Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 148(5)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33574039

RESUMEN

In mice, the entry of germ cells into meiosis crucially depends on the expression of stimulated by retinoic acid gene 8 (Stra8). Stra8 is expressed specifically in pre-meiotic germ cells of females and males, at fetal and postnatal stages, respectively, but the mechanistic details of its spatiotemporal regulation are yet to be defined. In particular, there has been considerable debate regarding whether retinoic acid is required, in vivo, to initiate Stra8 expression in the mouse fetal ovary. We show that the distinctive anterior-to-posterior pattern of Stra8 initiation, characteristic of germ cells in the fetal ovary, is faithfully recapitulated when 2.9 kb of the Stra8 promoter is used to drive eGFP expression. Using in vitro transfection assays of cutdown and mutant constructs, we identified two functional retinoic acid responsive elements (RAREs) within this 2.9 kb regulatory element. We also show that the transcription factor DMRT1 enhances Stra8 expression, but only in the presence of RA and the most proximal RARE. Finally, we used CRISPR/Cas9-mediated targeted mutation studies to demonstrate that both RAREs are required for optimal Stra8 expression levels in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Germinativas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Sitios de Unión , Sistemas CRISPR-Cas/genética , Femenino , Desarrollo Fetal/genética , Feto/citología , Feto/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Germinativas/citología , Meiosis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis , Ovario/citología , Ovario/metabolismo , Regiones Promotoras Genéticas , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/farmacología , Tretinoina/farmacología
2.
Development ; 148(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34913465

RESUMEN

Spermatogonial differentiation and meiotic initiation during spermatogenesis are tightly regulated by a number of genes, including those encoding enzymes for miRNA biogenesis. However, whether and how single miRNAs regulate these processes remain unclear. Here, we report that miR-202, a member of the let-7 family, prevents precocious spermatogonial differentiation and meiotic initiation in spermatogenesis by regulating the timely expression of many genes, including those for key regulators such as STRA8 and DMRT6. In miR-202 knockout (KO) mice, the undifferentiated spermatogonial pool is reduced, accompanied by age-dependent decline of fertility. In KO mice, SYCP3, STRA8 and DMRT6 are expressed earlier than in wild-type littermates, and Dmrt6 mRNA is a direct target of miR-202-5p. Moreover, the precocious spermatogonial differentiation and meiotic initiation were also observed in KO spermatogonial stem cells when cultured and induced in vitro, and could be partially rescued by the knockdown of Dmrt6. Therefore, we have not only shown that miR-202 is a regulator of meiotic initiation but also identified a previously unknown module in the underlying regulatory network.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , MicroARNs/genética , Espermatogénesis/genética , Espermatogonias/crecimiento & desarrollo , Testículo/crecimiento & desarrollo , Células Madre Germinales Adultas/citología , Animales , Proteínas de Ciclo Celular/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Fertilidad/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Meiosis/genética , Ratones , Ratones Noqueados , Espermatogonias/metabolismo , Testículo/metabolismo , Factores de Transcripción/genética
3.
Biol Reprod ; 109(2): 184-191, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37279768

RESUMEN

The development of oocytes occurs over a broad time frame, starting at the earliest stages of embryogenesis and continuing into adulthood. Conditional knockout technologies such as the Cre/loxP recombination system are useful for analyzing oocyte development at specific stages, but not every time frame has appropriate Cre drivers, for instance, during oocyte meiotic initiation through early prophase I in the embryo. Here, we generated a novel knockin mouse line that produces a bicistronic transcript from the endogenous Stra8 locus that includes a "self-cleaving" 2A peptide upstream of cre. This allows for high efficiency cleavage and production of both proteins individually and results in expression of cre in both male and female gonads at the biologically relevant stage. Fluorescent reporter analysis confirms that this line recapitulates endogenous Stra8 expression in both sexes and does not affect fertility of heterozygous nor homozygous mice. This line, named Stra8P2Acre, adds to the repertoire of germ-cell specific cre driver lines and, importantly, allows for deletion of target genes during key embryonic oocyte developmental stages, including early events in meiosis. Summary Sentence Generation of a novel cre recombinase knockin to the Stra8 locus allows production of Stra8 and cre without affecting fertility.


Asunto(s)
Células Germinativas , Integrasas , Ratones , Masculino , Femenino , Animales , Células Germinativas/metabolismo , Integrasas/genética , Integrasas/metabolismo , Oocitos/metabolismo , Proteínas/metabolismo , Ratones Transgénicos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo
4.
Mol Reprod Dev ; 90(5): 275-286, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36966461

RESUMEN

Meiosis, a key step in spermatogenesis, is affected by many factors. Current studies have shown that long noncoding RNAs (lncRNAs) are potential factors regulating meiosis, and their regulatory mechanisms have received much attention. However, little research has been done on its regulatory mechanism in the spermatogenesis of roosters. Here, we found that lncRNA involved in meiosis and spermatogenesis (lncRNA-IMS) was involved in the regulation of Stra8 by gga-miR-31-5p and hindered the inhibition of Stra8 by gga-miR-31-5p. The acquisition and loss of function experiments demonstrated that lncRNA-IMS was involved in meiosis and spermatogenesis. In addition, we predicted and determined the core promoter region of lncRNA-IMS. Prediction of transcription factors, deletion/overexpression of binding sites, knockdown/overexpression of Jun, and dual-luciferase reporter analysis confirmed that Jun positively activated transcription of lncRNA-IMS. Our findings further enrich the TF-lncRNA-miRNA-mRNA regulatory network during male meiosis and provide new ideas for studying the molecular mechanism of meiosis and spermatogenesis in chicken spermatogonial stem cells.


Asunto(s)
Células Madre Germinales Adultas , Proteínas Aviares , Meiosis , MicroARNs , ARN Largo no Codificante , Animales , Masculino , Células Madre Germinales Adultas/metabolismo , Pollos/genética , Pollos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Aviares/metabolismo
5.
Mol Biol Rep ; 50(1): 631-640, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36371553

RESUMEN

BACKGROUND: Devising of an appropriate in vitro culture method for germ cells differentiation in the presence of soluble factors has attracted considerable attention, which results will provide new insight into reproductive biology. In this study, we compared the effects of forskolin, retinoic acid (RA) or granulosa cell-conditioned medium in the presence or absence of granulosa cell co-culturing on germ cell differentiation from embryonic stem cells (ESCs). METHODS AND RESULTS: Embryonic stem cells were differentiated using embryoid bodies (EBs) for 5 days, and then EB-derived cells were co-cultured with or without adult mouse granulosa cells using monolayer protocol and treated with 50 µM forskolin, 1 µM RA and 50% granulosa cell-conditioned medium for 4 days. Granulosa cell-conditioned medium significantly increased the levels of Scp3, Rec8, Mvh and Gdf9 expression in the granulosa cell co-culture method compared to untreated cells. A significant elevation of Stra8, Rec8 and Mvh was observed after treatment with RA in the absence of granulosa cells and there was no significant increase in the levels of expression of germ cell-specific genes after treatment with forskolin compared to control. Furthermore, forskolin and RA significantly increased viability and proliferation of germ-like cells, compared with granulosa cell-conditioned medium. CONCLUSIONS: Our study revealed that granulosa cell-conditioned medium and RA effectively can induce germ cell differentiation from ESCs, however combined application of granulosa cell-conditioned medium and co-culturing with granulosa cells had synergic effect on germ cell development in vitro as optimized protocol.


Asunto(s)
Células Germinativas , Tretinoina , Animales , Femenino , Ratones , Tretinoina/farmacología , Técnicas de Cocultivo , Colforsina/farmacología , Colforsina/metabolismo , Medios de Cultivo Condicionados/farmacología , Medios de Cultivo Condicionados/metabolismo , Diferenciación Celular , Células Cultivadas , Células Germinativas/metabolismo , Células de la Granulosa/metabolismo
6.
J Reprod Dev ; 69(3): 139-146, 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-36927827

RESUMEN

In mouse fetal gonads, germ cell development is accompanied by changes in cell cycle mode in response to external signals and intrinsic mechanisms of cells. During fetal development, male germ cells undergo G0/G1 arrest, while female germ cells exit the mitotic cell cycle and enter meiosis. In fetal testes, NANOS2 and CYP26B1 force germ cells to stay in G0/G1 arrest phase, preventing them from entering the meiotic cell cycle. In the fetal ovary, external signals, such as RA, BMP, and WNT, promote the competency of female germ cells to enter the meiotic cell cycle. MEIOSIN and STRA8 ensure the establishment of the meiotic cell cycle by activating meiotic genes, such that meiotic entry coincides with the S phase. This review discusses germ cell development from the viewpoint of cell cycle regulation and highlights the mechanism of the entry of germ cells into meiosis.


Asunto(s)
Células Germinativas , Tretinoina , Masculino , Femenino , Ratones , Animales , Meiosis , Testículo/metabolismo , Diferenciación Celular , Mamíferos/metabolismo
7.
Int J Mol Sci ; 23(20)2022 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-36293427

RESUMEN

Meiosis is the unique division of germ cells resulting in the recombination of the maternal and paternal genomes and the production of haploid gametes. In mammals, it begins during the fetal life in females and during puberty in males. In both cases, entering meiosis requires a timely switch from the mitotic to the meiotic cell cycle and the transition from a potential pluripotent status to meiotic differentiation. Revealing the molecular mechanisms underlying these interrelated processes represents the essence in understanding the beginning of meiosis. Meiosis facilitates diversity across individuals and acts as a fundamental driver of evolution. Major differences between sexes and among species complicate the understanding of how meiosis begins. Basic meiotic research is further hindered by a current lack of meiotic cell lines. This has been recently partly overcome with the use of primordial-germ-cell-like cells (PGCLCs) generated from pluripotent stem cells. Much of what we know about this process depends on data from model organisms, namely, the mouse; in mice, the process, however, appears to differ in many aspects from that in humans. Identifying the mechanisms and molecules controlling germ cells to enter meiosis has represented and still represents a major challenge for reproductive medicine. In fact, the proper execution of meiosis is essential for fertility, for maintaining the integrity of the genome, and for ensuring the normal development of the offspring. The main clinical consequences of meiotic defects are infertility and, probably, increased susceptibility to some types of germ-cell tumors. In the present work, we report and discuss data mainly concerning the beginning of meiosis in mammalian female germ cells, referring to such process in males only when pertinent. After a brief account of this process in mice and humans and an historical chronicle of the major hypotheses and progress in this topic, the most recent results are reviewed and discussed.


Asunto(s)
Meiosis , Células Madre Pluripotentes , Humanos , Masculino , Femenino , Ratones , Animales , Meiosis/genética , Células Germinativas/metabolismo , Diferenciación Celular , Mamíferos/genética
8.
Int J Mol Sci ; 23(22)2022 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-36430291

RESUMEN

Fertilization is a complex process that requires successive stages and culminates in the adhesion/fusion of gamete membranes. If the question of the involvement of oocyte integrins has been swept away by deletion experiments, that of the involvement of sperm integrins remains to be further characterized. In the present study, we addressed the question of the feasibility of sperm-oocyte adhesion/fusion and early implantation in the absence of sperm ß1 integrin. Males and females with ß1 integrin-depleted sperm and oocytes were mated, and fertilization outcome was monitored by a gestational ultrasound analysis. Results suggest that although the sperm ß1 integrin participates in gamete adhesion/fusion, it is dispensable for fertilization in mice. However, sperm- and/or oocyte-originated integrin ß1 is essential for post-implantation development. Redundancy phenomena could be at the origin of a compensatory expression or alternative dimerization pattern.


Asunto(s)
Integrina beta1 , Interacciones Espermatozoide-Óvulo , Femenino , Ratones , Masculino , Animales , Integrina beta1/genética , Integrina beta1/metabolismo , Semen/metabolismo , Oocitos/metabolismo , Espermatozoides/metabolismo , Fertilización , Integrinas/metabolismo
9.
J Cell Mol Med ; 25(1): 383-396, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33236849

RESUMEN

STRA8 (Stimulated by Retinoic Acid Gene 8) controls the crucial decision of germ cells to engage meiotic division up and down-regulating genes involved in the meiotic programme. It has been proven as an amplifier of genes involved in cell cycle control and chromosome events, however, how STRA8 functions as negative regulator are not well understood. In this study, we demonstrate that STRA8 can interact with itself and with other basic Helix-Loop-Helix (bHLH) transcription factors through its HLH domain and that this domain is important for its ability to negatively interfere with the Ebox-mediated transcriptional activity of bHLH transcription factors. Significantly, we show that STRA8 interacts with TCF3/E47, a class I bHLH transcription factors, and with SOHLH1, a gonadal-specific bHLH, in male germ cells obtained from prepuberal mouse testis. We demonstrated that STRA8, indirectly, is able to exert a negative control on the SOHLH1-dependent stimulation of c-KIT expression in late differentiating spermatogonia and preleptotene spermatocytes. Although part of this results were obtained only 'in vitro', they support the notion that STRA8 interacting with different transcription factors, besides its established role as 'amplifier' of meiotic programme, is able to finely modulate the balance between spermatogonia proliferation, differentiation and acquisition of meiotic competence.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Masculino , Unión Proteica , Proteínas Proto-Oncogénicas c-kit/genética
10.
Biol Reprod ; 105(2): 503-518, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-33959758

RESUMEN

Retinoic acid (RA) induces spermatogonial differentiation, but the mechanism by which it operates remains largely unknown. We developed a germ cell culture assay system to study genes involved in spermatogonial differentiation triggered by RA. Stimulated by RA 8 (Stra8), a RA-inducible gene, is indispensable for meiosis initiation, and its deletion results in a complete block of spermatogenesis at the pre-leptotene/zygotene stage. To interrogate the role of Stra8 in RA mediated differentiation of spermatogonia, we derived germ cell cultures from the neonatal testis of both wild type and Stra8 knock-out mice. We provide the first evidence that Stra8 plays a crucial role in modulating the responsiveness of undifferentiated spermatogonia to RA and facilitates transition to a differentiated state. Stra8-mediated differentiation is achieved through the downregulation of a large portfolio of genes and pathways, most notably including genes involved in the spermatogonial stem cell self-renewal process. We also report here for the first time the role of transcription elongation regulator-1 like (Tcerg1l) as a downstream effector of RA-induced spermatogonial differentiation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Embrión de Mamíferos/embriología , Ratones/genética , Espermatogonias , Factores de Elongación Transcripcional/genética , Tretinoina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Masculino , Ratones/embriología , Factores de Elongación Transcripcional/metabolismo
11.
Mol Reprod Dev ; 88(2): 128-140, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33400349

RESUMEN

Spermatogonial development is a key process during spermatogenesis to prepare germ cells to enter meiosis. While the initial point of spermatogonial differentiation is well-characterized, the development of spermatogonia from the onset of differentiation to the point of meiotic entry has not been well defined. Further, STRA8 is highly induced at the onset of spermatogonial development but its function in spermatogonia has not been defined. To better understand how STRA8 impacts spermatogonia, we performed RNA-sequencing in both wild-type and STRA8 knockout mice at multiple timepoints during retinoic acid (RA)-stimulated spermatogonial development. As expected, in spermatogonia from wild-type mice we found that steady-state levels of many transcripts that define undifferentiated progenitor cells were decreased while transcripts that define the differentiating spermatogonia were increased as a result of the actions of RA. However, the spermatogonia from STRA8 knockout mice displayed a muted RA response such that there were more transcripts typical of undifferentiated cells and fewer transcripts typical of differentiating cells following RA action. While spermatogonia from STRA8 knockout mice can ultimately form spermatocytes that fail to complete meiosis, it appears that the defect likely begins as a result of altered messenger RNA levels during spermatogonial differentiation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Regulación del Desarrollo de la Expresión Génica , Espermatogénesis/fisiología , Espermatogonias/crecimiento & desarrollo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Masculino , Meiosis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , RNA-Seq , Espermatogénesis/efectos de los fármacos , Espermatogénesis/genética , Transcripción Genética , Tretinoina/farmacología
12.
J Cell Physiol ; 235(3): 3033-3042, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31541472

RESUMEN

Promyelocytic leukaemia zinc finger (PLZF) is a key factor in inhibiting differentiation of spermatogonial progenitor cells (SPCs), but the underlying mechanisms are still largely unknown. In this study, the regulation of PLZF on Kit, Stra8, Sohlh2, and Dmrt1 (SPCs differentiation related genes) was investigated. We found some PLZF potential binding sites existed in the promoters of Kit, Stra8, Sohlh2, and Dmrt1. Additionally, the expressions of KIT, STRA8, SOHLH2, and DMRT1 were upregulated when PLZF was knockdown in SPCs. Furthermore, chromatin immunoprecipitation quantitative polymerase chain reaction revealed PLZF directly bound to the promoters of Kit, Stra8, Sohlh2, and Dmrt1. Besides, dual luciferase assay verified PLZF repressed those gene expressions. Collectively, our finding indicate that PLZF binds to the promoter regions of Kit, Stra8, Sohlh2, and Dmrt1 to regulate SPCs differentiation, which facilitate us to further understand the regulatory mechanism of PLZF in SPCs fates.


Asunto(s)
Células Madre Germinales Adultas/metabolismo , Diferenciación Celular/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Espermatogonias/metabolismo , Animales , Expresión Génica/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regulación hacia Arriba
13.
Genesis ; 57(7-8): e23327, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31313882

RESUMEN

In the embryonic gonads of mice, the genetic and epigenetic regulatory programs for germ cell sex specification and meiosis induction or suppression are intertwined. The quest for garnering comprehensive understanding of these programs has led to the emergence of retinoic acid (RA) as an important extrinsic factor, which regulates initiation of meiosis in female fetal germ cells that have attained a permissive epigenetic ground state. In contrast, germ cells in fetal testis are protected from the exposure to RA due to the activity of CYP26B1, an RA metabolizing enzyme, which is highly expressed in fetal testis. In this review, we provide an overview of the molecular mechanisms operating in fetal gonads of mice, which enable regulation of meiosis via RA signaling.


Asunto(s)
Gónadas/embriología , Meiosis , Tretinoina/metabolismo , Animales , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Ratones , Transducción de Señal
14.
J Exp Zool B Mol Dev Evol ; 332(7): 269-278, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31580014

RESUMEN

Our knowledge of mechanisms involved in the meiosis of chicken germ cells is very limited. In mammalian fetal ovaries, the onset of meiosis is dependent on retinoic acid and subsequent upregulation of the Stra8 gene. To clarify the mechanism of meiotic initiation in chicken germ cells, we investigated the role of Cyp26b1, a retinoic acid-degrading enzyme. The Cyp26b1-inhibitor, ketoconazole was used to treat the ex vivo-cultured stage 36 gonads/mesonephroi. Then, the progression of meiosis was studied by histological and immunohistochemical analysis and the level of the transcript for Stra8 was evaluated by a quantitative reverse transcription-polymerase chain reaction in individual ketoconazole-treated gonads after 6 days in culture. The results revealed that meiosis was induced in both testes and right ovary upon inhibition of Cyp26b1 in the ex vivo-cultured gonads, despite downregulation of Stra8 messenger RNA in the treated gonads. Also, meiosis was observed only when mesonephros was cultured alongside the left ovary. These findings demonstrate that in chicken, Stra8 is not the only factor for the entrance into meiosis, and Cyp26b1 and mesonephros play critical regulatory roles for the sex-specific timing of meiotic initiation in birds.


Asunto(s)
Células Germinativas/citología , Meiosis , Mesonefro , Ácido Retinoico 4-Hidroxilasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Diferenciación Celular , Embrión de Pollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Cetoconazol/farmacología , Masculino , Ovario/embriología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ácido Retinoico 4-Hidroxilasa/efectos de los fármacos , Testículo/embriología
15.
Endocr Regul ; 53(2): 93-99, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-31517623

RESUMEN

OBJECTIVE: Stem cell therapy, specifically, pre-induction of mesenchymal stem cells toward male germ-like cells may be useful in patients with azoospermia. The aim of this study was to evaluate in vitro differentiation of mouse bone marrow-derived mesenchymal stem cells (BMSCs) into male germ-like cells by indirect co-culture with testicular cells in the presence of bone morphogenetic protein 4 (BMP4). METHODS: Experimental groups included: control (mouse BMSCs), treatment group-1 (BMSCs treated with BMP4), treatment group-2 (indirect co-culture of BMSCs with mouse testicular cells in the presence of BMP4) and treatment group-3 (indirect co-culture of BMSCs with testicular cells). BMSCs-derived male germ-like cells were evaluated by the expression of Dazl, and Stra8 using RT-qPCR. RESULTS: Stra8 gene expression was significantly increased in the treatment group-2 and Dazl gene was significantly increased in the treatment group-1 compared to other groups. In conclusion, indirect co-culturing of BMSCs with testicular cells and BMP4 leads to the differentiation of BMSCs into male germ-like cells which express specific male germ-like genes. Testicular cells released factors that contributed to the differentiation of BMSCs into male germ progenitor cells. CONCLUSION: This study suggests that mesenchymal stem cells may be differentiated into male germ-like cells and therefore, may be a novel treatment option for men with azoospermia.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Testículo/citología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Células Cultivadas , Técnicas de Cocultivo/métodos , Células Germinativas/efectos de los fármacos , Células Germinativas/fisiología , Humanos , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Espermatozoides/fisiología
16.
Proc Natl Acad Sci U S A ; 112(18): E2347-56, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25902548

RESUMEN

Mammalian spermatogenesis--the transformation of stem cells into millions of haploid spermatozoa--is elaborately organized in time and space. We explored the underlying regulatory mechanisms by genetically and chemically perturbing spermatogenesis in vivo, focusing on spermatogonial differentiation, which begins a series of amplifying divisions, and meiotic initiation, which ends these divisions. We first found that, in mice lacking the retinoic acid (RA) target gene Stimulated by retinoic acid gene 8 (Stra8), undifferentiated spermatogonia accumulated in unusually high numbers as early as 10 d after birth, whereas differentiating spermatogonia were depleted. We thus conclude that Stra8, previously shown to be required for meiotic initiation, also promotes (but is not strictly required for) spermatogonial differentiation. Second, we found that injection of RA into wild-type adult males induced, independently, precocious spermatogonial differentiation and precocious meiotic initiation; thus, RA acts instructively on germ cells at both transitions. Third, the competencies of germ cells to undergo spermatogonial differentiation or meiotic initiation in response to RA were found to be distinct, periodic, and limited to particular seminiferous stages. Competencies for both transitions begin while RA levels are low, so that the germ cells respond as soon as RA levels rise. Together with other findings, our results demonstrate that periodic RA-STRA8 signaling intersects with periodic germ-cell competencies to regulate two distinct, cell-type-specific responses: spermatogonial differentiation and meiotic initiation. This simple mechanism, with one signal both starting and ending the amplifying divisions, contributes to the prodigious output of spermatozoa and to the elaborate organization of spermatogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Células Germinativas/citología , Espermatogénesis , Tretinoina/química , Animales , Diferenciación Celular , Proliferación Celular , Cruzamientos Genéticos , Masculino , Meiosis , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Espermatogonias/citología , Espermatozoides/citología , Testículo/metabolismo
17.
Andrologia ; 50(1)2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28488736

RESUMEN

Rapamycin (mTOR inhibitor) has been reported to have negative effect on human male gonadal function. Previously, we showed that mTOR signalling molecules are expressed during early spermatogenesis in mice. The objective of this study was to investigate the role of mTOR signalling in meiosis both during the first wave of spermatogenesis and also during adult spermatogenesis. Day 5 post-partum mice were administered rapamycin and retinoic acid (RA; a Stra8 activator), and expression of p-p70S6K and Stra8 proteins was evaluated. p-p70S6K and Stra8 protein expressions decreased in post-natal testes after rapamycin treatment. Stra8 protein expression increased after RA and rapamycin+RA administrations in post-natal testes. In adult mice, rapamycin was administrated for 1 or 4 weeks. Morphological analysis for testicular damage and TUNEL assay was performed. After rapamycin administration, germ cell loss increased in adult testes. Ultrastructural analysis revealed disorganised testicular morphology and vacuolisation. The number of apoptotic germ cells increased after 4 weeks rapamycin administration. Stra8 and Dmc1 expressions decreased in 4 weeks rapamycin group, whereas Sycp3 and VASA expression did not change. Our findings suggest that mTOR pathway has an important role in meiotic progress of male germ cells both during first wave of spermatogenesis and in adult mice.


Asunto(s)
Meiosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Testículo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Sirolimus/farmacología , Espermatogénesis/efectos de los fármacos , Testículo/metabolismo , Tretinoina/farmacología
18.
J Cell Biochem ; 118(12): 4844-4853, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28543764

RESUMEN

Stra8 (stimulated by retinoic acid gene 8) is a specific gene that is expressed in mammalian germ cells during transition from mitosis to meiosis and plays a key role in the initiation of meiosis in mammals and birds. So, the evaluation of the Stra8 pathway in cSSCs may provide a deeper insight into mammalian spermatogenesis. miRNA was also an important regulating factor for meiosis of SSCs. However, there is currently no data indicating that miRNA regulate the meiosis of SSCs via Stra8. Here, we predicted the prospective miRNA targeting to Stra8 using the online Bioinformatics database-Targetscan, and performed an analysis of the dual-luciferase recombinant vector, pGL3-CMV-LUC-MCS-Stra8-3'UTR. miR-31 mimics (miR-31m), miR-31 inhibitors (miR-31i), Control (NC, scrambled oligonucleotides transfection) were transfected into cSSCs; Stra8 and miRNA were analyzed by RT-qPCR, immunofluorescence, and Western blot. The detection of haploid was conducted by flow cytometry. The results showed that miR-31 regulates meiosis of cSSCs via targeting Stra8 in vitro and in vivo. Our study identifies a new regulatory pathway that miR-31 targets Stra8 and inhibits spermatogenesis. J. Cell. Biochem. 118: 4844-4853, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Madre Germinales Adultas/metabolismo , Proteínas Aviares/metabolismo , Meiosis/fisiología , MicroARNs/metabolismo , Espermatogénesis/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Células Madre Germinales Adultas/citología , Animales , Proteínas Aviares/genética , Embrión de Pollo , Pollos , Masculino , MicroARNs/genética
19.
BMC Genomics ; 17: 368, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27189481

RESUMEN

With more than 30,000 species, ray-finned fish represent approximately half of vertebrates. The evolution of ray-finned fish was impacted by several whole genome duplication (WGD) events including a teleost-specific WGD event (TGD) that occurred at the root of the teleost lineage about 350 million years ago (Mya) and more recent WGD events in salmonids, carps, suckers and others. In plants and animals, WGD events are associated with adaptive radiations and evolutionary innovations. WGD-spurred innovation may be especially relevant in the case of teleost fish, which colonized a wide diversity of habitats on earth, including many extreme environments. Fish biodiversity, the use of fish models for human medicine and ecological studies, and the importance of fish in human nutrition, fuel an important need for the characterization of gene expression repertoires and corresponding evolutionary histories of ray-finned fish genes. To this aim, we performed transcriptome analyses and developed the PhyloFish database to provide (i) de novo assembled gene repertoires in 23 different ray-finned fish species including two holosteans (i.e. a group that diverged from teleosts before TGD) and 21 teleosts (including six salmonids), and (ii) gene expression levels in ten different tissues and organs (and embryos for many) in the same species. This resource was generated using a common deep RNA sequencing protocol to obtain the most exhaustive gene repertoire possible in each species that allows between-species comparisons to study the evolution of gene expression in different lineages. The PhyloFish database described here can be accessed and searched using RNAbrowse, a simple and efficient solution to give access to RNA-seq de novo assembled transcripts.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Evolución Molecular , Peces/genética , Duplicación de Gen , Expresión Génica , Genoma , Animales , Biología Computacional/métodos , Peces/clasificación , Perfilación de la Expresión Génica , Filogenia , Transcriptoma , Navegador Web
20.
Biochem Biophys Res Commun ; 480(4): 635-640, 2016 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-27794480

RESUMEN

Germline stem cells (GSCs) are attractive biological models because of their strict control on pluripotency gene expression, and their potential for huge epigenetic changes in a short period of time. Few data exists on the cooperative impact of GSC-specific genes on differentiated cells. In this study, we over-expressed 3 GSC-specific markers, STELLA, OCT4 and NANOS2, collectively designated as (SON), using the novel polycistronic lentiviral gene construct FUM-FD, in HEK293T cells and evaluated promoter activity of the Stra8 GSC marker gene We could show that HEK293T cells expressed pluripotency and GSC markers following ectopic expression of the SON genes. We also found induction of pluripotency markers after serum starvation in non-transduced HEK293T cells. Expression profiling of SON-expressing and serum-starved cells at mRNA and protein level showed the potential of SON factors and serum starvation in the induction of ESRRB, NANOG, OCT4 and REX1 expression. Additionally, the data indicated that the mouse Stra8 promoter could only be activated in a subpopulation of HEK293T cells, regardless of SON gene expression. We conclude that heterogeneous population of the HEK293T cells might be easily shifted towards expression of the pluripotency markers by ectopic expression of the SON factors or by growth in serum depleted media.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Células HEK293/citología , Células HEK293/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Diferenciación Celular/fisiología , Proteínas Cromosómicas no Histona , Humanos , Células Madre Pluripotentes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA