Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Development ; 151(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38512324

RESUMEN

The conserved MRE11-RAD50-NBS1/Xrs2 complex is crucial for DNA break metabolism and genome maintenance. Although hypomorphic Rad50 mutation mice showed normal meiosis, both null and hypomorphic rad50 mutation yeast displayed impaired meiosis recombination. However, the in vivo function of Rad50 in mammalian germ cells, particularly its in vivo role in the resection of meiotic double strand break (DSB) ends at the molecular level remains elusive. Here, we have established germ cell-specific Rad50 knockout mouse models to determine the role of Rad50 in mitosis and meiosis of mammalian germ cells. We find that Rad50-deficient spermatocytes exhibit defective meiotic recombination and abnormal synapsis. Mechanistically, using END-seq, we demonstrate reduced DSB formation and abnormal DSB end resection occurs in mutant spermatocytes. We further identify that deletion of Rad50 in gonocytes leads to complete loss of spermatogonial stem cells due to genotoxic stress. Taken together, our results reveal the essential role of Rad50 in mammalian germ cell meiosis and mitosis, and provide in vivo views of RAD50 function in meiotic DSB formation and end resection at the molecular level.


Asunto(s)
Roturas del ADN de Doble Cadena , Animales , Masculino , Ratones , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Mutación con Pérdida de Función , Mamíferos/metabolismo , Meiosis/genética , Mutación , Espermatocitos/metabolismo , Células Germinativas/metabolismo , Ácido Anhídrido Hidrolasas/genética , Ácido Anhídrido Hidrolasas/metabolismo
3.
J Mol Cell Biol ; 14(11)2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-36484653

RESUMEN

Spermatogenesis is a highly complex developmental process that typically consists of mitosis, meiosis, and spermiogenesis. DNA/RNA helicase DHX36, a unique guanine-quadruplex (G4) resolvase, plays crucial roles in a variety of biological processes. We previously showed that DHX36 is highly expressed in male germ cells with the highest level in zygotene spermatocytes. Here, we deleted Dhx36 in advanced germ cells with Stra8-GFPCre and found that a Dhx36 deficiency in the differentiated spermatogonia leads to meiotic defects and abnormal spermiogenesis. These defects in late stages of spermatogenesis arise from dysregulated transcription of G4-harboring genes, which are required for meiosis. Thus, this study reveals that Dhx36 plays crucial roles in late stages of spermatogenesis.


Asunto(s)
ARN Helicasas , ARN , Masculino , ADN/genética , ADN Helicasas/genética , Meiosis , ARN Helicasas/genética , Espermatocitos , Espermatogénesis/genética , Animales , Ratones
4.
J Adv Res ; 51: 45-57, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36396044

RESUMEN

INTRODUCTION: The R-loop is a naturally formed three-strand nucleic acid structure that recently has been reported to participate in multiple biological processes and helped answer some previously unexplained scientific questions. Meiosis process involves multiple chromatin-related events such as DNA double-stranded breaks (DSB) formation, repairing and transcriptional dynamics. OBJECTIVES: Explore the regulatory roles and physiological functions of R-loops in the mammalian meiosis process. METHODS: In our study, using genome-wide S9.6 CUT & Tag seq, we first mapped the genomic distribution and dynamic changes of R-loop during the meiotic process in mice, from spermatogonia to secondary spermatocytes. And we further explore the role of R-loop in physiological conditions by constructing conditional knockout mice of Rnaseh1, which deleted the R-loop endonuclease before meiosis entry. RESULTS: R-loop predominantly distributes at promoter-related regions and varies across different meiotic stages. By joint analysis with the corresponding transcriptome, we found that the R-loop was closely related to transcription during the meiotic process. The high frequency of promoter-related R-loop in meiotic cells is usually accompanied by high transcription activity, and we further verified this in the leptotene/zygotene to the pachytene transition process. Moreover, the lack of RNase H1 caused sterility in male mice with R-loop accumulation and abnormal DSB repair in spermatocytes. Further analysis showed that abnormal R-loop accumulation in the leptotene/zygotene stages influenced transcriptional regulation in the pachytene stage. CONCLUSION: The mutual regulation of the R-loop and transcription plays an essential role in spermatogenesis. And R-loop is also important for the normal repair process of DSB during meiosis.


Asunto(s)
Roturas del ADN de Doble Cadena , Estructuras R-Loop , Masculino , Ratones , Animales , Meiosis/genética , Espermatogénesis/genética , Espermatocitos , Ratones Noqueados , Mamíferos/genética
5.
Asian J Androl ; 23(6): 547-548, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34708720
6.
Cell Death Dis ; 12(11): 989, 2021 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-34689175

RESUMEN

Proper follicle development is very important for the production of mature oocytes, which is essential for the maintenance of female fertility. This complex biological process requires precise gene regulation. The most abundant modification of mRNA, N6-methyladenosine (m6A), is involved in many RNA metabolism processes, including RNA splicing, translation, stability, and degradation. Here, we report that m6A plays essential roles during oocyte and follicle development. Oocyte-specific inactivation of the key m6A methyltransferase Mettl3 with Gdf9-Cre caused DNA damage accumulation in oocytes, defective follicle development, and abnormal ovulation. Mechanistically, combined RNA-seq and m6A methylated RNA immunoprecipitation sequencing (MeRIP-seq) data from oocytes revealed, that we found METTL3 targets Itsn2 for m6A modification and then enhances its stability to influence the oocytes meiosis. Taken together, our findings highlight the crucial roles of mRNA m6A modification in follicle development and coordination of RNA stabilization during oocyte growth.


Asunto(s)
Adenosina/análogos & derivados , Metiltransferasas/metabolismo , Oocitos/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Adenosina/metabolismo , Animales , Femenino , Ratones
7.
Asian J Androl ; 23(6): 590-599, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33533741

RESUMEN

Azoospermia patients who carry a monogenetic mutation that causes meiotic arrest may have their biological child through genetic correction in spermatogonial stem cells (SSCs). However, such therapy for infertility has not been experimentally investigated yet. In this study, a mouse model with an X-linked testis-expressed 11 (TEX11) mutation (Tex11PM/Y) identified in azoospermia patients exhibited meiotic arrest due to aberrant chromosome segregation. Tex11PM/Y SSCs could be isolated and expanded in vitro normally, and the mutation was corrected by clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated endonuclease 9 (Cas9), leading to the generation of repaired SSC lines. Whole-genome sequencing demonstrated that the mutation rate in repaired SSCs is comparable with that of autonomous mutation in untreated Tex11PM/Y SSCs, and no predicted off-target sites are modified. Repaired SSCs could restore spermatogenesis in infertile males and give rise to fertile offspring at a high efficiency. In summary, our study establishes a paradigm for the treatment of male azoospermia by combining in vitro expansion of SSCs and gene therapy.


Asunto(s)
Células Madre Germinales Adultas/metabolismo , Infertilidad Masculina/genética , Mutación/genética , Animales , Azoospermia/genética , Infertilidad Masculina/terapia , Masculino , Ratones , Espermatogénesis/genética
8.
Stem Cell Reports ; 15(4): 968-982, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33053361

RESUMEN

Sertoli cells are the major component of the spermatogonial stem cell (SSC) niche; however, regulatory mechanisms in Sertoli cells that dictate SSC fate decisions remain largely unknown. Here we revealed features of the N6-methyladenosine (m6A) mRNA modification in Sertoli cells and demonstrated the functions of WTAP, the key subunit of the m6A methyltransferase complex in spermatogenesis. m6A-sequencing analysis identified 21,909 m6A sites from 15,365 putative m6A-enriched transcripts within 6,122 genes, including many Sertoli cell-specific genes. Conditional deletion of Wtap in Sertoli cells resulted in sterility and the progressive loss of the SSC population. RNA sequencing and ribosome nascent-chain complex-bound mRNA sequencing analyses suggested that alternative splicing events of transcripts encoding SSC niche factors were sharply altered and translation of these transcripts were severely dysregulated by Wtap deletion. Collectively, this study uncovers a novel regulatory mechanism of the SSC niche and provide insights into molecular interactions between stem cells and their cognate niches in mammals.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Factores de Empalme de ARN/metabolismo , Células de Sertoli/citología , Células de Sertoli/metabolismo , Espermatogonias/citología , Nicho de Células Madre , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Infertilidad Masculina/patología , Masculino , Ratones Noqueados , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Espermatogonias/metabolismo , Nicho de Células Madre/genética , Transcripción Genética
9.
Development ; 147(6)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32094118

RESUMEN

The most significant feature of meiosis is the recombination process during prophase I. CXXC finger protein 1 (CXXC1) binds to CpG islands and mediates the deposition of H3K4me3 by the SETD1 complex. CXXC1 is also predicted to recruit H3K4me3-marked regions to the chromosome axis for the generation of double-strand breaks (DSBs) in the prophase of meiosis. Therefore, we deleted Cxxc1 before the onset of meiosis with Stra8-Cre The conditional knockout mice were completely sterile with spermatogenesis arrested at MII. Knockout of Cxxc1 led to a decrease in the H3K4me3 level from the pachytene to the MII stage and caused transcriptional disorder. Many spermatogenesis pathway genes were expressed early leading to abnormal acrosome formation in arrested MII cells. In meiotic prophase, deletion of Cxxc1 caused delayed DSB repair and improper crossover formation in cells at the pachytene stage, and more than half of the diplotene cells exhibited precocious homologous chromosome segregation in both male and female meiosis. Cxxc1 deletion also led to a significant decrease of H3K4me3 enrichment at DMC1-binding sites, which might compromise DSB generation. Taken together, our results show that CXXC1 is essential for proper meiotic crossover formation in mice and suggest that CXXC1-mediated H3K4me3 plays an essential role in meiotic prophase of spermatogenesis and oogenesis.


Asunto(s)
Intercambio Genético/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Meiosis/fisiología , Transactivadores/fisiología , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Masculino , Meiosis/genética , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oogénesis/genética , Procesamiento Proteico-Postraduccional/genética , Espermatogénesis/genética , Transactivadores/genética
10.
Cell Res ; 30(3): 256-268, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32047271

RESUMEN

Meiotic recombination is initiated by the formation of double-strand breaks (DSBs), which are repaired as either crossovers (COs) or noncrossovers (NCOs). In most mammals, PRDM9-mediated H3K4me3 controls the nonrandom distribution of DSBs; however, both the timing and mechanism of DSB fate control remain largely undetermined. Here, we generated comprehensive epigenomic profiles of synchronized mouse spermatogenic cells during meiotic prophase I, revealing spatiotemporal and functional relationships between epigenetic factors and meiotic recombination. We find that PRDM9-mediated H3K4me3 at DSB hotspots, coinciding with H3K27ac and H3K36me3, is intimately connected with the fate of the DSB. Our data suggest that the fate decision is likely made at the time of DSB formation: earlier formed DSBs occupy more open chromatins and are much more competent to proceed to a CO fate. Our work highlights an intrinsic connection between PRDM9-mediated H3K4me3 and the fate decision of DSBs, and provides new insight into the control of CO homeostasis.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Espermatogonias , Animales , Epigénesis Genética , Masculino , Profase Meiótica I , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Espermatogonias/citología , Espermatogonias/metabolismo
11.
Asian J Androl ; 22(2): 169-176, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31134915

RESUMEN

Spermatogonial development is a vital prerequisite for spermatogenesis and male fertility. However, the exact mechanisms underlying the behavior of spermatogonia, including spermatogonial stem cell (SSC) self-renewal and spermatogonial proliferation and differentiation, are not fully understood. Recent studies demonstrated that the mTOR complex 1 (mTORC1) signaling pathway plays a crucial role in spermatogonial development, but whether MTOR itself was also involved in any specific process of spermatogonial development remained undetermined. In this study, we specifically deleted Mtor in male germ cells of mice using Stra8-Cre and assessed its effect on the function of spermatogonia. The Mtor knockout (KO) mice exhibited an age-dependent perturbation of testicular development and progressively lost germ cells and fertility with age. These age-related phenotypes were likely caused by a delayed initiation of Mtor deletion driven by Stra8-Cre. Further examination revealed a reduction of differentiating spermatogonia in Mtor KO mice, suggesting that spermatogonial differentiation was inhibited. Spermatogonial proliferation was also impaired in Mtor KO mice, leading to a diminished spermatogonial pool and total germ cell population. Our results show that MTOR plays a pivotal role in male fertility and is required for spermatogonial proliferation and differentiation.


Asunto(s)
Proliferación Celular/genética , Espermatogénesis/genética , Espermatogonias/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Testículo/metabolismo , Animales , Fertilidad/genética , Masculino , Ratones , Ratones Noqueados , Espermatogonias/citología , Serina-Treonina Quinasas TOR/genética , Testículo/citología
12.
Biochim Biophys Acta Gene Regul Mech ; 1862(3): 403-411, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30391644

RESUMEN

Mammalian spermatogenesis is a highly specialized differentiation process involving precise regulatory mechanisms at the transcriptional, posttranscriptional, and translational levels. Emerging evidence has shown that N6-methyladenosine (m6A), an epitranscriptomic regulator of gene expression, can influence pre-mRNA splicing, mRNA export, turnover, and translation, which are controlled in the male germline to ensure coordinated gene expression. In this review, we summarize the typical features of m6A RNA modification on mRNA during male germline development, and highlight the function of writers, erasers, and readers of m6A during mouse spermatogenesis.


Asunto(s)
Adenina/análogos & derivados , Procesamiento Postranscripcional del ARN , Espermatogénesis , Adenina/metabolismo , Animales , Humanos , Masculino
13.
Cell Res ; 28(9): 879-896, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30061742

RESUMEN

A systematic interrogation of male germ cells is key to complete understanding of molecular mechanisms governing spermatogenesis and the development of new strategies for infertility therapies and male contraception. Here we develop an approach to purify all types of homogeneous spermatogenic cells by combining transgenic labeling and synchronization of the cycle of the seminiferous epithelium, and subsequent single-cell RNA-sequencing. We reveal extensive and previously uncharacterized dynamic processes and molecular signatures in gene expression, as well as specific patterns of alternative splicing, and novel regulators for specific stages of male germ cell development. Our transcriptomics analyses led us to discover discriminative markers for isolating round spermatids at specific stages, and different embryo developmental potentials between early and late stage spermatids, providing evidence that maturation of round spermatids impacts on embryo development. This work provides valuable insights into mammalian spermatogenesis, and a comprehensive resource for future studies towards the complete elucidation of gametogenesis.


Asunto(s)
Análisis de Secuencia de ARN , Análisis de la Célula Individual , Espermatogénesis/genética , Animales , Masculino , Ratones , Ratones Endogámicos
14.
Biol Reprod ; 99(4): 817-827, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29733332

RESUMEN

Male infertility has become an increasingly common health concern in recent years. Apart from environmental factors, nutrition, lifestyle, and sexually transmitted diseases, genetic defects are important causes of male infertility. Many genes have been demonstrated to be associated with male infertility. However, the roles of some functional genes in infertility, especially those that are specifically expressed in the reproductive system, remain to be elucidated. Here, we demonstrated that the testis-specific gene coiled-coil domain-containing 87 (Ccdc87) is critical for male fertility. Reverse-transcriptase polymerase chain reaction and western blot analyses revealed that the Ccdc87 mRNA and protein were only expressed in mouse testis. Ccdc87 expression first appeared at postnatal day 14 and remained at a relatively high level until adulthood. Male mice lacking Ccdc87 gene (Ccdc87-/-) were found to be subfertile. Approximately 20% of Ccdc87-null sperm from the testis and epididymis displayed severe abnormity of acrosome and cell nucleus. Sperm isolated from the cauda epididymides of Ccdc87-/- mice exhibited decreased initial motility but did not show any change in capacitation. Additionally, Ccdc87 disruption led to the impotency of sperm spontaneous and progesterone-induced acrosome reaction. Moreover, in vitro fertilization assays indicated that the fertilizing capacity of Ccdc87-/- sperm was significantly reduced. Taken together, these findings provide a new clue to understand the genetic causes of male infertility.


Asunto(s)
Fertilidad/fisiología , Proteínas/genética , Proteínas/fisiología , Espermatozoides/fisiología , Reacción Acrosómica/genética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Femenino , Fertilidad/genética , Expresión Génica , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Infertilidad Masculina/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Motilidad Espermática/genética , Interacciones Espermatozoide-Óvulo/genética , Espermatozoides/anomalías , Espermatozoides/ultraestructura , Testículo/metabolismo
15.
J Biol Chem ; 293(24): 9188-9197, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29716999

RESUMEN

Spermatogenesis is precisely controlled by complex gene expression programs and involves epigenetic reprogramming, including histone modification and DNA methylation. SET domain-containing 2 (SETD2) is the predominant histone methyltransferase catalyzing the trimethylation of histone H3 lysine 36 (H3K36me3) and plays key roles in embryonic stem cell differentiation and somatic cell development. However, its role in male germ cell development remains elusive. Here, we demonstrate an essential role of Setd2 for spermiogenesis, the final stage of spermatogenesis. Using RNA-seq, we found that, in postnatal mouse testes, Setd2 mRNA levels dramatically increase in 14-day-old mice. Using a germ cell-specific Setd2 knockout mouse model, we also found that targeted Setd2 knockout in germ cells causes aberrant spermiogenesis with acrosomal malformation before step 8 of the round-spermatid stage, resulting in complete infertility. Furthermore, we noted that the Setd2 deficiency results in complete loss of H3K36me3 and significantly decreases expression of thousands of genes, including those encoding acrosin-binding protein 1 (Acrbp1) and protamines, required for spermatogenesis. Our findings thus reveal a previously unappreciated role of the SETD2-dependent H3K36me3 modification in spermiogenesis and provide clues to the molecular mechanisms in epigenetic disorders underlying male infertility.


Asunto(s)
Proteínas Portadoras/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Protaminas/genética , Espermatogénesis , Acrosoma/metabolismo , Acrosoma/patología , Animales , Células Cultivadas , Código de Histonas , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Espermátides/citología , Espermátides/metabolismo , Espermátides/patología
16.
Cell Res ; 27(10): 1216-1230, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28914256

RESUMEN

Spermatogenesis is a differentiation process during which diploid spermatogonial stem cells (SSCs) produce haploid spermatozoa. This highly specialized process is precisely controlled at the transcriptional, posttranscriptional, and translational levels. Here we report that N6-methyladenosine (m6A), an epitranscriptomic mark regulating gene expression, plays essential roles during spermatogenesis. We present comprehensive m6A mRNA methylomes of mouse spermatogenic cells from five developmental stages: undifferentiated spermatogonia, type A1 spermatogonia, preleptotene spermatocytes, pachytene/diplotene spermatocytes, and round spermatids. Germ cell-specific inactivation of the m6A RNA methyltransferase Mettl3 or Mettl14 with Vasa-Cre causes loss of m6A and depletion of SSCs. m6A depletion dysregulates translation of transcripts that are required for SSC proliferation/differentiation. Combined deletion of Mettl3 and Mettl14 in advanced germ cells with Stra8-GFPCre disrupts spermiogenesis, whereas mice with single deletion of either Mettl3 or Mettl14 in advanced germ cells show normal spermatogenesis. The spermatids from double-mutant mice exhibit impaired translation of haploid-specific genes that are essential for spermiogenesis. This study highlights crucial roles of mRNA m6A modification in germline development, potentially ensuring coordinated translation at different stages of spermatogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Diferenciación Celular/genética , Metiltransferasas/genética , Espermatogénesis/genética , Adenosina/análogos & derivados , Adenosina/genética , Animales , Proliferación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , ARN Mensajero/genética , Espermátides/crecimiento & desarrollo , Espermátides/metabolismo , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Espermatozoides/crecimiento & desarrollo , Espermatozoides/metabolismo , Células Madre/metabolismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo
17.
Development ; 143(9): 1502-11, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26965368

RESUMEN

Retinoic acid (RA) signaling is crucial for spermatogonial differentiation, which is a key step for spermatogenesis. We explored the mechanisms underlying spermatogonial differentiation by targeting expression of a dominant-negative mutant of retinoic acid receptor α (RARα) specifically to the germ cells of transgenic mice to subvert the activity of endogenous receptors. Here we show that: (1) inhibition of retinoid signaling in germ cells completely blocked spermatogonial differentiation identical to vitamin A-deficient (VAD) mice; (2) the blockage of spermatogonial differentiation by impaired retinoid signaling resulted from an arrest of entry of the undifferentiated spermatogonia into S phase; and (3) retinoid signaling regulated spermatogonial differentiation through controlling expression of its direct target genes, including replication-dependent core histone genes. Taken together, our results demonstrate that the action of retinoid signaling on spermatogonial differentiation in vivo is direct through the spermatogonia itself, and provide the first evidence that this is mediated by regulation of expression of replication-dependent core histone genes.


Asunto(s)
Diferenciación Celular/genética , Receptor alfa de Ácido Retinoico/genética , Puntos de Control de la Fase S del Ciclo Celular/genética , Transducción de Señal/genética , Espermatogénesis/genética , Espermatogonias/citología , Animales , Histonas/genética , Masculino , Ratones , Ratones Transgénicos , Receptor alfa de Ácido Retinoico/metabolismo , Espermatogonias/metabolismo , Testículo/metabolismo , Tretinoina/metabolismo , Deficiencia de Vitamina A
18.
Proc Natl Acad Sci U S A ; 110(2): 543-8, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23267101

RESUMEN

Retinoic acid (RA), an active vitamin A derivative, is essential for mammalian spermatogenesis. Genetic studies have revealed that oxidation of vitamin A to retinal by retinol dehydrogenase 10 (RDH10) is critical for embryonic RA biosynthesis. However, physiological roles of RDH10 in postnatal RA synthesis remain unclear, given that Rdh10 loss-of-function mutations lead to early embryonic lethality. We conducted in vivo genetic studies of Rdh10 in postnatal mouse testes and found that an RDH10 deficiency in Sertoli cells, but not in germ cells, results in a mild germ cell depletion phenotype. A deficiency of RDH10 in both Sertoli and germ cells in juvenile mice results in a blockage of spermatogonial differentiation, similar to that seen in vitamin A-deficient animals. This defect in spermatogenesis arises from a complete deficiency in juvenile testicular RA synthesis and can be rescued by retinoid administration. Thus, in juvenile mice, the primary, but not exclusive, source of RA in the testes is Sertoli cells. In contrast, adult Rdh10-deficient mice exhibit phenotypically normal spermatogenesis, indicating that during development a change occurs in either the cellular source of RA or the retinaldehyde dehydrogenase involved in RA synthesis.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Espermatogénesis/fisiología , Oxidorreductasas de Alcohol/deficiencia , Animales , Cartilla de ADN/genética , Galactósidos , Técnicas Histológicas , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Indoles , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/metabolismo , Testículo/metabolismo , Tretinoina/metabolismo , Vitamina A/metabolismo
19.
Biol Reprod ; 86(3): 72, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22116806

RESUMEN

Increasing evidence indicates that microRNAs (miRNAs) may be critical players in spermatogenesis. The miRNA expression profiles of THY1(+)-enriched undifferentiated spermatogonia were characterized, and members of Mir-17-92 (Mirc1) and its paralog Mir-106b-25 (Mirc3) clusters are significantly downregulated during retinoic acid-induced spermatogonial differentiation, both in vitro and in vivo. The repression of microRNA clusters Mir-17-92 (Mirc1) and Mir-106b-25 (Mirc3) by retinoic acid in turn potentially upregulates the expression of Bim, Kit, Socs3, and Stat3. The male germ cell-specific Mir-17-92 (Mirc1) knockout mice exhibit small testes, a lower number of epididymal sperm, and mild defect in spermatogenesis. Absence of Mir-17-92 (Mirc1) in male germ cells dramatically increases expression of Mir-106b-25 (Mirc3) cluster miRNAs in the germ cells. These results suggest that Mir-17-92 (Mirc1) cluster and Mir-106b-25 (Mirc3) cluster miRNAs possibly functionally cooperate in regulating spermatogonial development.


Asunto(s)
Diferenciación Celular/genética , MicroARNs/fisiología , Espermatogénesis/fisiología , Animales , Regulación hacia Abajo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Espermatogénesis/genética , Testículo/citología , Testículo/fisiología , Tretinoina/farmacología
20.
Biol Reprod ; 85(1): 189-97, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21430230

RESUMEN

Spermatogonial differentiation is orchestrated by the precise control of gene expression involving retinoic acid signaling. MicroRNAs have emerged as important regulators of spermatogenesis, and here we show that the Mirlet7 family miRNAs are expressed in mouse spermatogonia and spermatocytes. Retinoic acid significantly leads to the induction of Mirlet7 miRNAs through suppression of Lin28. We further confirmed both in vitro and in vivo that expressions of Mycn, Ccnd1, and Col1a2, which are targets of Mirlet7, were downregulated during spermatogonial differentiation. These results suggest that Mirlet7 family miRNAs play a role in retinoic acid-induced spermatogonial differentiation.


Asunto(s)
Diferenciación Celular , MicroARNs/metabolismo , Espermatogonias/metabolismo , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/metabolismo , Receptores de Ácido Retinoico/metabolismo , Espermatogonias/citología , Tretinoina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...