Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Andrology ; 12(4): 899-917, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-37772683

RESUMO

BACKGROUND: Acetaminophen and ibuprofen are widely administered to babies due to their presumed safety as over-the-counter drugs. However, no reports exist on the effects of cyclooxygenase inhibitors on undifferentiated spermatogonia and spermatogonial stem cells. Infancy represents a critical period for spermatogonial stem cell formation and disrupting spermatogonial stem cells or their precursors may be associated with infertility and testicular cancer formation. OBJECTIVES: The goal of this study was to examine the molecular and functional impact of cyclooxygenase inhibition and silencing on early steps of undifferentiated spermatogonia (u spg) and spermatogonial stem cell development, to assess the potential reproductive risk of pharmaceutical cyclooxygenase inhibitors. METHODS: The effects of cyclooxygenase inhibition were assessed using the mouse C18-4 undifferentiated juvenile spermatogonial cell line model, previously shown to include cells with spermatogonial stem cell features, by measuring prostaglandins, cell proliferation, and differentiation, using cyclooxygenase 1- and cyclooxygenase 2-selective inhibitors NS398, celecoxib, and FR122047, acetaminophen, and ibuprofen. Cyclooxygenase 1 gene silencing was achieved using a stable short-hairpin RNA approach and clone selection, then assessing gene and protein expression in RNA sequencing, quantitative real-time polymerase chain reaction, and immunofluorescence studies. RESULTS: Cyclooxygenase 2 inhibitors NS398 and celecoxib, as well as acetaminophen, but not ibuprofen, dose-dependently decreased retinoic acid-induced expression of the spg differentiation gene Stra8, while NS398 decreased the spg differentiation marker Kit, suggesting that cyclooxygenase 2 is positively associated with spg differentiation. In contrast, short-hairpin RNA-based cyclooxygenase 1 silencing in C18-4 cells altered cellular morphology and upregulated Stra8 and Kit, implying that cyclooxygenase 1 prevented spg differentiation. Furthermore, RNA sequencing analysis of cyclooxygenase 1 knockdown cells indicated the activation of several signaling pathways including the TGFb, Wnt, and Notch pathways, compared to control C18-4 cells. Notch pathway genes were upregulated by selective cyclooxygenase inhibitors, acetaminophen and ibuprofen. CONCLUSION: We report that cyclooxygenase 1 and 2 differentially regulate undifferentiated spermatogonia/spermatogonial stem cell differentiation. Cyclooxygenases regulate Notch3 expression, with the Notch pathway targeted by PGD2. These data suggest an interaction between the eicosanoid and Notch signaling pathways that may be critical for the development of spermatogonial stem cells and subsequent spermatogenesis, cautioning about using cyclooxygenase inhibitors in infants.


Assuntos
Nitrobenzenos , Espermatogônias , Sulfonamidas , Neoplasias Testiculares , Humanos , Masculino , Animais , Camundongos , Espermatogônias/metabolismo , Neoplasias Testiculares/metabolismo , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 1/farmacologia , Ciclo-Oxigenase 2/metabolismo , Celecoxib/farmacologia , Celecoxib/metabolismo , Ibuprofeno/farmacologia , Acetaminofen , Espermatogênese/fisiologia , Diferenciação Celular/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , RNA/metabolismo , Testículo/metabolismo
2.
Front Endocrinol (Lausanne) ; 13: 896507, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35634494

RESUMO

Male reproductive function depends on the formation of spermatogonial stem cells from their neonatal precursors, the gonocytes. Previously, we identified several UPS enzymes dynamically altered during gonocyte differentiation. The present work focuses on understanding the role of the RING finger protein 149 (RNF149), an E3 ligase that we found to be strongly expressed in gonocytes and downregulated in spermatogonia. The quantification of RNF149 mRNA from postnatal day (PND) 2 to 35 (puberty) in rat testis, brain, liver, kidney, and heart indicated that its highest levels are found in the testis. RNF149 knock-down in PND3 rat gonocytes was performed to better understand its role in gonocyte development. While a proliferative cocktail of PDGF-BB and 17ß-estradiol (P+E) increased both the expression levels of the cell proliferation marker PCNA and RNF149 in mock cells, the effects of P+E on both genes were reduced in cells treated with RNF149 siRNA, suggesting that RNF149 expression is regulated during gonocyte proliferation and that there might be a functional link between RNF149 and PCNA. To examine RNF149 subcellular localization, EGFP-tagged RNF149 vectors were constructed, after determining the rat testis RNF149 mRNA sequence. Surprisingly, two variant transcripts were expressed in rat tissues, predicting truncated proteins, one containing the PA and the other the RING functional domains. Transfection in mouse F9 embryonal carcinoma cells and C18-4 spermatogonial cell lines showed differential subcellular profiles of the two truncated proteins. Overall, the results of this study support a role for RNF149 in gonocyte proliferation and suggest its transcription to variant mRNAs resulting in two proteins with different functional domains. Future studies will examine the respective roles of these variant proteins in the cell lines and isolated gonocytes.


Assuntos
Maturidade Sexual , Ubiquitina , Animais , Masculino , Camundongos , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Espermatogônias/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética
3.
Front Toxicol ; 4: 842565, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35295224

RESUMO

Increasing rates of infertility associated with declining sperm counts and quality, as well as increasing rates of testicular cancer are contemporary issues in the United States and abroad. These conditions are part of the Testicular Dysgenesis Syndrome, which includes a variety of male reproductive disorders hypothesized to share a common origin based on disrupted testicular development during fetal and neonatal stages of life. Male reproductive development is a highly regulated and complex process that relies on an intricate coordination between germ, Leydig, and Sertoli cells as well as other supporting cell types, to ensure proper spermatogenesis, testicular immune privilege, and endocrine function. The eicosanoid system has been reported to be involved in the regulation of fetal and neonatal germ cell development as well as overall testicular homeostasis. Moreover, non-steroidal anti-inflammatory drugs (NSAIDs) and analgesics with abilities to block eicosanoid synthesis by targeting either or both isoforms of cyclooxygenase enzymes, have been found to adversely affect male reproductive development. This review will explore the current body of knowledge on the involvement of the eicosanoid system in male reproductive development, as well as discuss adverse effects of NSAIDs and analgesic drugs administered perinatally, focusing on toxicities reported in the testis and on major testicular cell types. Rodent and epidemiological studies will be corroborated by findings in invertebrate models for a comprehensive report of the state of the field, and to add to our understanding of the potential long-term effects of NSAID and analgesic drug administration in infants.

4.
Proc Natl Acad Sci U S A ; 116(46): 23274-23283, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31591190

RESUMO

Reduced serum testosterone (T), or hypogonadism, affects millions of men and is associated with many pathologies, including infertility, cardiovascular diseases, metabolic syndrome, and decreased libido and sexual function. Administering T-replacement therapy (TRT) reverses many of the symptoms associated with low T levels. However, TRT is linked to side effects such as infertility and increased risk of prostate cancer and cardiovascular diseases. Thus, there is a need to obtain T-producing cells that could be used to treat hypogonadism via transplantation and reestablishment of T-producing cell lineages in the body. T is synthesized by Leydig cells (LCs), proposed to derive from mesenchymal cells of mesonephric origin. Although mesenchymal cells have been successfully induced into LCs, the limited source and possible trauma to donors hinders their application to clinical therapies. Alternatively, human induced pluripotent stem cells (hiPSCs), which are expandable in culture and have the potential to differentiate into all somatic cell types, have become the emerging source of autologous cell therapies. We have successfully induced the differentiation of hiPSCs into either human Leydig-like (hLLCs) or adrenal-like cells (hALCs) using chemically defined culture conditions. Factors critical for the development of LCs were added to both culture systems. hLLCs expressed all steroidogenic genes and proteins important for T biosynthesis, synthesized T rather than cortisol, secreted steroid hormones in response to dibutyryl-cAMP and 22(R)-hydroxycholesterol, and displayed ultrastructural features resembling LCs. By contrast, hALCs synthesized cortisol rather than T. The success in generating hiPSC-derived hLLCs with broad human LC (hLC) features supports the potential for hiPSC-based hLC regeneration.


Assuntos
Técnicas de Cultura de Células , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Células Intersticiais do Testículo/enzimologia , Testosterona/metabolismo , Expressão Gênica , Humanos , Células Intersticiais do Testículo/ultraestrutura , Masculino , Transcriptoma
5.
Sci Rep ; 7(1): 17759, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29259204

RESUMO

Spermatogenesis consists of a series of highly regulated processes that include mitotic proliferation, meiosis and cellular remodeling. Although alterations in gene expression are well known to modulate spermatogenesis, posttranscriptional mechanisms are less well defined. The ubiquitin proteasome system plays a significant role in protein turnover and may be involved in these posttranscriptional mechanisms. We previously identified ubiquitin ligase Huwe1 in the testis and showed that it can ubiquitinate histones. Since modulation of histones is important at many steps in spermatogenesis, we performed a complete characterization of the functions of Huwe1 in this process by examining the effects of its inactivation in the differentiating spermatogonia, spermatocytes and spermatids. Inactivation of Huwe1 in differentiating spermatogonia led to their depletion and formation of fewer pre-leptotene spermatocytes. The cell degeneration was associated with an accumulation of DNA damage response protein γH2AX, impaired downstream signalling and apoptosis. Inactivation of Huwe1 in spermatocytes indicated that Huwe1 is not essential for meiosis and spermiogenesis, but can result in accumulation of γH2AX. Collectively, these results provide a comprehensive survey of the functions of Huwe1 in spermatogenesis and reveal Huwe1's critical role as a modulator of the DNA damage response pathway in the earliest steps of spermatogonial differentiation.


Assuntos
Diferenciação Celular/fisiologia , Ligases/metabolismo , Meiose/fisiologia , Espermatogênese/fisiologia , Espermatogônias/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Feminino , Histonas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/metabolismo , Espermátides/metabolismo , Espermatócitos/metabolismo , Espermatogônias/fisiologia , Testículo/metabolismo , Testículo/fisiologia
6.
Endocrinology ; 158(11): 4000-4016, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28938460

RESUMO

Spermatogenesis is sustained by a heterogeneous population of spermatogonia that includes the spermatogonial stem cells. However, the mechanisms underlying their establishment from gonocyte embryonic precursors and their maintenance thereafter remain largely unknown. In this study, we report that inactivation of the ubiquitin ligase Huwe1 in male germ cells in mice led to the degeneration of spermatogonia in neonates and resulted in a Sertoli cell-only phenotype in the adult. Huwe1 knockout gonocytes showed a decrease in mitotic re-entry, which inhibited their transition to spermatogonia. Inactivation of Huwe1 in primary spermatogonial culture or the C18-4 cell line resulted in cell degeneration. Degeneration of Huwe1 knockout spermatogonia was associated with an increased level of histone H2AX and an elevated DNA damage response that led to apparent mitotic catastrophe but not apoptosis or senescence. Blocking this increase in H2AX prevented the degeneration of Huwe1-depleted cells. Taken together, these results reveal a previously undefined role of Huwe1 in orchestrating the physiological DNA damage response in the male germline that contributes to the establishment and maintenance of spermatogonia.


Assuntos
Diferenciação Celular/genética , Dano ao DNA/genética , Espermatogênese/genética , Espermatogônias/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Células Cultivadas , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Proteínas Supressoras de Tumor
7.
Int J Mol Sci ; 17(9)2016 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-27608010

RESUMO

Translocator protein 18 kDa (TSPO) is a high affinity cholesterol- and drug-binding protein highly expressed in steroidogenic cells, such as Leydig cells, where it plays a role in cholesterol mitochondrial transport. We have previously shown that TSPO is expressed in postnatal day 3 rat gonocytes, precursors of spermatogonial stem cells. Gonocytes undergo regulated phases of proliferation and migration, followed by retinoic acid (RA)-induced differentiation. Understanding these processes is important since their disruption may lead to the formation of carcinoma in situ, a precursor of testicular germ cell tumors (TGCTs). Previously, we showed that TSPO ligands do not regulate gonocyte proliferation. In the present study, we found that TSPO expression is downregulated in differentiating gonocytes. Similarly, in F9 embryonal carcinoma cells, a mouse TGCT cell line with embryonic stem cell properties, there is a significant decrease in TSPO expression during RA-induced differentiation. Silencing TSPO expression in gonocytes increased the stimulatory effect of RA on the expression of the differentiation marker Stra8, suggesting that TSPO exerts a repressive role on differentiation. Furthermore, in normal human testes, TSPO was located not only in Leydig cells, but also in discrete spermatogenic phases such as the forming acrosome of round spermatids. By contrast, seminomas, the most common type of TGCT, presented high levels of TSPO mRNA. TSPO protein was expressed in the cytoplasmic compartment of seminoma cells, identified by their nuclear expression of the transcription factors OCT4 and AP2G. Thus, TSPO appears to be tightly regulated during germ cell differentiation, and to be deregulated in seminomas, suggesting a role in germ cell development and pathology.


Assuntos
Proteínas de Transporte/metabolismo , Receptores de GABA-A/metabolismo , Receptores de GABA/metabolismo , Seminoma/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Humanos , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Receptores de GABA/genética , Receptores de GABA-A/genética , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia
8.
Environ Res ; 150: 496-512, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27423704

RESUMO

Plasticizers are indispensable additives providing flexibility and malleability to plastics. Among them, several phthalates, including di (2-ethylhexyl) phthalate (DEHP), have emerged as endocrine disruptors, leading to their restriction in consumer products and creating a need for new, safer plasticizers. The goal of this project was to use in vitro functional screening tools to select novel non-toxic plasticizers suitable for further in vivo evaluation. A panel of novel compounds with satisfactory plasticizer properties and biodegradability were tested, along with several commercial plasticizers, such as diisononyl-cyclohexane-1,2-dicarboxylate (DINCH®). MEHP, the monoester metabolite of DEHP was also included as reference compound. Because phthalates target mainly testicular function, including androgen production and spermatogenesis, we used the mouse MA-10 Leydig and C18-4 spermatogonial cell lines as surrogates to examine cell survival, proliferation, steroidogenesis and mitochondrial integrity. The most promising compounds were further assessed on organ cultures of rat fetal and neonatal testes, corresponding to sensitive developmental windows. Dose-response studies revealed the toxicity of most maleates and fumarates, while identifying several dibenzoate and succinate plasticizers as innocuous on Leydig and germ cells. Interestingly, DINCH®, a plasticizer marketed as a safe alternative to phthalates, exerted a biphasic effect on steroid production in MA-10 and fetal Leydig cells. MEHP was the only plasticizer inducing the formation of multinucleated germ cells (MNG) in organ culture. Overall, organ cultures corroborated the cell line data, identifying one dibenzoate and one succinate as the most promising candidates. The adoption of such collaborative approaches for developing new chemicals should help prevent the development of compounds potentially harmful to human health.


Assuntos
Ácidos Carboxílicos/toxicidade , Plastificantes/toxicidade , Animais , Bioensaio , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Masculino , Camundongos , Ratos Sprague-Dawley , Reprodução/efeitos dos fármacos , Testículo/citologia
9.
Mol Endocrinol ; 30(7): 763-82, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27167610

RESUMO

Fatty acid metabolism and steroid biosynthesis are 2 major pathways shared by peroxisomes and mitochondria. Both organelles are in close apposition to the endoplasmic reticulum, with which they communicate via interorganelle membrane contact sites to promote cellular signaling and the exchange of ions and lipids. To date, no convincing evidence of the direct contact between peroxisomes and mitochondria was reported in mammalian cells. Hormone-induced, tightly controlled steroid hormone biosynthesis requires interorganelle interactions. Using immunofluorescent staining and live-cell imaging, we found that dibutyryl-cAMP treatment of MA-10 mouse tumor Leydig cells rapidly induces peroxisomes to approach mitochondria and form peroxisome-mitochondrial contact sites/fusion, revealed by the subcellular distribution of the endogenous acyl-coenzyme A-binding domain (ACBD)2/ECI2 isoform A generated by alternative splicing, and further validated using a proximity ligation assay. This event occurs likely via a peroxisome-like structure, which is mediated by peroxisomal and mitochondrial matrix protein import complexes: peroxisomal import receptor peroxisomal biogenesis factor 5 (PEX5), and the mitochondrial import receptor subunit translocase of outer mitochondrial membrane 20 homolog (yeast) protein. Similar results were obtained using the mLTC-1 mouse tumor Leydig cells. Ectopic expression of the ACBD2/ECI2 isoform A in MA-10 cells led to increased basal and hormone-stimulated steroid formation, indicating that ACBD2/ECI2-mediated peroxisomes-mitochondria interactions favor in the exchange of metabolites and/or macromolecules between these 2 organelles in support of steroid biosynthesis. Considering the widespread occurrence of the ACBD2/ECI2 protein, we propose that this protein might serve as a tool to assist in understanding the contact between peroxisomes and mitochondria.


Assuntos
Dodecenoil-CoA Isomerase/metabolismo , Células Intersticiais do Testículo/metabolismo , Mitocôndrias/metabolismo , Peroxissomos/metabolismo , Esteroides/metabolismo , Animais , Bucladesina/farmacologia , Linhagem Celular , Dodecenoil-CoA Isomerase/genética , Citometria de Fluxo , Células Intersticiais do Testículo/efeitos dos fármacos , Masculino , Camundongos , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Receptor 1 de Sinal de Orientação para Peroxissomos/metabolismo , Peroxissomos/efeitos dos fármacos , RNA Interferente Pequeno
10.
Toxicology ; 355-356: 21-30, 2016 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-27181934

RESUMO

Previous work in our laboratory demonstrated that in-utero exposure to a mixture of the phytoestrogen Genistein (GEN), and plasticizer DEHP, induces short- and long-term alterations in testicular gene and protein expression different from individual exposures. These studies identified fetal and adult Leydig cells as sensitive targets for low dose endocrine disruptor (ED) mixtures. To further investigate the direct effects and mechanisms of toxicity of GEN and DEHP, MA-10 mouse tumor Leydig cells were exposed in-vitro to varying concentrations of GEN and MEHP, the principal bioactive metabolite of DEHP. Combined 10µM GEN+10µM MEHP had a stimulatory effect on basal progesterone production. Consistent with increased androgenicity, the mRNA of steroidogenic and cholesterol mediators Star, Cyp11a, Srb1 and Hsl, as well as upstream orphan nuclear receptors Nr2f2 and Sf1 were all significantly increased uniquely in the mixture treatment group. Insl3, a sensitive marker of Leydig endocrine disruption and cell function, was significantly decreased by combined GEN+MEHP. Lipid analysis by high-performance thin layer chromatography demonstrated the ability of combined 10µM combined GEN+MEHP, but not individual exposures, to increase levels of several neutral lipids and phospholipid classes, indicating a generalized deregulation of lipid homeostasis. Further investigation by qPCR analysis revealed a concomitant increase in cholesterol (Hmgcoa) and phospholipid (Srebp1c, Fasn) mediator mRNAs, suggesting the possible involvement of upstream LXRα agonism. These results suggest a deregulation of MA-10 Leydig function in response to a combination of GEN+MEHP. We propose a working model for GEN+MEHP doses relevant to human exposure involving LXR agonism and activation of other transcription factors. Taken more broadly, this research highlights the importance of assessing the impact of ED mixtures in multiple toxicological models across a range of environmentally relevant doses.


Assuntos
Dietilexilftalato/análogos & derivados , Disruptores Endócrinos/toxicidade , Genisteína/toxicidade , Células Intersticiais do Testículo/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Cromatografia em Camada Fina , Dietilexilftalato/administração & dosagem , Dietilexilftalato/toxicidade , Relação Dose-Resposta a Droga , Disruptores Endócrinos/administração & dosagem , Genisteína/administração & dosagem , Homeostase , Células Intersticiais do Testículo/patologia , Masculino , Camundongos , Fosfolipídeos/metabolismo , Reação em Cadeia da Polimerase , Progesterona/biossíntese , Esteroides/biossíntese , Fatores de Transcrição/metabolismo
11.
Steroids ; 103: 105-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26302977

RESUMO

For decades, only few tissues and cell types were defined as steroidogenic, capable of de novo steroid synthesis from cholesterol. However, with the refinement of detection methods, several tissues have now been added to the list of steroidogenic tissues. Besides their critical role as long-range acting hormones, steroids are also playing more discreet roles as local mediators and signaling molecules within the tissues they are produced. In testis, steroidogenesis is carried out by the Leydig cells through a broad network of proteins, mediating cholesterol delivery to CYP11A1, the first cytochrome of the steroidogenic cascade, and the sequential action of enzymes insuring the production of active steroids, the main one being testosterone. The knowledge that male germ cells can be directly regulated by steroids and that they express several steroidogenesis-related proteins led us to hypothesize that germ cells could produce steroids, acting as autocrine, intracrine and juxtacrine modulators, as a way to insure synchronized progression within spermatogenic cycles, and preventing inappropriate cell behaviors between neighboring cells. Gene expression and protein analyses of mouse and rat germ cells from neonatal gonocytes to spermatozoa showed that most steroidogenesis-associated genes are expressed in germ cells, showing cell type-, spermatogenic cycle-, and age-specific expression profiles. Highly expressed genes included genes involved in steroidogenesis and other cell functions, such as Acbd1 and 3, Tspo and Vdac1-3, and genes involved in fatty acids metabolism or synthesis, including Hsb17b4 10 and 12, implying broader roles than steroid synthesis in germ cells. These results support the possibility of an additional level of regulation of spermatogenesis exerted between adjacent germ cells.


Assuntos
Biomarcadores/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Esteroides/biossíntese , Testículo/metabolismo , Animais , Western Blotting , Células Cultivadas , Perfilação da Expressão Gênica , Células Germinativas/citologia , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/citologia
12.
Asian J Androl ; 17(3): 403-14, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25677133

RESUMO

Apoptosis is an integral part of the spermatogenic process, necessary to maintain a proper ratio of Sertoli to germ cell numbers and provide an adequate microenvironment to germ cells. Apoptosis may also represent a protective mechanism mediating the elimination of abnormal germ cells. Extensive apoptosis occurs between the first and second postnatal weeks, at the point when gonocytes, precursors of spermatogonial stem cells, should have migrated toward the basement membrane of the tubules and differentiated into spermatogonia. The mechanisms regulating this process are not well-understood. Gonocytes undergo phases of proliferation, migration, and differentiation which occur in a timely and closely regulated manner. Gonocytes failing to migrate and differentiate properly undergo apoptosis. Inadequate gonocyte differentiation has been suggested to lead to testicular germ cell tumor (TGCT) formation. Here, we examined the expression levels of apoptosis-related genes during gonocyte differentiation by quantitative real-time polymerase chain reaction, identifying 48 pro- and anti-apoptotic genes increased by at least two-fold in rat gonocytes induced to differentiate by retinoic acid, when compared to untreated gonocytes. Further analysis of the most highly expressed genes identified the pro-apoptotic genes Gadd45a and Cycs as upregulated in differentiating gonocytes and in spermatogonia compared with gonocytes. These genes were also significantly downregulated in seminomas, the most common type of TGCT, compared with normal human testicular tissues. These results indicate that apoptosis-related genes are actively regulated during gonocyte differentiation. Moreover, the down-regulation of pro-apoptotic genes in seminomas suggests that they could represent new therapeutic targets in the treatment of TGCTs.


Assuntos
Células-Tronco Adultas/patologia , Apoptose/genética , Diferenciação Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Seminoma/genética , Neoplasias Testiculares/genética , Células-Tronco Adultas/efeitos dos fármacos , Animais , Caspase 9/genética , Diferenciação Celular/efeitos dos fármacos , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Regulação para Baixo/genética , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Ratos , Ratos Sprague-Dawley , Seminoma/patologia , Neoplasias Testiculares/patologia , Tretinoína/farmacologia , Proteínas GADD45
13.
Reproduction ; 149(3): R139-57, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25670871

RESUMO

The production of spermatozoa relies on a pool of spermatogonial stem cells (SSCs), formed in infancy from the differentiation of their precursor cells, the gonocytes. Throughout adult life, SSCs will either self-renew or differentiate, in order to maintain a stem cell reserve while providing cells to the spermatogenic cycle. By contrast, gonocytes represent a transient and finite phase of development leading to the formation of SSCs or spermatogonia of the first spermatogenic wave. Gonocyte development involves phases of quiescence, cell proliferation, migration, and differentiation. Spermatogonia, on the other hand, remain located at the basement membrane of the seminiferous tubules throughout their successive phases of proliferation and differentiation. Apoptosis is an integral part of both developmental phases, allowing for the removal of defective cells and the maintenance of proper germ-Sertoli cell ratios. While gonocytes and spermatogonia mitosis are regulated by distinct factors, they both undergo differentiation in response to retinoic acid. In contrast to postpubertal spermatogenesis, the early steps of germ cell development have only recently attracted attention, unveiling genes and pathways regulating SSC self-renewal and proliferation. Yet, less is known on the mechanisms regulating differentiation. The processes leading from gonocytes to spermatogonia have been seldom investigated. While the formation of abnormal gonocytes or SSCs could lead to infertility, defective gonocyte differentiation might be at the origin of testicular germ cell tumors. Thus, it is important to better understand the molecular mechanisms regulating these processes. This review summarizes and compares the present knowledge on the mechanisms regulating mammalian gonocyte and spermatogonial differentiation.


Assuntos
Diferenciação Celular/genética , Células Germinativas/metabolismo , Células de Sertoli/metabolismo , Espermatogônias/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Espermatogênese/genética
14.
Endocrinology ; 156(1): 346-59, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25380237

RESUMO

Neonatal gonocytes are direct precursors of spermatogonial stem cells, the cell pool that supports spermatogenesis. Although unipotent in vivo, gonocytes express pluripotency genes common with embryonic stem cells. Previously, we found that all-trans retinoic acid (RA) induced the expression of differentiation markers and a truncated form of platelet-derived growth factor receptor (PDGFR)ß in rat gonocytes, as well as in F9 mouse embryonal carcinoma cells, an embryonic stem cell-surrogate that expresses somatic lineage markers in response to RA. The present study is focused on identifying the signaling pathways involved in RA-induced gonocyte and F9 cell differentiation. Mitogen-activated protein kinase kinase (MEK) 1/2 activation was required during F9 cell differentiation towards somatic lineage, whereas its inhibition potentiated RA-induced Stra8 expression, suggesting that MEK1/2 acts as a lineage specification switch in F9 cells. In both cell types, RA increased the expression of the spermatogonial/premeiotic marker Stra8, which is in line with F9 cells being at a stage before somatic-germline lineage specification. Inhibiting PDGFR kinase activity reduced RA-induced Stra8 expression. Interestingly, RA increased the expression of PDGFRα variant forms in both cell types. Together, these results suggest a potential cross talk between RA and PDGFR signaling pathways in cell differentiation. RA receptor-α inhibition partially reduced RA effects on Stra8 in gonocytes, indicating that RA acts in part via RA receptor-α. RA-induced gonocyte differentiation was significantly reduced by inhibiting SRC (v-src avian sarcoma [Schmidt-Ruppin A-2] viral oncogene) and JAK2/STAT5 (Janus kinase 2/signal transducer and activator of transcription 5) activities, implying that these signaling molecules play a role in gonocyte differentiation. These results suggest that gonocyte and F9 cell differentiation is regulated via cross talk between RA and PDGFRs using different downstream pathways.


Assuntos
Células-Tronco de Carcinoma Embrionário/citologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Tretinoína/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Linhagem Celular , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Ceratolíticos/farmacologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Masculino , Camundongos , Fator de Crescimento Derivado de Plaquetas/genética , Ratos , Ratos Sprague-Dawley , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Testículo/citologia , Testículo/crescimento & desenvolvimento
15.
Mol Ther ; 22(10): 1779-91, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24947306

RESUMO

Low testosterone (T), a major cause of male hypogonadism and infertility, is linked to mood changes, fatigue, osteoporosis, reduced bone-mass index, and aging. The treatment of choice, T replacement therapy, has been linked with increased risk for prostate cancer and luteinizing hormone (LH) suppression, and shown to lead to infertility, cardiovascular diseases, and obesity. Alternate methods to induce T with lower side effects are desirable. In search of the mechanisms regulating T synthesis in the testes, we identified the 14-3-3ɛ protein adaptor as a negative regulator of steroidogenesis. Steroidogenesis begins in mitochondria. 14-3-3ɛ interacts with the outer mitochondrial membrane voltage-dependent anion channel (VDAC1) protein, forming a scaffold that limits the availability of cholesterol for steroidogenesis. We report the development of a tool able to induce endogenous T formation. Peptides able to penetrate testes conjugated to 14-3-3ɛ site of interaction with VDAC1 blocked 14-3-3ɛ-VDAC1 interactions while at the same time increased VDAC1-translocator protein (18 kDa) interactions that induced steroid formation in rat testes, leading to increased serum T levels. These peptides rescued intratesticular and serum T formation in adult male rats treated with gonadotropin-releasing hormone antagonist, which dampened LH and T production.


Assuntos
Proteínas 14-3-3/metabolismo , Androgênios/metabolismo , Mitocôndrias/metabolismo , Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química , Animais , Linhagem Celular , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Transporte Proteico , Ratos , Proteínas Recombinantes de Fusão/administração & dosagem , Esteroides/biossíntese , Testículo/efeitos dos fármacos , Testículo/metabolismo , Canal de Ânion 1 Dependente de Voltagem/química
16.
J Biol Chem ; 287(19): 15380-94, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22427666

RESUMO

Cholesterol is the sole precursor of steroid hormones in the body. The import of cholesterol to the inner mitochondrial membrane, the rate-limiting step in steroid biosynthesis, relies on the formation of a protein complex that assembles at the outer mitochondrial membrane called the transduceosome. The transduceosome contains several mitochondrial and cytosolic components, including the steroidogenic acute regulatory protein (STAR). Human chorionic gonadotropin (hCG) induces de novo synthesis of STAR, a process shown to parallel maximal steroid production. In the hCG-dependent steroidogenic MA-10 mouse Leydig cell line, the 14-3-3γ protein was identified in native mitochondrial complexes by mass spectrometry and immunoblotting, and its levels increased in response to hCG treatment. The 14-3-3 proteins bind and regulate the activity of many proteins, acting via target protein activation, modification and localization. In MA-10 cells, cAMP induces 14-3-3γ expression parallel to STAR expression. Silencing of 14-3-3γ expression potentiates hormone-induced steroidogenesis. Binding motifs of 14-3-3γ were identified in components of the transduceosome, including STAR. Immunoprecipitation studies demonstrate a hormone-dependent interaction between 14-3-3γ and STAR that coincides with reduced 14-3-3γ homodimerization. The binding site of 14-3-3γ on STAR was identified to be Ser-194 in the STAR-related sterol binding lipid transfer (START) domain, the site phosphorylated in response to hCG. Taken together, these results demonstrate that 14-3-3γ negatively regulates steroidogenesis by binding to Ser-194 of STAR, thus keeping STAR in an unfolded state, unable to induce maximal steroidogenesis. Over time 14-3-3γ homodimerizes and dissociates from STAR, allowing this protein to induce maximal mitochondrial steroid formation.


Assuntos
Proteínas 14-3-3/metabolismo , Gonadotropina Coriônica/farmacologia , Células Intersticiais do Testículo/efeitos dos fármacos , Fosfoproteínas/metabolismo , Esteroides/biossíntese , Proteínas 14-3-3/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , AMP Cíclico/farmacologia , Expressão Gênica/efeitos dos fármacos , Immunoblotting , Imunoprecipitação , Células Intersticiais do Testículo/metabolismo , Masculino , Espectrometria de Massas , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Dados de Sequência Molecular , Fosfoproteínas/genética , Ligação Proteica/efeitos dos fármacos , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina/genética , Serina/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo
17.
Biol Reprod ; 82(5): 825-36, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20089883

RESUMO

We previously found that platelet-derived growth factor (PDGF) and 17beta-estradiol stimulate gonocyte proliferation in a dose-dependent, nonadditive manner. In the present study, we report that gonocytes express RAF1, MAP2K1, and MAPK1/3. Inhibition of RAF1 and MAP2K1/2, but not phosphoinositide-3-kinase, blocked PDGF-induced proliferation. AG-370, an inhibitor of PDGF receptor kinase activity, suppressed not only PDGF-induced proliferation but also that induced by 17beta-estradiol. In addition, RAF1 and MAP2K1/2 inhibitors blocked 17beta-estradiol-activated proliferation. The estrogen receptor antagonist ICI 182780 inhibited both the effects of 17beta-estradiol and PDGF. PDGF lost its stimulatory effect when steroid-depleted serum or no serum was used. Similarly, 17beta-estradiol did not induce gonocyte proliferation in the absence of PDGF. The xenoestrogens genistein, bisphenol A, and DES, but not coumestrol, stimulated gonocyte proliferation in a dose-dependent and PDGF-dependent manner similarly to 17beta-estradiol. Their effects were blocked by ICI 182780, suggesting that they act via the estrogen receptor. AG-370 blocked genistein and bisphenol A effects, demonstrating their requirement of PDGF receptor activation in a manner similar to 17beta-estradiol. These results demonstrate the interdependence of PDGF and estrogen pathways in stimulating in vitro gonocyte proliferation, suggesting that this critical step in gonocyte development might be regulated in vivo by the coordinated action of PDGF and estrogen. Thus, the inappropriate exposure of gonocytes to xenoestrogens might disrupt the crosstalk between the two pathways and potentially interfere with gonocyte development.


Assuntos
Proliferação de Células , Estradiol/fisiologia , Células Germinativas/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Espermatogênese/fisiologia , Animais , Imuno-Histoquímica , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-raf , Ratos , Ratos Sprague-Dawley , Receptor Cross-Talk/fisiologia , Transdução de Sinais/fisiologia , Espermatogônias/fisiologia , Testículo/citologia , Testículo/metabolismo
18.
Prog Lipid Res ; 49(3): 218-34, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20043945

RESUMO

Golgi body-mediated signaling has been linked to its fragmentation and regeneration during the mitotic cycle of the cell. During this process, Golgi-resident proteins are released to the cytosol and interact with other signaling molecules to regulate various cellular processes. Acyl-coenzyme A binding domain containing 3 protein (ACBD3) is a Golgi protein involved in several signaling events. ACBD3 protein was previously known as peripheral-type benzodiazepine receptor and cAMP-dependent protein kinase associated protein 7 (PAP7), Golgi complex-associated protein of 60kDa (GCP60), Golgi complex-associated protein 1 (GOCAP1), and Golgi phosphoprotein 1 (GOLPH1). In this review, we present the gene ontology of ACBD3, its relations to other Acyl-coenzyme A binding domain containing (ACBD) proteins, and its biological function in steroidogenesis, apoptosis, neurogenesis, and embryogenesis. We also discuss the role of ACBD3 in asymmetric cell division and cancer. New findings about ACBD3 may help understand this newly characterized signaling molecule and stimulate further research into its role in molecular endocrinology, neurology, and stem cell biology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/classificação , Apoptose , Desenvolvimento Embrionário , Complexo de Golgi/metabolismo , Humanos , Proteínas de Membrana/análise , Proteínas de Membrana/classificação , Neurogênese , Estrutura Terciária de Proteína , Transdução de Sinais
19.
Microsc Res Tech ; 72(11): 773-86, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19565636

RESUMO

Concerns have been raised about the possible role of the phytoestrogen genistein and the xenoestrogen Bisphenol A (BPA) as endocrine disruptors. In the present study, we examined the effects of fetal exposure to genistein and BPA on the Mitogen-activated protein kinase (MAPK) pathway and on testicular cell populations in neonatal and adult rat testes. At postnatal day (PND) 3, genistein (0.1-10 mg/kg/day) and BPA (1-200 mg/kg/day) induced Raf1 and Erk1/2 mRNA and protein increases in testes, mainly in Sertoli cells. No changes were seen for Mek1. At PND60, Erk1/2 protein expression remained robust in Sertoli cells and in some spermatogonia. Raf1 was predominant in Leydig cells while Mek1 was expressed strongly in spermatogonia, and they were both expressed in pachytene spermatocytes. No consistent change was seen in these proteins at PND60. Transient effects were observed on germ cell populations, while the only remaining effect on adult testicular cells was an increase in Leydig cell number. Rats exposed in utero to the two compounds did not present significant changes in circulating testosterone levels, suggesting normally functioning adult Leydig cells. Furthermore, Sertoli cell numbers were not affected by exposure to genistein and BPA. Finally, around 10% of genistein and BPA exposed rats were sterile, whereas all control rats were fertile. These data suggest that fetal exposure to genistein and BPA exerts transient effects in rat testes and that the changes observed at PND3 did not correlate with relevant changes in germ cell populations, Leydig cell function, or fertility in the adult.


Assuntos
Estrogênios/farmacologia , Células Intersticiais do Testículo/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células de Sertoli/efeitos dos fármacos , Espermatócitos/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Compostos Benzidrílicos , Contagem de Células , Perfilação da Expressão Gênica , Genisteína/farmacologia , Infertilidade/induzido quimicamente , Masculino , Fenóis/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Testículo/crescimento & desenvolvimento
20.
Birth Defects Res C Embryo Today ; 87(1): 1-26, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19306346

RESUMO

Male germ cells, the repository cells of the genome, comprise several successive developmental stages starting in the embryo and ending up with the spermatozoon. Gonocytes represent the fetal and neonatal stages preceding the formation of spermatogonial stem cells. Recent findings shows that germline stem cells can be driven to pluripotency and used as alternative for embryonic stem cells prompted more effort in identifying the processes regulating the development of their precursors, the testicular gonocytes. Also called pre- or pro-spermatogonia, gonocytes represent not one, but several successive developmental stages between the time at which the germ cell becomes resident in the forming fetal testis to the time it migrates to the basement membrane of the seminiferous cord to adopt a spermatogonial phenotype. This review summarizes the findings regarding the genetic identity of gonocytes, providing a description of the "common" gene expression profiles of fetal and neonatal gonocytes, as well as information on the main regulatory factors of gonocyte functions. A better comprehension of gonocyte development should help in the understanding of how germline stem cells are formed, possibly providing valuable clues on the origins of germ cell tumors or infertility.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias/fisiologia , Túbulos Seminíferos/citologia , Espermatogênese , Espermatogônias/fisiologia , Androgênios/metabolismo , Animais , Apoptose/fisiologia , Movimento Celular/fisiologia , Disruptores Endócrinos/metabolismo , Humanos , Masculino , Espermatogônias/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA