoreactive inhibin (ir-inhibin) was measured by RIA. Activin/inhibin subunit mRNA expression was measured by quantitative real-time PCR. Both IL-1 isoforms, but not IL-6, stimulated activin A secretion through increased synthesis of beta(A)-subunit mRNA. IL-1 also stimulated activin A secretion by testicular peritubular cells. In contrast to the effect on activin A, IL-1 suppressed inhibin beta(B)-subunit and, to a lesser extent, alpha-subunit mRNA expression, thereby reducing basal and FSH-stimulated inhibin B secretion by the Sertoli cells. Conversely, FSH inhibited basal activin A secretion and antagonised the stimulatory effects of IL-1. Dibutyryl cAMP partially inhibited the action of IL-1 on activin A secretion, but had no significant effect on basal activin A secretion. Secretion of inhibin A was low in all treatment groups. These data demonstrate that IL-1 and FSH/cAMP exert a reciprocal regulation of activin A and inhibin B synthesis and release by the Sertoli cell, and suggest a role for activin A as a potential feedback regulator of IL-1 and IL-6 activity in the testis during normal spermatogenesis and in inflammation.


Assuntos
Ativinas/farmacologia , Hormônio Foliculoestimulante/farmacologia , Subunidades beta de Inibinas/farmacologia , Inibinas/farmacologia , Interleucina-1/farmacologia , Células de Sertoli/metabolismo , Animais , Bucladesina/farmacologia , Técnicas de Cultura de Células , Ensaio de Imunoadsorção Enzimática/métodos , Retroalimentação Fisiológica , Interleucina-6/farmacologia , Masculino , RNA Mensageiro/análise , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/efeitos dos fármacos , Estimulação Química
16.
Mol Hum Reprod ; 11(12): 871-80, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16421219

RESUMO

Many of the proteins and their encoding genes involved in spermatogenesis are unknown, making the specific diagnosis and treatment of infertility in males difficult and highlighting the importance of identifying new genes that are involved in spermatogenesis. Through genome-wide chemical mutagenesis using N-ethyl-N-nitrosourea (ENU) and a three-generation breeding scheme to isolate recessive mutations, we have identified mouse lines with a range of abnormalities relevant to human male fertility. Abnormal phenotypes included hypospermatogenesis, Sertoli cell-only (SCO) seminiferous tubules, germ-cell arrest and abnormal spermiogenesis and were accompanied, in some, with abnormal serum levels of reproductive hormones. In total, from 65 mouse lines, 14 showed a reproductive phenotype consistent with a recessive mutation. This study shows that it is feasible to use ENU mutagenesis as an effective and rapid means of generating mouse models relevant to furthering our understanding of human male infertility. Spermatozoa and genomic DNA from all mouse lines, including those with abnormal reproductive tract parameters, have been cryopreserved for the regeneration of lines as required. This repository will form a valuable resource for the identification and analysis of key regulators of multiple aspects of male fertility.


Assuntos
Etilnitrosoureia/toxicidade , Fertilidade/fisiologia , Ativinas , Animais , Apoptose , Cruzamentos Genéticos , Feminino , Fertilidade/efeitos dos fármacos , Fertilidade/genética , Hormônio Foliculoestimulante/sangue , Masculino , Camundongos , Camundongos Mutantes , Mutagênese , Mutagênicos , Tamanho do Órgão , Preservação do Sêmen , Testículo/anatomia & histologia , Testículo/efeitos dos fármacos
17.
Mol Cell Endocrinol ; 225(1-2): 57-64, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15451568

RESUMO

The role of the inhibins, activins and follistatins in testicular function are being more clearly defined following studies describing the cellular localisation of these proteins to the testis and the availability of specific assay systems enabling measurement of these proteins. Taken together with the results of targetted gene inactivation experiments, several concepts emerge. Inhibin B is predominantly produced by the Sertoli cell in many adult male mammals whereas there is a perinatal peak of inhibin A in the rat. In contrast, activin A has its highest concentrations in the immediate post-natal period during which it is involved in the developmental regulation of both germ cells and Sertoli cells being modulated by follistatin. Activin A levels are considerably lower in the adult testis but Sertoli cell production is stimulated by interleukin-1 and inhibited by FSH. Little is known about the production of activin B due to the absence of a suitable assay but the beta(B) subunit mRNA is expressed in germ cells and Sertoli cells and is stage-dependent. This pattern of expression suggest that it may be involved in autocrine or paracrine actions within the seminiferous epithelium.


Assuntos
Hormônios Gonadais/fisiologia , Testículo/fisiologia , Ativinas/genética , Ativinas/metabolismo , Ativinas/fisiologia , Animais , Folistatina/genética , Folistatina/metabolismo , Folistatina/fisiologia , Regulação da Expressão Gênica/fisiologia , Hormônios Gonadais/genética , Humanos , Inibinas/fisiologia , Masculino
19.
J Endocrinol ; 182(1): 69-80, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15225132

RESUMO

A series of experiments were conducted in adult ewes to delineate the release profile of activin A and its relationship to other cytokines following an i.v. injection of the bacterial cell wall component, lipopolysaccharide (LPS). Following this challenge, plasma activin A increased rapidly and appeared to be released in a biphasic manner, slightly preceding the release of tumour necrosis factor-alpha (TNFalpha) and before elevation of interleukin (IL)-6 and follistatin levels. The concentration of activin A was correlated with body temperature during the response to LPS. A second experiment compared cytokine concentrations in matched blood and cerebrospinal fluid (CSF) samples. This revealed that activin A was not released centrally in the CSF following a peripheral LPS injection, nor was TNFalpha or the activin binding protein, follistatin, but IL-6 showed a robust elevation. In a third experiment, the stimulus for activin A release was examined by blocking prostaglandin synthesis. Flurbiprofen, a prostaglandin synthesis inhibitor, effectively attenuated the fever response to LPS and partly inhibited cortisol release, but the cytokine profiles were unaffected. Finally, the bioactivity of TNFalpha and/or IL-1 was blocked using soluble receptor antagonists. These treatments did not affect the initial release of activin A, but blockade of TNFalpha depressed the second activin peak. These studies define more rigorously the release of activin A into the circulation following acute inflammatory challenge. The response is rapid and probably biphasic, independent of prostaglandin- mediated pathways and does not depend upon stimulation by TNFalpha or IL-1. The data suggest that activin A release is an early event in the inflammatory cascade following the interaction of LPS with its cellular receptor.


Assuntos
Ativinas/metabolismo , Infecções Bacterianas/imunologia , Subunidades beta de Inibinas/metabolismo , Doenças dos Ovinos/imunologia , Ativinas/sangue , Animais , Infecções Bacterianas/sangue , Inibidores de Ciclo-Oxigenase/farmacologia , Feminino , Flurbiprofeno/farmacologia , Folistatina/sangue , Subunidades beta de Inibinas/sangue , Injeções Intravenosas , Interleucina-6/sangue , Lipopolissacarídeos/farmacologia , Ovinos , Doenças dos Ovinos/sangue , Fatores de Tempo , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...