Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Andrology ; 9(5): 1652-1661, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33998165

RESUMO

BACKGROUND: Circulating prolactin concentration in rodents and humans is sexually dimorphic. Oestrogens are a well-characterised stimulator of prolactin release. Circulating prolactin fluctuates throughout the menstrual/oestrous cycle of females in response to oestrogen levels, but remains continually low in males. We have previously identified androgens as an inhibitor of prolactin release through characterisation of males of a mouse line with a conditional pituitary androgen receptor knockout (PARKO) which have an increase in circulating prolactin, but unchanged lactotroph number. OBJECTIVES: In the present study, we aimed to specify the cell type that androgens act on to repress prolactin release. MATERIALS AND METHODS: PARKO, lactotroph-specific, Pit1 lineage-specific and neural-specific conditional androgen receptor knockout male mice were investigated using prolactin ELISA, pituitary electron microscopy, immunohistochemistry and qRT-PCR. RESULTS: Lactotroph-specific, Pit1 lineage-specific and neural-specific conditional AR knockouts did not duplicate the high circulating prolactin seen in the PARKO line. Using electron microscopy to examine ultrastructure, we showed that pituitary androgen receptor knockout male mice develop lactotrophs that resemble those seen in female mice. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males. When expression of selected oestrogen-regulated anterior pituitary genes was examined, there were no differences in expression level between controls and knockouts. DISCUSSION: The cell type that androgens act on to repress prolactin release is not the lactotroph, cells in the Pit1-lineage, or the dopaminergic neurons in the hypothalamus. PARKO males develop a female-specific lactotroph ultrastructure that this is likely to contribute to the increase in circulating prolactin. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males, which suggests that removal of both circulating oestrogens and androgens reduces the stimulation of pituitary prolactin release. CONCLUSION: Further investigation is needed into prolactin regulation by changes in androgen-oestrogen balance, which is involved sexual dimorphism of development and diseases including hyperprolactinemia.


Assuntos
Hiperprolactinemia/genética , Lactotrofos , Receptores Androgênicos/deficiência , Animais , Estrogênios/metabolismo , Masculino , Camundongos , Camundongos Knockout , Hipófise/metabolismo , Prolactina/metabolismo
2.
Reproduction ; 161(5): 593-602, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33784244

RESUMO

The epididymis is an androgen-responsive organ, whose structure and functions are modulated by the coordination between androgen and epididymal cues. Highly regulated molecular interaction within the epididymis is required to support viable sperm development necessary for subsequent fertilization. In the present study, we extended our earlier findings on a promising epididymal protein, quiescin sulfhydryl oxidase 2 (QSOX2), and demonstrated a positive correlation between testosterone and QSOX2 protein synthesis through the use of loss- and restore-of-function animal models. Moreover, based on transcriptomic analyses and 2D culture system, we determined that an additional polarized effect of glutamate is indispensable for the regulatory action of testosterone on QSOX2 synthesis. In conclusion, we propose noncanonical testosterone signaling supports epididymal QSOX2 protein synthesis, providing a novel perspective on the regulation of sperm maturation within the epididymis.


Assuntos
Epididimo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Maturação do Esperma , Testosterona/farmacologia , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Proteínas de Transporte/metabolismo , Epididimo/citologia , Epididimo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética
3.
J Neuroendocrinol ; 32(10): e12903, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32959418

RESUMO

The anterior and intermediate lobes of the pituitary are composed of endocrine cells, as well as vasculature and supporting cells, such as folliculostellate cells. Folliculostellate cells form a network with several postulated roles in the pituitary, including production of paracrine signalling molecules and cytokines, coordination of endocrine cell hormone release, phagocytosis, and structural support. Folliculostellate cells in rats are characterised by expression of S100B protein, and in humans by glial fibrillary acid protein. However, there is evidence for another network of supporting cells in the anterior pituitary that has properties of mural cells, such as vascular smooth muscle cells and pericytes. The present study aims to characterise the distribution of cells that express the mural cell marker platelet derived growth factor receptor beta (PDGFRß) in the mouse pituitary and establish whether these cells are folliculostellate. By immunohistochemical localisation, we determine that approximately 80% of PDGFRß+ cells in the mouse pituitary have a non-perivascular location and 20% are pericytes. Investigation of gene expression in a magnetic cell sorted population of PDGFRß+ cells shows that, despite a mostly non-perivascular location, this population is enriched for mural cell markers but not enriched for rat or human folliculostellate cell markers. This is confirmed by immunohistochemistry. The present study concludes that a mural cell network is present throughout the anterior pituitary of the mouse and that this population does not express well-characterised human or rat folliculostellate cell markers.


Assuntos
Comunicação Celular/fisiologia , Hipófise/citologia , Animais , Biomarcadores/metabolismo , Células Endócrinas/citologia , Células Endócrinas/fisiologia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Pericitos/citologia , Pericitos/fisiologia , Hipófise/metabolismo , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Transcrição SOXB1/metabolismo
4.
Front Endocrinol (Lausanne) ; 11: 599869, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33584538

RESUMO

Adrenal androgens are fundamental mediators of ovarian folliculogenesis, embryonic implantation, and breast development. Although adrenal androgen function in target tissues are well characterized, there is little research covering the role of androgen-signaling within the adrenal itself. Adrenal glands express AR which is essential for the regression of the X-zone in male mice. Female mice also undergo X-zone regression during their first pregnancy, however whether this is also controlled by AR signaling is unknown. To understand the role of the androgen receptor (AR) in the female adrenal, we utilized a Cyp11a1-Cre to specifically ablate AR from the mouse adrenal cortex. Results show that AR-signaling is dispensable for adrenal gland development in females, and for X-zone regression during pregnancy, but is required to suppress elevation of corticosterone levels post-partum. Additionally, following disruption to adrenal AR, aberrant spindle cell development is observed in young adult females. These results demonstrate sexually dimorphic regulation of the adrenal X-zone by AR and point to dysfunctional adrenal androgen signaling as a possible mechanism in the early development of adrenal spindle cell hyperplasia.


Assuntos
Córtex Suprarrenal/citologia , Androgênios/farmacologia , Corticosterona/metabolismo , Período Pós-Parto/metabolismo , Substâncias Protetoras/farmacologia , Receptores Androgênicos/química , Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/metabolismo , Animais , Feminino , Masculino , Camundongos , Período Pós-Parto/efeitos dos fármacos , Gravidez , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transdução de Sinais
5.
Sci Rep ; 9(1): 10457, 2019 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-31320667

RESUMO

Androgens are known to be an essential regulator of male health. Androgen receptor (AR) is widely expressed throughout the adrenal cortex, yet the wider role for androgen signalling in the adrenal remains underexplored. To investigate AR-dependent and AR-independent androgen signalling in the adrenal, we used a novel mouse model with a specific ablation of androgen receptor in the adrenal cortex with or without reduction of circulating androgen levels by castration. Our results describe AR expression in the human and mouse adrenal and highlight that the mouse is a viable model to investigate androgen signalling in the adrenal cortex. We show androgen signalling via AR is required for X-zone regression during puberty. Furthermore, cortex measurements define differences in X-zone morphology depending on whether circulating androgens or AR have been removed. We show androgens promote both cortical cell differentiation and apoptosis but are dispensable for the formation of the definitive cortex. Additionally, investigation of aged mice with AR ablation reveals severe cortex disruption, spindle cell hyperplasia and X-zone expansion. The data described herein demonstrates AR-signalling is required to facilitate X-zone regression, cell clearance and to protect against adrenal degeneration during ageing.


Assuntos
Córtex Suprarrenal/citologia , Envelhecimento/fisiologia , Androgênios/farmacologia , Substâncias Protetoras/farmacologia , Receptores Androgênicos/fisiologia , Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Apoptose , Castração , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
6.
BMC Dev Biol ; 19(1): 8, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30995907

RESUMO

BACKGROUND: The interstitium of the mouse testis contains Leydig cells and a small number of steroidogenic cells with adrenal characteristics which may be derived from the fetal adrenal during development or may be a normal subset of the developing fetal Leydig cells. Currently it is not known what regulates development and/or proliferation of this sub-population of steroidogenic cells in the mouse testis. Androgen receptors (AR) are essential for normal testicular function and in this study we have examined the role of the AR in regulating interstitial cell development. RESULTS: Using a mouse model which lacks gonadotropins and AR (hpg.ARKO), stimulation of luteinising hormone receptors in vivo with human chorionic gonadotropin (hCG) caused a marked increase in adrenal cell transcripts/protein in a group of testicular interstitial cells. hCG also induced testicular transcripts associated with basic steroidogenic function in these mice but had no effect on adult Leydig cell-specific transcript levels. In hpg mice with functional AR, treatment with hCG induced Leydig cell-specific function and had no effect on adrenal transcript levels. Examination of mice with cell-specific AR deletion and knockdown of AR in a mouse Leydig cell line suggests that AR in the Leydig cells are likely to regulate these effects. CONCLUSIONS: This study shows that in the mouse the androgen receptor is required both to prevent development of testicular cells with adrenal characteristics and to ensure development of an adult Leydig cell phenotype.


Assuntos
Gonadotropina Coriônica/metabolismo , Desenvolvimento Embrionário/fisiologia , Células Intersticiais do Testículo/citologia , Hormônio Luteinizante/metabolismo , Receptores Androgênicos/biossíntese , Animais , Contagem de Células , Linhagem Celular Tumoral , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Modelos Animais , Fenótipo , Receptores Androgênicos/genética
7.
Sci Rep ; 9(1): 3250, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824750

RESUMO

The hypothalamic-pituitary-adrenal (HPA) axis regulates responses to internal and external stressors. Many patients diagnosed with conditions such as depression or anxiety also have hyperactivity of the HPA axis. Hyper-stimulation of the HPA axis results in sustained elevated levels of glucocorticoids which impair neuronal function and can ultimately result in a psychiatric disorder. Studies investigating Glucocorticoid Receptor (GR/NR3C1) in the brain have primarily focused on the forebrain, however in recent years, the hindbrain has become a region of interest for research into the development of anxiety and depression, though the role of GR signalling in the hindbrain remains poorly characterised. To determine the role of glucocorticoid signalling in the hindbrain we have developed a novel mouse model that specifically ablates hindbrain GR to ascertain its role in behaviour, HPA-axis regulation and adrenal structure. Our study highlights that ablation of GR in the hindbrain results in excessive barbering, obsessive compulsive digging and lack of cage exploration. These mice also develop kyphosis, elevated circulating corticosterone and severe adrenal cortex disruption. Together, this data demonstrates a role for hindbrain GR signalling in regulating stress-related behaviour and identifies a novel mouse model to allow further investigation into the pathways impacting stress and anxiety.


Assuntos
Receptores de Glucocorticoides/metabolismo , Rombencéfalo/metabolismo , Estresse Psicológico/metabolismo , Córtex Suprarrenal/patologia , Animais , Comportamento Animal , Peso Corporal , Modelos Animais de Doenças , Feminino , Cifose/complicações , Cifose/diagnóstico por imagem , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tomografia por Emissão de Pósitrons , Recombinação Genética/genética , Rombencéfalo/diagnóstico por imagem , Estresse Psicológico/diagnóstico por imagem , Tomografia Computadorizada por Raios X
8.
BMC Res Notes ; 11(1): 252, 2018 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-29690918

RESUMO

OBJECTIVE: Steroid hormones are responsible for the control of a wide range of physiological processes such as development, growth, reproduction, metabolism, and aging. Because of the variety of enzymes, substrates and products that take part in steroidogenesis and the compartmentalisation of its constituent reactions, it is a complex process to visualise and document. One of the goals of systems biology is to quantitatively describe the behaviour of complex biological systems that involve the interaction of many components. This can be done by representing these interactions visually in a pathway model and then optionally constructing a mathematical model of the interactions. RESULTS: We have used the modified Edinburgh Pathway Notation to construct a framework diagram describing human steroidogenic pathways, which will be of use to endocrinologists. To demonstrate further utility, we show how such models can be parameterised with empirical data within the software Graphia Professional, to recapitulate specific examples of steroid hormone production, and also to mimic gene knockout. These framework models support in silico hypothesis generation and testing with utility across endocrine endpoints, with significant potential to reduce costs, time and animal numbers, whilst informing the design of planned studies.


Assuntos
Hormônios Esteroides Gonadais/biossíntese , Modelos Biológicos , Esteroides/biossíntese , Humanos
9.
Nat Protoc ; 13(4): 705-722, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29543794

RESUMO

A major endeavor of systems biology is the construction of graphical and computational models of biological pathways as a means to better understand their structure and function. Here, we present a protocol for a biologist-friendly graphical modeling scheme that facilitates the construction of detailed network diagrams, summarizing the components of a biological pathway (such as proteins and biochemicals) and illustrating how they interact. These diagrams can then be used to simulate activity flow through a pathway, thereby modeling its dynamic behavior. The protocol is divided into four sections: (i) assembly of network diagrams using the modified Edinburgh Pathway Notation (mEPN) scheme and yEd network editing software with pathway information obtained from published literature and databases of molecular interaction data; (ii) parameterization of the pathway model within yEd through the placement of 'tokens' on the basis of the known or imputed amount or activity of a component; (iii) model testing through visualization and quantitative analysis of the movement of tokens through the pathway, using the network analysis tool Graphia Professional and (iv) optimization of model parameterization and experimentation. This is the first modeling approach that combines a sophisticated notation scheme for depicting biological events at the molecular level with a Petri net-based flow simulation algorithm and a powerful visualization engine with which to observe the dynamics of the system being modeled. Unlike many mathematical approaches to modeling pathways, it does not require the construction of a series of equations or rate constants for model parameterization. Depending on a model's complexity and the availability of information, its construction can take days to months, and, with refinement, possibly years. However, once assembled and parameterized, a simulation run, even on a large model, typically takes only seconds. Models constructed using this approach provide a means of knowledge management, information exchange and, through the computation simulation of their dynamic activity, generation and testing of hypotheses, as well as prediction of a system's behavior when perturbed.


Assuntos
Biologia Computacional/métodos , Gráficos por Computador , Simulação por Computador , Biologia de Sistemas/métodos , Algoritmos , Redes e Vias Metabólicas , Modelos Biológicos , Mapas de Interação de Proteínas , Software
10.
Sci Rep ; 7(1): 8991, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28827578

RESUMO

The tamoxifen-inducible Cre system is a popular transgenic method for controlling the induction of recombination by Cre at a specific time and in a specific cell type. However, tamoxifen is not an inert inducer of recombination, but an established endocrine disruptor with mixed agonist/antagonist activity acting via endogenous estrogen receptors. Such potentially confounding effects should be controlled for, but >40% of publications that have used tamoxifen to generate conditional knockouts have not reported even the minimum appropriate controls. To highlight the importance of this issue, the present study investigated the long-term impacts of different doses of a single systemic tamoxifen injection on the testis and the wider endocrine system. We found that a single dose of tamoxifen less than 10% of the mean dose used for recombination induction, caused adverse effects to the testis and to the reproductive endocrine system that persisted long-term. These data raise significant concerns about the widespread use of tamoxifen induction of recombination, and highlight the importance of including appropriate controls in all pathophysiological studies using this means of induction.


Assuntos
Antagonistas de Estrogênios/administração & dosagem , Antagonistas de Estrogênios/efeitos adversos , Efeitos Adversos de Longa Duração , Tamoxifeno/administração & dosagem , Tamoxifeno/efeitos adversos , Testículo/efeitos dos fármacos , Administração Intravenosa , Animais , Histocitoquímica , Imuno-Histoquímica , Masculino , Camundongos Endogâmicos C57BL , Testículo/patologia
11.
PLoS Biol ; 14(8): e1002530, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27509052

RESUMO

There is a need for formalised diagrams that both summarise current biological pathway knowledge and support modelling approaches that explain and predict their behaviour. Here, we present a new, freely available modelling framework that includes a biologist-friendly pathway modelling language (mEPN), a simple but sophisticated method to support model parameterisation using available biological information; a stochastic flow algorithm that simulates the dynamics of pathway activity; and a 3-D visualisation engine that aids understanding of the complexities of a system's dynamics. We present example pathway models that illustrate of the power of approach to depict a diverse range of systems.


Assuntos
Algoritmos , Biologia Computacional/métodos , Modelos Biológicos , Transdução de Sinais , Animais , Simulação por Computador , Humanos , Reprodutibilidade dos Testes
12.
Methods Mol Biol ; 1443: 219-48, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27246343

RESUMO

Conditional gene targeting has revolutionized molecular genetic analysis of nuclear receptor proteins, however development and analysis of such conditional knockouts is far from simple, with many caveats and pitfalls waiting to snare the novice or unprepared. In this chapter, we describe our experience of generating and analyzing mouse models with conditional ablation of the androgen receptor (AR) from tissues of the reproductive system and other organs. The guidance, suggestions, and protocols outlined in the chapter provide the key starting point for analyses of conditional-ARKO mice, completing them as described provides an excellent framework for further focussed project-specific analyses, and applies equally well to analysis of reproductive tissues from any mouse model generated through conditional gene targeting.


Assuntos
Marcação de Genes/métodos , Técnicas Imunoenzimáticas/métodos , Microscopia de Fluorescência/métodos , Reação em Cadeia da Polimerase em Tempo Real/métodos , Receptores Androgênicos/fisiologia , Animais , Genes Reporter , Genótipo , Hormônios/metabolismo , Integrases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Receptores Androgênicos/genética , Testículo/metabolismo , Transgenes
13.
Best Pract Res Clin Endocrinol Metab ; 29(4): 595-605, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26303086

RESUMO

Androgens such as testosterone are steroid hormones essential for normal male reproductive development and function. Mutations of androgen receptors (AR) are often found in patients with disorders of male reproductive development, and milder mutations may be responsible for some cases of male infertility. Androgens exert their action through AR and its signalling in the testis is essential for spermatogenesis. AR is not expressed in the developing germ cell lineage so is thought to exert its effects through testicular Sertoli and peri-tubular myoid (PTM) cells. AR signalling in spermatogenesis has been investigated in rodent models where testosterone levels are chemically supressed or models with transgenic disruption of AR. These models have pinpointed the steps of spermatogenesis that require AR signalling, specifically maintenance of spermatogonial numbers, blood-testis barrier integrity, completion of meiosis, adhesion of spermatids and spermiation, together these studies detail the essential nature of androgens in the promotion of male fertility.


Assuntos
Infertilidade Masculina/metabolismo , Receptores Androgênicos/metabolismo , Espermatogênese , Animais , Humanos , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Masculino , Mutação , Receptores Androgênicos/genética
14.
PLoS One ; 10(3): e0121657, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25799562

RESUMO

Production of the androgen testosterone is controlled by a negative feedback loop within the hypothalamic-pituitary-gonadal (HPG) axis. Stimulation of testicular Leydig cells by pituitary luteinising hormone (LH) is under the control of hypothalamic gonadotrophin releasing hormone (GnRH), while suppression of LH secretion by the pituitary is controlled by circulating testosterone. Exactly how androgens exert their feedback control of gonadotrophin secretion (and whether this is at the level of the pituitary), as well as the role of AR in other pituitary cell types remains unclear. To investigate these questions, we exploited a transgenic mouse line (Foxg1 Cre/+; AR fl/y) which lacks androgen receptor in the pituitary gland. Both circulating testosterone and gonadotrophins are unchanged in adulthood, demonstrating that AR signalling is dispensable in the male mouse pituitary for testosterone-dependent regulation of LH secretion. In contrast, Foxg1 Cre/+; AR fl/y males have a significant increase in circulating prolactin, suggesting that, rather than controlling gonadotrophins, AR-signalling in the pituitary acts to suppress aberrant prolactin production in males.


Assuntos
Hipófise/citologia , Prolactina/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Animais , Regulação da Expressão Gênica , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Receptores Androgênicos/genética
15.
Am J Reprod Immunol ; 74(1): 12-25, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25598450

RESUMO

PROBLEM: Previous studies demonstrated a strong association between low androgen levels and reduced capacity to mount an inflammatory response. However, the mechanisms underlying these observations are largely not understood. METHODS OF STUDY: Generation of CD4+CD25+Foxp3+ regulatory T cells in Leydig cell-conditioned media was determined by flow cytometry and ELISA. Influence of testosterone on cytokine response was measured in LPS-stimulated testicular macrophages, Sertoli and peritubular cells. RESULTS: Leydig cell-conditioned media dose-dependently stimulated expression of transcription factor Foxp3 and secretion of IL-10 in splenic CD4+ T cells, an effect abolished by addition of the anti-androgen flutamide. In isolated Sertoli and peritubular cells, testosterone pre-treatment suppressed the LPS-induced inflammatory response on TNF-α mRNA expression, while no effect was evident in testicular macrophages (TM). CONCLUSIONS: Androgens can influence the immune system under normal conditions by the generation and functional differentiation of regulatory T cells and in testicular inflammation by direct effect on Sertoli and peritubular cells.


Assuntos
Fatores de Transcrição Forkhead/biossíntese , Interleucina-10/biossíntese , Células Intersticiais do Testículo/imunologia , Linfócitos T Reguladores/citologia , Testosterona/metabolismo , Antagonistas de Androgênios/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Cultivadas , Quimiocina CCL2/biossíntese , Meios de Cultivo Condicionados/farmacologia , Flutamida/farmacologia , Inflamação/imunologia , Interleucina-10/metabolismo , Macrófagos/imunologia , Masculino , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Células de Sertoli/imunologia , Linfócitos T Reguladores/imunologia , Testosterona/antagonistas & inibidores , Fator de Crescimento Transformador beta/biossíntese , Fator de Necrose Tumoral alfa/genética
16.
FASEB J ; 29(3): 894-910, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25404712

RESUMO

Leydig cell number and function decline as men age, and low testosterone is associated with all "Western" cardio-metabolic disorders. However, whether perturbed androgen action within the adult Leydig cell lineage predisposes individuals to this late-onset degeneration remains unknown. To address this, we generated a novel mouse model in which androgen receptor (AR) is ablated from ∼75% of adult Leydig stem cell/cell progenitors, from fetal life onward (Leydig cell AR knockout mice), permitting interrogation of the specific roles of autocrine Leydig cell AR signaling through comparison to adjacent AR-retaining Leydig cells, testes from littermate controls, and to human testes, including from patients with complete androgen insensitivity syndrome (CAIS). This revealed that autocrine AR signaling is dispensable for the attainment of final Leydig cell number but is essential for Leydig cell maturation and regulation of steroidogenic enzymes in adulthood. Furthermore, these studies reveal that autocrine AR signaling in Leydig cells protects against late-onset degeneration of the seminiferous epithelium in mice and inhibits Leydig cell apoptosis in both adult mice and patients with CAIS, possibly via opposing aberrant estrogen signaling. We conclude that autocrine androgen action within Leydig cells is essential for the lifelong support of spermatogenesis and the development and lifelong health of Leydig cells.


Assuntos
Síndrome de Resistência a Andrógenos/patologia , Androgênios/farmacologia , Apoptose/efeitos dos fármacos , Células Intersticiais do Testículo/patologia , Receptores Androgênicos/fisiologia , Testículo/patologia , Adolescente , Adulto , Síndrome de Resistência a Andrógenos/tratamento farmacológico , Síndrome de Resistência a Andrógenos/metabolismo , Animais , Comunicação Autócrina , Western Blotting , Células Cultivadas , Criança , Humanos , Técnicas Imunoenzimáticas , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Substâncias Protetoras/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/metabolismo , Adulto Jovem
17.
Development ; 141(10): 2139-49, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24803659

RESUMO

Sertoli cells (SCs) regulate testicular fate in the differentiating gonad and are the main regulators of spermatogenesis in the adult testis; however, their role during the intervening period of testis development, in particular during adult Leydig cell (ALC) differentiation and function, remains largely unknown. To examine SC function during fetal and prepubertal development we generated two transgenic mouse models that permit controlled, cell-specific ablation of SCs in pre- and postnatal life. Results show that SCs are required: (1) to maintain the differentiated phenotype of peritubular myoid cells (PTMCs) in prepubertal life; (2) to maintain the ALC progenitor population in the postnatal testis; and (3) for development of normal ALC numbers. Furthermore, our data show that fetal LCs function independently from SC, germ cell or PTMC support in the prepubertal testis. Together, these findings reveal that SCs remain essential regulators of testis development long after the period of sex determination. These findings have significant implications for our understanding of male reproductive disorders and wider androgen-related conditions affecting male health.


Assuntos
Diferenciação Celular , Células Intersticiais do Testículo/fisiologia , Epitélio Seminífero/citologia , Células de Sertoli/fisiologia , Maturidade Sexual/fisiologia , Testículo/citologia , Testículo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Masculino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Túbulos Seminíferos/citologia
18.
Proc Natl Acad Sci U S A ; 111(18): E1924-32, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753613

RESUMO

Fetal growth plays a role in programming of adult cardiometabolic disorders, which in men, are associated with lowered testosterone levels. Fetal growth and fetal androgen exposure can also predetermine testosterone levels in men, although how is unknown, because the adult Leydig cells (ALCs) that produce testosterone do not differentiate until puberty. To explain this conundrum, we hypothesized that stem cells for ALCs must be present in the fetal testis and might be susceptible to programming by fetal androgen exposure during masculinization. To address this hypothesis, we used ALC ablation/regeneration to identify that, in rats, ALCs derive from stem/progenitor cells that express chicken ovalbumin upstream promoter transcription factor II. These stem cells are abundant in the fetal testis of humans and rodents, and lineage tracing in mice shows that they develop into ALCs. The stem cells also express androgen receptors (ARs). Reduction in fetal androgen action through AR KO in mice or dibutyl phthalate (DBP) -induced reduction in intratesticular testosterone in rats reduced ALC stem cell number by ∼40% at birth to adulthood and induced compensated ALC failure (low/normal testosterone and elevated luteinizing hormone). In DBP-exposed males, this failure was probably explained by reduced testicular steroidogenic acute regulatory protein expression, which is associated with increased histone methylation (H3K27me3) in the proximal promoter. Accordingly, ALCs and ALC stem cells immunoexpressed increased H3K27me3, a change that was also evident in ALC stem cells in fetal testes. These studies highlight how a key component of male reproductive development can fundamentally reprogram adult hormone production (through an epigenetic change), which might affect lifetime disease risk.


Assuntos
Células-Tronco Adultas/fisiologia , Androgênios/fisiologia , Desenvolvimento Fetal/fisiologia , Células Intersticiais do Testículo/fisiologia , Células-Tronco Adultas/efeitos dos fármacos , Animais , Callithrix , Linhagem da Célula/fisiologia , Dibutilftalato/toxicidade , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Células-Tronco Fetais/efeitos dos fármacos , Células-Tronco Fetais/fisiologia , Humanos , Técnicas In Vitro , Células Intersticiais do Testículo/efeitos dos fármacos , Hormônio Luteinizante/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Gravidez , Ratos , Ratos Transgênicos , Ratos Wistar , Receptores Androgênicos/deficiência , Receptores Androgênicos/genética , Receptores Androgênicos/fisiologia , Regeneração , Testículo/embriologia , Testículo/fisiologia , Testosterona/deficiência , Testosterona/fisiologia
19.
PLoS One ; 9(1): e84541, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24404170

RESUMO

To permit conditional gene targeting of floxed alleles in steroidogenic cell-types we have generated a transgenic mouse line that expresses Cre Recombinase under the regulation of the endogenous Cytochrome P450 side chain cleavage enzyme (Cyp11a1) promoter. Mice Carrying the Cyp11a1-GC (GFP-Cre) allele express Cre Recombinase in fetal adrenal and testis, and adrenal cortex, testicular Leydig cells (and a small proportion of Sertoli cells), theca cells of the ovary, and the hindbrain in postnatal life. Circulating testosterone concentration is unchanged in Cyp11(+/GC) males, suggesting steroidogenesis is unaffected by loss of one allele of Cyp11a1, mice are grossly normal, and Cre Recombinase functions to recombine floxed alleles of both a YFP reporter gene and the Androgen Receptor (AR) in steroidogenic cells of the testis, ovary, adrenal and hindbrain. Additionally, when bred to homozygosity (Cyp11a1(GC/GC) ), knock-in of GFP-Cre to the endogenous Cyp11a1 locus results in a novel mouse model lacking endogenous Cyp11a1 (P450-SCC) function. This unique dual-purpose model has utility both for those wishing to conditionally target genes within steroidogenic cell types and for studies requiring mice lacking endogenous steroid hormone production.


Assuntos
Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Expressão Gênica , Marcação de Genes , Proteínas de Fluorescência Verde/genética , Integrases/genética , Ovário/metabolismo , Testículo/metabolismo , Animais , Feminino , Ordem dos Genes , Masculino , Camundongos , Camundongos Knockout , Ovário/citologia , Fenótipo , Testículo/citologia
20.
BMC Res Notes ; 5: 16, 2012 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-22230795

RESUMO

BACKGROUND: Androgen signalling is essential both for male development and function of the male reproductive system in adulthood. Within the adult testis, Germ cells (GC) do not express androgen receptor (AR) suggesting androgen-mediated promotion of spermatogenesis must act via AR-expressing somatic cell-types. Several recent studies have exploited the Cre/lox system of conditional gene-targeting to ablate AR function from key somatic cell-types in order to establish the cell-specific role of AR in promotion of male fertility. In this study, we have used a similar approach to specifically ablate AR-signalling from Vascular Endothelial (VE) cells, with a view to defining the significance of androgen signalling within this cell-type on spermatogenesis. FINDINGS: AR expression in VE cells of the testicular vasculature was confirmed using an antibody against AR. A Cre-inducible fluorescent reporter line was used to empirically establish the utility of a mouse line expressing Cre Recombinase driven by the Tie2-Promoter, for targeting VE cells. Immunofluorescent detection revealed expression of YFP (and therefore Cre Recombinase function) limited to VE cells and an interstitial population of cells, believed to be macrophages, that did not express AR. Mating of Tie2-Cre males to females carrying a floxed AR gene produced Vascular Endothelial Androgen Receptor Knockout (VEARKO) mice and littermate controls. Ablation of AR from all VE cells was confirmed; however, no significant differences in bodyweight or reproductive tissue weights could be detected in VEARKO animals and spermatogenesis and fertility was unaffected. CONCLUSIONS: We demonstrate the successful generation and empirical validation of a cell-specific knockout of AR from VE cells, and conclude that AR expression in VE cells is not essential for spermatogenesis or male fertility.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA