Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Reproduction ; 158(3): 267-280, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31299635

RESUMEN

Expression profiles and subcellular localisations of core Drosophila behaviour/human splicing (DBHS) proteins (PSPC1, SFPQ and NONO) and NEAT1, a long noncoding RNA (lncRNA), are investigated in developing and adult mouse testes. Core DBHS proteins are markers for the distinct subnuclear domain termed paraspeckles, while a long NEAT1 isoform scaffold facilitates paraspeckle nucleation. Paraspeckles contain many proteins (>40) and are broadly involved in RNA metabolism, including transcriptional regulation by protein sequestration, nuclear retention of A-to-I edited RNA transcripts to regulate translation and promoting survival during cellular stress. Immunohistochemistry reveals cell-specific profiles for core DBHS paraspeckle protein expression, indicating their functional diversity. PSPC1 is an androgen receptor co-activator, and it is detected in differentiating Sertoli cell nuclei from day 15 onwards, as they develop androgen responsiveness. PSPC1 is nuclear in the most mature male germ cell type present at each age, from foetal to adult life. In adult mouse testes, PSPC1 and SFPQ are present in Sertoli cells, spermatocytes and round spermatids, while the NEAT1 lncRNA appears in the punctate nuclear foci delineating paraspeckles only within Leydig cells. Identification of NEAT1 in the cytoplasm of spermatogonia and spermatocytes must reflect non-paraspeckle-related functions. NONO was absent from germ cells but nuclear in Sertoli cells. Reciprocal nuclear profiles of PSPC1 and γ-H2AX in spermatogenic cells suggest that each performs developmentally regulated roles in stress responses. These findings demonstrate paraspeckles and paraspeckle-related proteins contribute to diverse functions during testis development and spermatogenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Células Intersticiales del Testículo/metabolismo , Factor de Empalme Asociado a PTB/metabolismo , Proteínas de Unión al ARN/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Masculino , Ratones , Factor de Empalme Asociado a PTB/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/genética , Células de Sertoli/metabolismo , Testículo/crecimiento & desarrollo
2.
Development ; 145(13)2018 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-29899137

RESUMEN

Spermatogenesis in mammals is a very complex, highly organized process, regulated in part by testosterone and retinoic acid (RA). Much is known about how RA and testosterone signaling pathways independently regulate this process, but there is almost no information regarding whether these two signaling pathways directly interact and whether RA is crucial for steroidogenic cell function. This study uses a transgenic mouse line that expresses a dominant-negative form of RA receptor α (RAR-DN) and the steroidogenic cell-specific Cre mouse line, Cyp17iCre, to generate male mice with steroidogenic cells unable to perform RA signaling. Testes of mutant mice displayed increased apoptosis of pachytene spermatocytes, an increased number of macrophages in the interstitium and a loss of advanced germ cells. Additionally, blocking RA signaling in Leydig cells resulted in increased permeability of the blood-testis barrier, decreased levels of the steroidogenic enzyme cytochrome P450 17a1 and decreased testosterone levels. Surprisingly, the epididymides of the mutant mice also displayed an abnormal phenotype. This study demonstrates that RA signaling is required in steroidogenic cells for their normal function and, thus, for male fertility.


Asunto(s)
Barrera Hematotesticular/metabolismo , Fertilidad/fisiología , Receptor alfa de Ácido Retinoico/metabolismo , Transducción de Señal/fisiología , Espermatocitos/metabolismo , Espermatogénesis/fisiología , Animales , Barrera Hematotesticular/citología , Masculino , Ratones , Ratones Transgénicos , Receptor alfa de Ácido Retinoico/genética , Espermatocitos/citología , Esteroide 17-alfa-Hidroxilasa/genética , Esteroide 17-alfa-Hidroxilasa/metabolismo
3.
Biol Reprod ; 98(5): 722-738, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29408990

RESUMEN

Spermatogenesis in mammals occurs in a very highly organized manner within the seminiferous epithelium regulated by different cell types in the testis. Testosterone produced by Leydig cells regulates blood-testis barrier formation, meiosis, spermiogenesis, and spermiation. However, it is unknown whether Leydig cell function changes with the different stages of the seminiferous epithelium. This study utilized the WIN 18,446 and retinoic acid (RA) treatment regime combined with the RiboTag mouse methodology to synchronize male germ cell development and allow for the in vivo mapping of the Leydig cell translatome across the different stages of one cycle of the seminiferous epithelium. Using microarrays analysis, we identified 11 Leydig cell-enriched genes that were expressed in stage-specific manner such as the glucocorticoid synthesis and transport genes, Cyp21a1 and Serpina6. In addition, there were nine Leydig cell transcripts that change their association with polysomes in correlation with the different stages of the spermatogenic cycle including Egr1. Interestingly, the signal intensity of EGR1 and CYP21 varied among Leydig cells in the adult asynchronous testis. However, testosterone levels across the different stages of germ cell development did not cycle. These data show, for the first time, that Leydig cell gene expression changes in a stage-specific manner during the cycle of the seminiferous epithelium and indicate that a heterogeneous Leydig cell population exists in the adult mouse testis.


Asunto(s)
Células Intersticiales del Testículo/metabolismo , Polirribosomas/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Barrera Hematotesticular , Expresión Génica , Células Intersticiales del Testículo/citología , Masculino , Ratones , Epitelio Seminífero/citología , Epitelio Seminífero/metabolismo , Esteroide 21-Hidroxilasa/genética , Esteroide 21-Hidroxilasa/metabolismo , Testículo/citología , Transcortina/genética , Transcortina/metabolismo
4.
Dev Biol ; 432(2): 229-236, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29037932

RESUMEN

The onset of spermatogenesis occurs in response to retinoic acid (RA), the active metabolite of vitamin A. However, whether RA plays any role during establishment of the spermatogonial stem cell (SSC) pool is unknown. Because designation of the SSC population and the onset of RA signaling in the testis that induces differentiation have similar timing, this study asked whether RA influenced SSC establishment. Whole mount immunofluorescence and flow cytometric analysis using the Id4-eGfp transgenic reporter mouse line revealed an enrichment for ID4-EGFP+ cells within the testis following inhibition of RA synthesis by WIN 18,446 treatment. Transplantation analyses confirmed a significant increase in the number of SSCs in testes from RA-deficient animals. Conversely, no difference in the ID4-EGFP+ population or change in SSC number were detected following exposure to an excess of RA. Collectively, reduced RA altered the number of SSCs present in the neonatal testis but precocious RA exposure in the neonatal testis did not, suggesting that RA deficiency causes a greater proportion of progenitor undifferentiated spermatogonia to retain their SSC state past the age when the pool is thought to be determined.


Asunto(s)
Espermatogénesis/fisiología , Tretinoina/metabolismo , Células Madre Germinales Adultas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal/efectos de los fármacos , Espermatogénesis/genética , Espermatogonias/citología , Testículo/metabolismo
5.
Proc Natl Acad Sci U S A ; 114(28): E5635-E5644, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28630288

RESUMEN

The PIWI-interacting RNA (piRNA) pathway is essential for retrotransposon silencing. In piRNA-deficient mice, L1-overexpressing male germ cells exhibit excessive DNA damage and meiotic defects. It remains unknown whether L1 expression simply highlights piRNA deficiency or actually drives the germ-cell demise. Specifically, the sheer abundance of genomic L1 copies prevents reliable quantification of new insertions. Here, we developed a codon-optimized L1 transgene that is controlled by an endogenous mouse L1 promoter. Importantly, DNA methylation dynamics of a single-copy transgene were indistinguishable from those of endogenous L1s. Analysis of Mov10l1-/- testes established that de novo methylation of the L1 transgene required the intact piRNA pathway. Consistent with loss of DNA methylation and programmed reduction of H3K9me2 at meiotic onset, the transgene showed 1,400-fold increase in RNA expression and consequently 70-fold increase in retrotransposition in postnatal day 14 Mov10l1-/- germ cells compared with the wild-type. Analysis of adult Mov10l1-/- germ-cell fractions indicated a stage-specific increase of retrotransposition in the early meiotic prophase. However, extrapolation of the transgene data to endogenous L1s suggests that it is unlikely insertional mutagenesis alone accounts for the Mov10l1-/- phenotype. Indeed, pharmacological inhibition of reverse transcription did not rescue the meiotic defect. Cumulatively, these results establish the occurrence of productive L1 mobilization in the absence of an intact piRNA pathway but leave open the possibility of processes preceding L1 integration in triggering meiotic checkpoints and germ-cell death. Additionally, our data suggest that many heritable L1 insertions originate from individuals with partially compromised piRNA defense.


Asunto(s)
Meiosis , ARN Interferente Pequeño/metabolismo , Retroelementos , Transgenes , Regiones no Traducidas 5' , Animales , Codón , Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Masculino , Metilación , Ratones , Ratones Transgénicos , Sistemas de Lectura Abierta , Fenotipo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Espermatocitos/metabolismo , Espermatogénesis , Testículo/metabolismo
6.
Biol Reprod ; 95(4): 81, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27488029

RESUMEN

Retinoic acid (RA), the active metabolite of vitamin A, is known to be required for the differentiation of spermatogonia. The first round of spermatogenesis initiates in response to RA and occurs in patches along the length of the seminiferous tubule. However, very little is known about the individual differentiating spermatogonial populations and their progression through the cell cycle due to the heterogeneous nature of the onset of spermatogenesis. In this study, we utilized WIN 18,446 and RA as tools to generate testes enriched with different populations of spermatogonia to further investigate 1) the undifferentiated to differentiating spermatogonial transition, 2) the progression of the differentiating spermatogonia through the cell cycle, and 3) Sertoli cell number in response to altered RA levels. WIN 18,446/RA-treated neonatal mice were used to determine when synchronous S phases occurred in the differentiating spermatogonial population following treatment. Five differentiating spermatogonial S phase windows were identified between spermatogonial differentiation and formation of preleptotene spermatocytes. In addition, a slight increase in Sertoli cell number was observed following RA treatment, possibly implicating a role for RA in Sertoli cell cycle progression. This study has enhanced our understanding of the spermatogonial populations present in the neonatal testis during the onset of spermatogenesis by mapping the cell cycle kinetics of both the undifferentiated and the differentiating spermatogonial populations and identifying the precise timing of when specific individual differentiating spermatogonial populations are enriched within the testis following synchrony, thus providing an essential tool for further study of the differentiating spermatogonia.


Asunto(s)
Espermatogénesis/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Tretinoina/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Diaminas/farmacología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Microscopía Fluorescente , Túbulos Seminíferos/metabolismo , Células de Sertoli/citología , Células de Sertoli/efectos de los fármacos , Transducción de Señal , Espermatogénesis/fisiología , Espermatogonias/citología , Espermatogonias/fisiología , Testículo/citología , Testículo/efectos de los fármacos , Testículo/fisiología , Tretinoina/fisiología
7.
Results Probl Cell Differ ; 58: 135-66, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27300178

RESUMEN

The core of the decision to commit to either oogenesis or spermatogenesis lies in the timing of meiotic entry. Primordial germ cells within the fetal ovary become committed to the female pathway prior to birth and enter meiosis during embryonic development. In the fetal testis, however, the germ cells are protected from this signal before birth and instead receive this trigger postnatally. There is a growing body of evidence to indicate that RA is the meiosis-inducing factor in both sexes, with the gender-specific timing of meiotic entry controlled via degradation of this molecule only within the fetal testis. This chapter will review our current understanding of how RA controls germ cell fate in both the embryonic ovary and postnatal testis, highlighting the key studies that have led to the hypothesis that RA can drive the commitment to meiosis in both sexes and discussing the current debate over whether RA truly is the meiosis-inducing factor in the fetal ovary.


Asunto(s)
Células Germinativas/citología , Oogénesis/fisiología , Transducción de Señal/fisiología , Espermatogénesis/fisiología , Tretinoina/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Humanos , Masculino , Oogénesis/genética , Transducción de Señal/genética , Espermatogénesis/genética , Tretinoina/metabolismo
8.
Mol Pharmacol ; 89(5): 560-74, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26921399

RESUMEN

All-trans-retinoic acid (atRA) is the active metabolite of vitamin A. The liver is the main storage organ of vitamin A, but activation of the retinoic acid receptors (RARs) in mouse liver and in human liver cell lines has also been shown. AlthoughatRA treatment improves mitochondrial function in skeletal muscle in rodents, its role in modulating mitochondrial function in the liver is controversial, and little data are available regarding the human liver. The aim of this study was to determine whetheratRA regulates hepatic mitochondrial activity.atRA treatment increased the mRNA and protein expression of multiple components of mitochondrialß-oxidation, tricarboxylic acid (TCA) cycle, and respiratory chain. Additionally,atRA increased mitochondrial biogenesis in human hepatocytes and in HepG2 cells with and without lipid loading based on peroxisome proliferator activated receptor gamma coactivator 1αand 1ßand nuclear respiratory factor 1 mRNA and mitochondrial DNA quantification.atRA also increasedß-oxidation and ATP production in HepG2 cells and in human hepatocytes. Knockdown studies of RARα, RARß, and PPARδrevealed that the enhancement of mitochondrial biogenesis andß-oxidation byatRA requires peroxisome proliferator activated receptor delta. In vivo in mice,atRA treatment increased mitochondrial biogenesis markers after an overnight fast. Inhibition ofatRA metabolism by talarozole, a cytochrome P450 (CYP) 26 specific inhibitor, increased the effects ofatRA on mitochondrial biogenesis markers in HepG2 cells and in vivo in mice. These studies show thatatRA regulates mitochondrial function and lipid metabolism and that increasingatRA concentrations in human liver via CYP26 inhibition may increase mitochondrial biogenesis and fatty acidß-oxidation and provide therapeutic benefit in diseases associated with mitochondrial dysfunction.


Asunto(s)
Mitocondrias Hepáticas/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , PPAR delta/agonistas , Transducción de Señal , Tretinoina/metabolismo , Regulación hacia Arriba , Animales , Benzotiazoles/farmacología , Células Cultivadas , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/enzimología , Biogénesis de Organelos , PPAR delta/antagonistas & inhibidores , PPAR delta/genética , PPAR delta/metabolismo , Interferencia de ARN , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Ácido Retinoico 4-Hidroxilasa , Receptor alfa de Ácido Retinoico , Triazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos
9.
Biol Reprod ; 94(1): 12, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26632609

RESUMEN

Perturbations in the vitamin A metabolism pathway could be a significant cause of male infertility, as well as a target toward the development of a male contraceptive, necessitating the need for a better understanding of how testicular retinoic acid (RA) concentrations are regulated. Quantitative analyses have recently demonstrated that RA is present in a pulsatile manner along testis tubules. However, it is unclear if the aldehyde dehydrogenase (ALDH) enzymes, which are responsible for RA synthesis, contribute to the regulation of these RA concentration gradients. Previous studies have alluded to fluctuations in ALDH enzymes across the spermatogenic cycle, but these inferences have been based primarily on qualitative transcript localization experiments. Here, we show via various quantitative methods that the three well-known ALDH enzymes (ALDH1A1, ALDH1A2, and ALDH1A3), and an ALDH enzyme previously unreported in the murine testis (ALDH8A1), are not expressed in a stage-specific manner in the adult testis, but do fluctuate throughout juvenile development in perfect agreement with the first appearance of each advancing germ cell type. We also show, via treatments with a known ALDH inhibitor, that lowered testicular RA levels result in an increase in blood-testis barrier permeability, meiotic recombination, and meiotic defects. Taken together, these data further our understanding of the complex regulatory actions of RA on various spermatogenic events and, in contrast with previous studies, also suggest that the ALDH enzymes are not responsible for regulating the recently measured RA pulse.


Asunto(s)
Aldehído Deshidrogenasa/biosíntesis , Espermatogénesis/genética , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/genética , Animales , Biotina/metabolismo , Barrera Hematotesticular/efectos de los fármacos , Emparejamiento Cromosómico/efectos de los fármacos , Diaminas/farmacología , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/genética , Isoenzimas/metabolismo , Masculino , Meiosis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Espermatogénesis/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Tretinoina/metabolismo
10.
Biol Reprod ; 93(1): 19, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26040672

RESUMEN

The active metabolite of vitamin A, retinoic acid (RA), is known to be essential for spermatogenesis. Changes to RA levels within the seminiferous epithelium can alter the development of male germ cells, including blocking their differentiation completely. Excess RA has been shown to cause germ cell death in both neonatal and adult animals, yet the cells capable of degrading RA within the testis have yet to be investigated. One previous study alluded to a requirement for one of the RA degrading enzymes, CYP26B1, in Sertoli cells but no data exist to determine whether germ cells possess the ability to degrade RA. To bridge this gap, the roles of CYP26A1 and CYP26B1 within the seminiferous epithelium were investigated by creating single and dual conditional knockouts of these enzymes in either Sertoli or germ cells. Analysis of these knockout models revealed that deletion of both Cyp26a1 and Cyp26b1 in either cell type resulted in increased vacuolization within the seminiferous tubules, delayed spermatid release, and an increase in the number of STRA8-positive spermatogonia, but spermatozoa were still produced and the animals were found to be fertile. However, elimination of CYP26B1 activity within both germ and Sertoli cells resulted in severe male subfertility, with a loss of advanced germ cells from the seminiferous epithelium. These data indicate that CYP26 activity within either Sertoli or germ cells is essential for the normal progression of spermatogenesis and that its loss can result in reduced male fertility.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Epitelio Seminífero/enzimología , Espermatogénesis/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/genética , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Noqueados , Ácido Retinoico 4-Hidroxilasa , Células de Sertoli/metabolismo , Espermatogonias/metabolismo , Espermatozoides/metabolismo
11.
Biochem Pharmacol ; 95(3): 177-92, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25764981

RESUMEN

all-trans retinoic acid (atRA), the active metabolite of vitamin A, is an essential signaling molecule. Specifically the concentrations of atRA are spatiotemporally controlled in target tissues such as the liver and the testes. While the enzymes of the aldehyde dehydrogenase 1A family (ALDH1A) are believed to control the synthesis of atRA, a direct relationship between altered ALDH1A activity and tissue atRA concentrations has never been shown. To test whether inhibition of ALDH1A enzymes decreases atRA concentrations in a tissue specific manner, the potent ALDH1A inhibitor WIN 18,446 was used to inhibit ALDH1A activity in mice. The ALDH1A expression, atRA formation kinetics, ALDH1A inhibition by WIN 18,446 and WIN 18,446 disposition were used to predict the time course and extent of inhibition of atRA formation in the testis and liver. The effect of WIN 18,446 on atRA concentrations in testis, liver and serum were measured following single and multiple doses of WIN 18,446. ALDH1A1 and ALDH1A2 were responsible for the majority of atRA formation in the testis while ALDH1A1 and aldehyde oxidase contributed to atRA formation in the liver. Due to the different complement of enzymes contributing to atRA formation in different tissues and different inhibition of ALDH1A1 and ALDH1A2 by WIN 18,446, WIN 18,446 caused only a 50% decrease in liver atRA but testicular atRA decreased over 90%. Serum atRA concentrations were also reduced. These data demonstrate that inhibition of ALDH1A enzymes will decrease atRA concentrations in a tissue specific manner and selective ALDH1A inhibition could be used to alter atRA concentrations in select target tissues.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Tretinoina/farmacología , Aldehído Deshidrogenasa/química , Familia de Aldehído Deshidrogenasa 1 , Secuencia de Aminoácidos , Animales , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Retinal-Deshidrogenasa , Distribución Tisular , Tretinoina/farmacocinética
12.
Mol Biol Cell ; 26(8): 1543-58, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25694451

RESUMEN

Importin (IMP) superfamily members mediate regulated nucleocytoplasmic transport, which is central to key cellular processes. Although individual IMPα proteins exhibit dynamic synthesis and subcellular localization during cellular differentiation, including during spermatogenesis, little is known of how this affects cell fate. To investigate how IMPαs control cellular development, we conducted a yeast two-hybrid screen for IMPα2 cargoes in embryonic day 12.5 mouse testis, a site of peak IMPα2 expression coincident with germ-line masculization. We identified paraspeckle protein 1 (PSPC1), the original defining component of nuclear paraspeckles, as an IMPα2-binding partner. PSPC1-IMPα2 binding in testis was confirmed in immunoprecipitations and pull downs, and an enzyme-linked immunosorbent assay-based assay demonstrated direct, high-affinity PSPC1 binding to either IMPα2/IMPß1 or IMPα6/IMPß1. Coexpression of full-length PSPC1 and IMPα2 in HeLa cells yielded increased PSPC1 localization in nuclear paraspeckles. High-throughput image analysis of >3500 cells indicated IMPα2 levels can directly determine PSPC1-positive nuclear speckle numbers and size; a transport-deficient IMPα2 isoform or small interfering RNA knockdown of IMPα2 each reduced endogenous PSPC1 accumulation in speckles. This first validation of an IMPα2 nuclear import cargo in fetal testis provides novel evidence that PSPC1 delivery to paraspeckles, and consequently paraspeckle function, may be controlled by modulated synthesis of specific IMPs.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Testículo/metabolismo , Transporte Activo de Núcleo Celular , Animales , Inmunoprecipitación , Masculino , Ratones , Testículo/embriología , Técnicas del Sistema de Dos Híbridos , alfa Carioferinas
13.
Dev Biol ; 397(1): 140-9, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25446031

RESUMEN

In the testis, a subset of spermatogonia retains stem cell potential, while others differentiate to eventually become spermatozoa. This delicate balance must be maintained, as defects can result in testicular cancer or infertility. Currently, little is known about the gene products and signaling pathways directing these critical cell fate decisions. Retinoic acid (RA) is a requisite driver of spermatogonial differentiation and entry into meiosis, yet the mechanisms activated downstream are undefined. Here, we determined a requirement for RA in the expression of KIT, a receptor tyrosine kinase essential for spermatogonial differentiation. We found that RA signaling utilized the PI3K/AKT/mTOR signaling pathway to induce the efficient translation of mRNAs for Kit, which are present but not translated in undifferentiated spermatogonia. Our findings provide an important molecular link between a morphogen (RA) and the expression of KIT protein, which together direct the differentiation of spermatogonia throughout the male reproductive lifespan.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Espermatogénesis , Tretinoina/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Espermatogonias/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Testículo/metabolismo
14.
J Lipid Res ; 56(2): 342-57, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25502770

RESUMEN

Retinoic acid (RA), the active metabolite of vitamin A, is required for spermatogenesis and many other biological processes. RA formation requires irreversible oxidation of retinal to RA by aldehyde dehydrogenase enzymes of the 1A family (ALDH1A). While ALDH1A1, ALDH1A2, and ALDH1A3 all form RA, the expression pattern and relative contribution of these enzymes to RA formation in the testis is unknown. In this study, novel methods to measure ALDH1A protein levels and intrinsic RA formation were used to accurately predict RA formation velocities in individual human testis samples and an association between RA formation and intratesticular RA concentrations was observed. The distinct localization of ALDH1A in the testis suggests a specific role for each enzyme in controlling RA formation. ALDH1A1 was found in Sertoli cells, while only ALDH1A2 was found in spermatogonia, spermatids, and spermatocytes. In the absence of cellular retinol binding protein (CRBP)1, ALDH1A1 was predicted to be the main contributor to intratesticular RA formation, but when CRBP1 was present, ALDH1A2 was predicted to be equally important in RA formation as ALDH1A1. This study provides a comprehensive novel methodology to evaluate RA homeostasis in human tissues and provides insight to how the individual ALDH1A enzymes mediate RA concentrations in specific cell types.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Testículo/metabolismo , Anciano , Anciano de 80 o más Años , Aldehído Deshidrogenasa/genética , Familia de Aldehído Deshidrogenasa 1 , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Cromatografía Liquida , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Espectrometría de Masas en Tándem , Tretinoina/metabolismo
15.
Biol Reprod ; 92(2): 37, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25519186

RESUMEN

The asynchronous cyclic nature of spermatogenesis is essential for continual sperm production and is one of the hallmarks of mammalian male fertility. While various mRNA and protein localization studies have indirectly implicated changing retinoid levels along testis tubules, no quantitative evidence for these changes across the cycle of the seminiferous epithelium currently exists. This study utilized a unique mouse model of induced synchronous spermatogenesis, localization of the retinoid-signaling marker STRA8, and sensitive quantification of retinoic acid concentrations to determine whether there are fluctuations in retinoid levels at each of the individual stages of germ cell differentiation and maturation to sperm. These data show that processive pulses of retinoic acid are generated during spermatogonial differentiation and are the likely trigger for cyclic spermatogenesis and allow us, for the first time, to understand how the cycle of the seminiferous epithelium is generated and maintained. In addition, this study represents the first direct quantification of a retinoid gradient controlling cellular differentiation in a postnatal tissue.


Asunto(s)
Espermatogénesis/efectos de los fármacos , Testículo/efectos de los fármacos , Tretinoina/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Masculino , Ratones , Receptores de Ácido Retinoico/metabolismo , Testículo/metabolismo , Tretinoina/metabolismo
16.
Biol Open ; 4(1): 1-12, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25505149

RESUMEN

The spermatogenic cycle describes the periodic development of germ cells in the testicular tissue. The temporal-spatial dynamics of the cycle highlight the unique, complex, and interdependent interaction between germ and somatic cells, and are the key to continual sperm production. Although understanding the spermatogenic cycle has important clinical relevance for male fertility and contraception, there are a number of experimental obstacles. For example, the lengthy process cannot be visualized through dynamic imaging, and the precise action of germ cells that leads to the emergence of testicular morphology remains uncharacterized. Here, we report an agent-based model that simulates the mouse spermatogenic cycle on a cross-section of the seminiferous tubule over a time scale of hours to years, while considering feedback regulation, mitotic and meiotic division, differentiation, apoptosis, and movement. The computer model is able to elaborate the germ cell dynamics in a time-lapse movie format, allowing us to trace individual cells as they change state and location. More importantly, the model provides mechanistic understanding of the fundamentals of male fertility, namely how testicular morphology and sperm production are achieved. By manipulating cellular behaviors either individually or collectively in silico, the model predicts causal events for the altered arrangement of germ cells upon genetic or environmental perturbations. This in silico platform can serve as an interactive tool to perform long-term simulation and to identify optimal approaches for infertility treatment and contraceptive development.

17.
PLoS Genet ; 10(8): e1004541, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25102060

RESUMEN

In all sexually reproducing organisms, cells of the germ line must transition from mitosis to meiosis. In mice, retinoic acid (RA), the extrinsic signal for meiotic initiation, activates transcription of Stra8, which is required for meiotic DNA replication and the subsequent processes of meiotic prophase. Here we report that RA also activates transcription of Rec8, which encodes a component of the cohesin complex that accumulates during meiotic S phase, and which is essential for chromosome synapsis and segregation. This RA induction of Rec8 occurs in parallel with the induction of Stra8, and independently of Stra8 function, and it is conserved between the sexes. Further, RA induction of Rec8, like that of Stra8, requires the germ-cell-intrinsic competence factor Dazl. Our findings strengthen the importance of RA and Dazl in the meiotic transition, provide important details about the Stra8 pathway, and open avenues to investigate early meiosis through analysis of Rec8 induction and function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Meiosis/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética , Tretinoina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Replicación del ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Germinativas/crecimiento & desarrollo , Masculino , Ratones , Mitosis/genética , Proteínas Nucleares/biosíntesis , Ovario/efectos de los fármacos , Ovario/crecimiento & desarrollo , Fosfoproteínas/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Transducción de Señal/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/crecimiento & desarrollo , Transcripción Genética/efectos de los fármacos , Tretinoina/administración & dosificación
18.
Spermatogenesis ; 3(1): e23180, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23687613

RESUMEN

Retinoic acid (RA) is required for the successful differentiation and meiotic entry of germ cells in the murine testis. The availability of RA to undifferentiated germ cells begins in a variable, uneven pattern during the first few days after birth and establishes the asynchronous pattern of germ cell differentiation in adulthood. It has been shown that synchronous spermatogenesis can be induced in 2 d postpartum mice, but not in adult mice, by treating vitamin A sufficient males with RA. In this study, neonatal males were treated at different ages with a single dose of RA and spermatogenesis was examined after recovery to adulthood. The failure of exogenous RA to alter asynchrony correlates with the appearance of meiotic preleptotene spermatocytes within the seminiferous epithelium.

20.
Curr Opin Endocrinol Diabetes Obes ; 20(3): 217-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23511242

RESUMEN

PURPOSE OF REVIEW: Description of new evidence to support the model for how retinoic acid regulates spermatogonial differentiation, male meiosis and the cycle of the seminiferous epithelium. RECENT FINDINGS: It has been known since the 1920s that vitamin A is essential for spermatogenesis. However, only recently has significant progress been made toward understanding how the active metabolite of vitamin A, retinoic acid, regulates spermatogenesis at multiple different differentiation steps, including the onset of meiosis. Current publications suggest that the initiation and maintenance of the cycle of the seminiferous epithelium is linked to retinoic-acid-driving spermatogonial differentiation and meiotic onset. SUMMARY: Retinoic acid appears to act in a pulsatile manner, periodically driving spermatogonial differentiation and meiotic onset at discrete points along testis tubules, and as a result, is likely to be responsible for generating and maintaining the cycle of the seminiferous epithelium.


Asunto(s)
Modelos Biológicos , Epitelio Seminífero/metabolismo , Espermatogénesis , Tretinoina/metabolismo , Animales , Humanos , Masculino , Meiosis , Receptores de Ácido Retinoico/metabolismo , Epitelio Seminífero/citología , Epitelio Seminífero/crecimiento & desarrollo , Transducción de Señal , Espermatozoides/citología , Espermatozoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA