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












Base de datos
Intervalo de año de publicación
1.
Dev Biol ; 509: 11-27, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38311163

RESUMEN

Undifferentiated spermatogonia are composed of a heterogeneous cell population including spermatogonial stem cells (SSCs). Molecular mechanisms underlying the regulation of various spermatogonial cohorts during their self-renewal and differentiation are largely unclear. Here we show that AKT1S1, an AKT substrate and inhibitor of mTORC1, regulates the homeostasis of undifferentiated spermatogonia. Although deletion of Akt1s1 in mouse appears not grossly affecting steady-state spermatogenesis and male mice are fertile, the subset of differentiation-primed OCT4+ spermatogonia decreased significantly, whereas self-renewing GFRα1+ and proliferating PLZF+ spermatogonia were sustained. Both neonatal prospermatogonia and the first wave spermatogenesis were greatly reduced in Akt1s1-/- mice. Further analyses suggest that OCT4+ spermatogonia in Akt1s1-/- mice possess altered PI3K/AKT-mTORC1 signaling, gene expression and carbohydrate metabolism, leading to their functionally compromised developmental potential. Collectively, these results revealed an important role of AKT1S1 in mediating the stage-specific signals that regulate the self-renewal and differentiation of spermatogonia during mouse spermatogenesis.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Espermatogonias , Masculino , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Testículo/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Espermatogénesis/genética , Diferenciación Celular/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
2.
Elife ; 122023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37096870

RESUMEN

Spermatogenesis depends on an orchestrated series of developing events in germ cells and full maturation of the somatic microenvironment. To date, the majority of efforts to study cellular heterogeneity in testis has been focused on single-cell gene expression rather than the chromatin landscape shaping gene expression. To advance our understanding of the regulatory programs underlying testicular cell types, we analyzed single-cell chromatin accessibility profiles in more than 25,000 cells from mouse developing testis. We showed that single-cell sequencing assay for transposase-accessible chromatin (scATAC-Seq) allowed us to deconvolve distinct cell populations and identify cis-regulatory elements (CREs) underlying cell-type specification. We identified sets of transcription factors associated with cell type-specific accessibility, revealing novel regulators of cell fate specification and maintenance. Pseudotime reconstruction revealed detailed regulatory dynamics coordinating the sequential developmental progressions of germ cells and somatic cells. This high-resolution dataset also unveiled previously unreported subpopulations within both the Sertoli and Leydig cell groups. Further, we defined candidate target cell types and genes of several genome-wide association study (GWAS) signals, including those associated with testosterone levels and coronary artery disease. Collectively, our data provide a blueprint of the 'regulon' of the mouse male germline and supporting somatic cells.


Asunto(s)
Cromatina , Testículo , Masculino , Embarazo , Femenino , Animales , Ratones , Cromatina/metabolismo , Testículo/metabolismo , Estudio de Asociación del Genoma Completo , Factores de Transcripción/metabolismo , Espermatogénesis/genética , Análisis de la Célula Individual
3.
Yi Chuan ; 44(7): 591-608, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35858771

RESUMEN

Spermatogonial stem cells (SSCs) are adult stem cells in the testis of male animals and have the ability in self-renewal and differentiation. SSCs are derived from primordial germ cells (PGCs) that are mitotically arrested in the embryo before birth. Following the birth of the animal, PGCs resume mitosis and migrate from the centre of the seminiferous tubules to the basement membrane. The descendent of PGCs (also called gonocytes) establish stable SSC colonies in about a week postnatally in order to support the life-long spermatogenesis. Whether SSCs at different developmental stages differ in their molecular and cellular characteristics is currently unclear. In the presented study, we conducted bioinformatics analyses using transcriptomics data established previously in the laboratory on OCT4 (encoded by the pluripotent gene Pou5f1) expressing SSCs from the neonatal (3 days-post-partum, 3-dpp), juvenile (7-dpp) and adult (2~3-month) mice, including screen of differentially expressed genes (DEGs), protein-protein interaction (PPI) network analysis of DEGs and clustering of sub-networks from PPI. GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) analyses were also performed on clustered sub-networks of the PPI. In addition, all genes were analyzed using GSEA (gene set enrichment analysis) based on GO, KEGG and HALLMARK gene sets. The results showed that SSCs have a large number of DEGs among OCT4-positive SSCs from neonatal, juvenile and adult mice. The distinguishable biological functions encoded by these DEGs include biosynthesis and energy metabolism, immune response, cell junction and expression of migration and cell differentiation-related genes. Significant changes in the cell membrane composition of OCT4-positive SSCs may not only cause hypersensitive immune reactions but also affect the cell-cell contact and responses to secreted cytokines in the extracellular environment. The results also suggest that OCT4-positive SSCs may shift metabolic state from oxidative phosphorylation to glycolysis and significantly reduce the transcription of genes related to ribosome formation during aging. These results provide new clues for future research on the regulatory mechanisms of male germline stem cell development, growth and aging.


Asunto(s)
Perfilación de la Expresión Génica , Espermatogonias , Animales , Diferenciación Celular , Masculino , Ratones , Espermatogénesis/genética , Células Madre , Testículo
4.
STAR Protoc ; 3(2): 101398, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35600921

RESUMEN

Spermatogenesis generates heterologous cell populations which, if not distinguished clearly, often hinder mechanistic and etiological studies. Here, we present a protocol to identify and isolate populations of mouse spermatogenic cells, including spermatogonial stem cells (SSCs), spermatocytes, and haploid spermatids. We also describe absolute quantification of mRNA copy numbers in SSCs. The isolated cells can be used for analyzing nascent protein synthesis and protein degradation, two main events that maintain cellular proteostasis important for healthy and long-term production of male gametes. For complete details on the use and execution of this protocol, please refer to Zou et al. (2021).


Asunto(s)
Células Madre Germinales Adultas , Espermátides , Animales , Haploidia , Masculino , Ratones , Espermátides/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/genética
5.
Genes (Basel) ; 12(12)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34946890

RESUMEN

cAMP-dependent protein kinase (PKA) signaling plays various roles during mammalian spermatogenesis, ranging from the regulation of gene expression to the modulation of sperm motility. However, the molecular mechanisms that govern the multifaceted functions of PKA during spermatogenesis remain largely unclear. We previously found that PKA regulatory subunit I α (RIα) and catalytic subunit α (Cα) co-sediment with polyribosomal fractions of mouse testis lysate on sucrose gradient and the stimulation of PKA activity facilitates protein synthesis in post-meiotic elongating spermatids, indicating that type I PKA is intricately associated with protein translation machinery and regulates protein synthesis during mouse spermiogenesis. Since PKA activity is often regulated by interacting proteins that form complexes with its regulatory subunits, the identification of PKA-RIα interacting proteins in post-meiotic spermatogenic cells will facilitate our understanding of its regulatory roles in protein synthesis and spermiogenesis. In the present study, we applied a yeast two-hybrid screen to identify PKA-Riα-binding proteins using a cDNA library generated from mouse round and elongating spermatids. Numerous proteins were found to potentially interact with PKA-RIα, including proteostasis modulators, metabolic enzymes, cytoskeletal regulators, and mitochondrial proteins, many of which are specifically expressed in testes. Consistently, the examination of MENA (mouse ENA/VASP homolog) in developing mouse testes suggested that post-meiotic spermatogenic cells express a short isoform of MENA that interacts with PKA-RIα in yeast two-hybrid assay. The identification of PKA-RIα interacting proteins provides us solid basis to further explore how PKA signaling regulates protein synthesis and cellular morphogenesis during mouse spermatogenesis.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Espermatogénesis , Animales , Biblioteca de Genes , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Isoformas de Proteínas , Espermatogénesis/genética , Técnicas del Sistema de Dos Híbridos
6.
iScience ; 24(12): 103396, 2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34825148

RESUMEN

Maintaining proteostasis is important for animal development. How proteostasis influences spermatogenesis that generates male gametes, spermatozoa, is not clear. We show that testis-specific paralog of ribosomal large subunit protein RPL39, RPL39L, is required for mouse spermatogenesis. Deletion of Rpl39l in mouse caused reduced proliferation of spermatogonial stem cells, malformed sperm mitochondria and flagella, leading to sub-fertility in males. Biochemical analyses revealed that lack of RPL39L deteriorated protein synthesis and protein quality control in spermatogenic cells, partly due to reduced biogenesis of ribosomal subunits and ribosome homeostasis. RPL39/RPL39L is likely assembled into ribosomes via H/ACA domain containing NOP10 complex early in ribosome biogenesis pathway. Furthermore, Rpl39l null mice exhibited compromised regenerative spermatogenesis after chemical insult and early degenerative spermatogenesis in aging mice. These data demonstrate that maintaining proteostasis is important for spermatogenesis, of which ribosome homeostasis maintained by ribosomal proteins coordinates translation machinery to the regulation of cellular growth.

7.
Int J Biochem Cell Biol ; 139: 106070, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34428590

RESUMEN

Accumulating evidences suggest that the composition and functional roles of ribosomes are heterogeneous in cells, partly due to the temporal-spatial expression of paralogous ribosomal proteins (RPs), of which functional relationships remain largely unexplored. In mouse, the X chromosome-linked RPL39 and its male germline specific paralog RPL39L are thought to express mutually exclusively due to the meiotic sex chromosome inactivation, hinders the understanding of their functional relationships. In the present study, we investigated the expression and functional relations of Rpl39 and Rpl39l in a proliferative mouse cell line, in which both genes are expressed simultaneously, with the expression level of Rpl39 higher than that of Rpl39l. Disruption of Rpl39 via CRISPR/Cas9 method caused decreased cell proliferation, nascent protein synthesis and altered mitochondrial functions, whereas double mutations of Rpl39 and Rpl39l augmented these phenotypes, suggesting that both RPs contribute to the cellular physiology. Consistently, overexpression of Rpl39, Rpl39l or an alanine mutant of RPL39, rescued cell proliferation similarly in Rpl39-/-::Rpl39l-/- dual gene null cells. Deletion of Rpl39l induced compensatory expression of Rpl39, rendering the deleterious effects of Rpl39l mutation. Supporting this, Rpl39l mutation was more detrimental to cells under a low serum condition, under which the compensatory expression of Rpl39 was inhibited. Moreover, the low serum condition induced expression of both genes, suggesting that they possess stress responsive roles. Taken together, these data indicate that both RPL39 and RPL39L influence cell proliferation via protein synthesis and mitochondrial functions, suggesting a link between protein translation and cellular metabolism through these ribosomal protein paralogs.


Asunto(s)
Ribosomas , Animales , Proliferación Celular , Ratones , Mitocondrias , Biosíntesis de Proteínas
8.
PLoS Genet ; 17(7): e1009369, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34237055

RESUMEN

Spermatogonial stem cells (SSC), the foundation of spermatogenesis and male fertility, possess lifelong self-renewal activity. Aging leads to the decline in stem cell function and increased risk of paternal age-related genetic diseases. In the present study, we performed a comparative genomic analysis of mouse SSC-enriched undifferentiated spermatogonia (Oct4-GFP+/KIT-) and differentiating progenitors (Oct4-GFP+/KIT+) isolated from young and aged testes. Our transcriptome data revealed enormous complexity of expressed coding and non-coding RNAs and alternative splicing regulation during SSC differentiation. Further comparison between young and aged undifferentiated spermatogonia suggested these differentiation programs were affected by aging. We identified aberrant expression of genes associated with meiosis and TGF-ß signaling, alteration in alternative splicing regulation and differential expression of specific lncRNAs such as Fendrr. Epigenetic profiling revealed reduced H3K27me3 deposition at numerous pro-differentiation genes during SSC differentiation as well as aberrant H3K27me3 distribution at genes in Wnt and TGF-ß signaling upon aging. Finally, aged undifferentiated spermatogonia exhibited gene body hypomethylation, which is accompanied by an elevated 5hmC level. We believe this in-depth molecular analysis will serve as a reference for future analysis of SSC aging.


Asunto(s)
Células Madre Germinales Adultas/citología , Células Madre Germinales Adultas/fisiología , Envejecimiento/fisiología , Epigenoma , 5-Metilcitosina/metabolismo , Envejecimiento/genética , Empalme Alternativo , Animales , Diferenciación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Lisina/genética , Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Largo no Codificante/genética , Testículo/citología
9.
Cell Prolif ; 54(5): e13000, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33666296

RESUMEN

OBJECTIVES: Mammalian spermatogenesis is a biological process of male gamete formation. Gonocytes are the only precursors of spermatogonial stem cells (SSCs) which develop into mature spermatozoa. DDX5 is one of DEAD-box RNA helicases and expresses in male germ cells, suggesting that Ddx5 plays important functions during spermatogenesis. Here, we explore the functions of Ddx5 in regulating the specification of gonocytes. MATERIALS AND METHODS: Germ cell-specific Ddx5 knockout (Ddx5-/- ) mice were generated. The morphology of testes and epididymides and fertility in both wild-type and Ddx5-/- mice were analysed. Single-cell RNA sequencing (scRNA-seq) was used to profile the transcriptome in testes from wild-type and Ddx5-/- mice at postnatal day (P) 2. Dysregulated genes were validated by single-cell qRT-PCR and immunofluorescent staining. RESULTS: In male mice, Ddx5 was expressed in germ cells at different stages of development. Germ cell-specific Ddx5 knockout adult male mice were sterile due to completely devoid of germ cells. Male germ cells gradually disappeared in Ddx5-/- mice from E18.5 to P6. Single-cell transcriptome analysis showed that genes involved in cell cycle and glial cell line-derived neurotrophic factor (GDNF) pathway were significantly decreased in Ddx5-deficient gonocytes. Notably, Ddx5 ablation impeded the proliferation of gonocytes. CONCLUSIONS: Our study reveals the critical roles of Ddx5 in fate determination of gonocytes, offering a novel insight into the pathogenesis of male sterility.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Células Germinativas/metabolismo , Animales , Animales Recién Nacidos , ARN Helicasas DEAD-box/genética , Regulación del Desarrollo de la Expresión Génica , Genotipo , Células Germinativas/citología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Infertilidad/metabolismo , Infertilidad/patología , Masculino , Ratones , Ratones Noqueados , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Testículo/metabolismo , Testículo/patología
10.
Development ; 147(2)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31969357

RESUMEN

The development and maintenance of the correct morphology of sperm is important for their functions. Cellular morphogenesis of sperm occurs during the post-meiotic developmental stage; however, little is known about what coordinates this process. In the present study, we investigated the role of A-kinase anchoring protein 3 (AKAP3) during mouse spermiogenesis, using both mouse genetics and proteomics. It was found that AKAP3 is essential for the formation of the specific subcellular structure of the sperm flagellum, motility of sperm and male fertility. Additionally, lack of AKAP3 caused global changes of the sperm proteome and mislocalization of sperm proteins, including accumulation of RNA metabolism and translation factors and displacement of PKA subunits in mature sperm, which may underlie misregulated PKA activity and immotility in sperm. Interestingly, sperm lacking a complete fibrous sheath from both Akap3 and Akap4 null mice accumulated F-actin filaments and morphological defects during post-testicular maturation in the epididymis. These results suggest that the subcellular structures of sperm could be formed via independent pathways, and elucidate the roles of AKAP3 during the coordinated synthesis and organization of the sperm proteome and sperm morphology.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Infertilidad Masculina/metabolismo , Espermatozoides/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Secuencia de Bases , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Epidídimo/metabolismo , Eliminación de Gen , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Proteoma/metabolismo , Transducción de Señal , Espermatozoides/anomalías , Espermatozoides/patología , Espermatozoides/ultraestructura , Fracciones Subcelulares/metabolismo
11.
Stem Cells Dev ; 28(20): 1365-1375, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31580778

RESUMEN

Glaucoma is characterized by retinal ganglion cell (RGC) degeneration and is the second leading cause of blindness worldwide. However, current treatments such as eye drop or surgery have limitations and do not target the loss of RGC. Regenerative therapy using embryonic stem cells (ESCs) holds a promising option, but ethical concern hinders clinical applications on human subjects. In this study, we employed spermatogonial stem cells (SSCs) as an alternative source of ESCs for cell-based regenerative therapy in mouse glaucoma model. We generated functional RGCs from SSCs with a two-step protocol without applying viral transfection or chemical induction. SSCs were first dedifferentiated to embryonic stem-like cells (SSC-ESCs) that resemble ESCs in morphology, gene expression signatures, and stem cell properties. The SSC-ESCs then differentiated toward retinal lineages. We showed SSC-ESC-derived retinal cells expressed RGC-specific marker Brn3b and functioned as bona fide RGCs. To allow in vivo RGC tracing, Brn3b-EGFP reporter SSC-ESCs were generated and the derived RGCs were subsequently transplanted into the retina of glaucoma mouse models by intravitreal injection. We demonstrated that the transplanted RGCs could survive in host retina for at least 10 days after transplantation. SSC-ESC-derived RGCs can thus potentially be a novel alternative to replace the damaged RGCs in glaucomatous retina.


Asunto(s)
Células Madre Germinales Adultas/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Glaucoma/terapia , Células Ganglionares de la Retina/trasplante , Células Madre Germinales Adultas/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Expresión Génica , Genes Reporteros , Glaucoma/inducido químicamente , Glaucoma/genética , Glaucoma/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , N-Metilaspartato/administración & dosificación , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Cultivo Primario de Células , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Testículo/citología , Testículo/metabolismo , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo
12.
Exp Cell Res ; 373(1-2): 71-79, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30266657

RESUMEN

During spermatogenesis, a group of undifferentiated spermatogonia undergoes an essential transition to a differentiating stage, which involves gain of Kit receptor. In the current study, we showed that a small non-coding RNA, miRNA-26b could induce transition from Kit- to Kit+ and inhibit proliferation of spermatogonia. A key transcriptional factor for undifferentiated spermatogonia, Plzf, was proven as a direct target of miR-26b. When undifferentiated spermatogonia were treated with Retinoic acid (RA), miR-26b was increased, further promoting RA-induced differentiation of spermatogonia. In addition, miR-26b could repress 5-hydroxymethylcytosine (5hmC) via repression of Tet3 in spermatogonia. These findings demonstrate that miR-26b might play a role in promoting the transition from Kit- to Kit+ SSCs.


Asunto(s)
MicroARNs/fisiología , Espermatogénesis , Espermatogonias/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , Apoptosis , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Masculino , Ratones , MicroARNs/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-kit/análisis , Espermatogonias/citología , Espermatogonias/efectos de los fármacos , Tretinoina/farmacología
13.
Biol Reprod ; 99(4): 773-788, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29701755

RESUMEN

Translational regulation plays a central role during post-meiotic development of male germ cells. Previous studies suggested that P-element induced wimpy testis like 1 (PIWIL1), a PIWI-interacting RNA (piRNA) binding protein that is critical for sperm development, participates in the maintenance and translational regulation of post-meiotic mRNAs in haploid spermatids. However, how PIWIL1 regulates protein translation remains largely unclear. Using biochemical assays, we show here that PIWIL1 utilizes different domains to interact with post-meiotic mRNAs and Poly-A binding protein cytoplasmic 1 (PABPC1), a general protein translation regulator. PIWIL1 binds 3'-untranslated regions (3'-UTRs) of several spermiogenic mRNAs via its N-terminal domain, whereas its interactions with PABPC1 are mediated through its N- and C-terminal domains in an RNA-dependent manner. Using a heterologous cell system, we analyzed its effects on protein translation via luciferase reporter assay and sucrose gradient sedimentation. It was found that PIWIL1 augments protein translation with PABPC1 in the presence of 3'-UTRs of post-meiotic mRNAs. While both the N-terminal RNA recognition motif (RRM) domain and the central linker region of PABPC1 stimulate translation, only the PIWI Argonaute and Zwille (PAZ) domain of PIWIL1 positively affects translation of reporter mRNAs. Interestingly, the PAZ domain was found absent from polysomal fractions, in contrast to the N- and C-terminal domains of PIWIL1. Taken together, the results suggest that PIWIL1 interacts with various partners using different domains and participates in translational regulation partly through 3'-UTRs. It will be of interest to further explore how PIWIL1 elicit its versatile functions, including translational regulation of post-meiotic mRNAs through intrinsic structural changes and extrinsic signals during mouse spermiogenesis under more physiological settings.


Asunto(s)
Proteínas Argonautas/química , Proteínas Argonautas/metabolismo , Proteína I de Unión a Poli(A)/química , Proteína I de Unión a Poli(A)/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Espermatogénesis/genética , Regiones no Traducidas 3' , Animales , Proteínas Argonautas/genética , Células HEK293 , Humanos , Masculino , Meiosis/genética , Ratones , Ratones Endogámicos C57BL , Proteína I de Unión a Poli(A)/genética , Biosíntesis de Proteínas , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Testículo/metabolismo
14.
Cell Regen ; 5: 1, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26839690

RESUMEN

Adult stem cells that reside in particular types of tissues are responsible for tissue homeostasis and regeneration. Cellular functions of adult stem cells are intricately related to the gene expression programs in those cells. Past research has demonstrated that regulation of gene expression at the transcriptional level can decisively alter cell fate of stem cells. However, cellular contents of mRNAs are sometimes not equivalent to proteins, the functional units of cells. It is increasingly realized that post-transcriptional and translational regulation of gene expression are also fundamental for stem cell functions. Compared to differentiated somatic cells, effects on cellular status manifested by varied expression of RNA-binding proteins and global protein synthesis have been demonstrated in several stem cell systems. Through the cooperation of both cis-elements of mRNAs and trans-acting RNA-binding proteins that are intimately associated with them, regulation of localization, stability, and translational status of mRNAs directly influences the self-renewal and differentiation of stem cells. Previous studies have uncovered some of the molecular mechanisms that underlie the functions of RNA-binding proteins in stem cells in invertebrate species. However, their roles in adult stem cells in mammals are just beginning to be unveiled. This review highlights some of the RNA-binding proteins that play important functions during the maintenance and differentiation of mouse male germline stem cells, the adult stem cells in the male reproductive organ.

15.
Reproduction ; 151(4): 351-67, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26762401

RESUMEN

Cell lineage determination during early embryogenesis has profound effects on adult animal development. Pre-patterning of embryos, such as that of Drosophila and Caenorhabditis elegans, is driven by asymmetrically localized maternal or zygotic factors, including mRNA species and RNA binding proteins. However, it is not clear how mammalian early embryogenesis is regulated and what the early cell fate determinants are. Here we show that, in mouse, mitochondrial ribosomal RNAs (mtrRNAs) are differentially distributed between 2-cell sister blastomeres. This distribution pattern is not related to the overall quantity or activity of mitochondria which appears equal between 2-cell sister blastomeres. Like in lower species, 16S mtrRNA is found to localize in the cytoplasm outside of mitochondria in mouse 2-cell embryos. Alterations of 16S mtrRNA levels in one of the 2-cell sister blastomere via microinjection of either sense or anti-sense RNAs drive its progeny into different cell lineages in blastocyst. These results indicate that mtrRNAs are differentially distributed among embryonic cells at the beginning of embryogenesis in mouse and they are functionally involved in the regulation of cell lineage allocations in blastocyst, suggesting an underlying molecular mechanism that regulates pre-implantation embryogenesis in mouse.


Asunto(s)
Blastocisto/citología , Blastómeros/citología , Linaje de la Célula/genética , Embrión de Mamíferos/citología , Desarrollo Embrionario/genética , ARN/genética , Animales , Blastocisto/metabolismo , Blastómeros/metabolismo , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mitocondrial , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Mol Reprod Dev ; 81(7): 595-607, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24687590

RESUMEN

cAMP-dependent protein kinase A (PKA) plays important regulatory roles during mouse spermatogenesis. PKA-mediated signaling has been shown to regulate gene expression, chromatin condensation, capacitation, and motility during sperm development and behavior, although how PKA is regulated in spatiotemporal manners during spermatogenesis is not fully understood. In the present study, we found that PKA subunit isoforms are expressed and localized differently in meiotic and post-meiotic mouse spermatogenic cells. Regulatory subunit I alpha (RIα) is expressed in spermatocytes and round spermatids, where it is localized diffusely throughout the cytoplasm of cells. During late spermiogenesis, RIα abundance gradually decreases. On the other hand, RIIα is expressed constantly throughout meiotic and post-meiotic stages, and is associated with cytoskeletal structures. Among several A kinase anchoring proteins (AKAPs) expressed in the testis, sperm-specific AKAP3 can be found in the cytoplasm of elongating spermatids and interacts with RIα, as demonstrated by both in vivo and in vitro experiments. In mature sperm, AKAP3 is exclusively found in the principal piece of the flagellum, coincident with only RIIα. Mutagenesis experiments further showed that the preferential interactions of AKAP3 with PKA regulatory subunits are mediated by two highly conserved amphipathic peptides located in the N-terminal region of AKAP3. Thus, AKAP3 is a dual-specificity molecule that modulates PKA isotypes in a spatiotemporal manner during mouse spermatogenesis.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Espermátides/química , Espermátides/metabolismo , Proteínas de Anclaje a la Quinasa A/análisis , Proteínas de Anclaje a la Quinasa A/química , Secuencia de Aminoácidos , Animales , AMP Cíclico/química , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/química , Células HEK293 , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Alineación de Secuencia
17.
Biol Reprod ; 90(6): 119, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24648398

RESUMEN

Mammalian spermatogenesis is regulated by coordinated gene expression in a spatiotemporal manner. The spatiotemporal regulation of major sperm proteins plays important roles during normal development of the male gamete, of which the underlying molecular mechanisms are poorly understood. A-kinase anchoring protein 3 (AKAP3) is one of the major components of the fibrous sheath of the sperm tail that is formed during spermiogenesis. In the present study, we analyzed the expression of sperm-specific Akap3 and the potential regulatory factors of its protein synthesis during mouse spermiogenesis. Results showed that the transcription of Akap3 precedes its protein synthesis by about 2 wk. Nascent AKAP3 was found to form protein complex with PKA and RNA binding proteins (RBPs), including PIWIL1, PABPC1, and NONO, as revealed by coimmunoprecipitation and protein mass spectrometry. RNA electrophoretic gel mobility shift assay showed that these RBPs bind sperm-specific mRNAs, of which proteins are synthesized during the elongating stage of spermiogenesis. Biochemical and cell biological experiments demonstrated that PIWIL1, PABPC1, and NONO interact with each other and colocalize in spermatids' RNA granule, the chromatoid body. In addition, NONO was found in extracytoplasmic granules in round spermatids, whereas PIWIL1 and PABPC1 were diffusely localized in cytoplasm of elongating spermatids, indicating their participation at different steps of mRNA metabolism during spermatogenesis. Interestingly, type I PKA subunits colocalize with PIWIL1 and PABPC1 in the cytoplasm of elongating spermatids and cosediment with the RBPs in polysomal fractions on sucrose gradients. Further biochemical analyses revealed that activation of PKA positively regulates AKAP3 protein synthesis without changing its mRNA level in elongating spermatids. Taken together, these results indicate that PKA signaling directly participates in the regulation of protein translation in postmeiotic male germ cells, underscoring molecular mechanisms that regulate protein synthesis during mouse spermiogenesis.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/fisiología , Espermátides/fisiología , Espermatogénesis/fisiología , Proteínas de Anclaje a la Quinasa A/metabolismo , Animales , Proteínas Argonautas/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Proteínas de Unión al ADN/metabolismo , Masculino , Ratones Endogámicos , Proteínas de Unión a Poli(A)/metabolismo , Biosíntesis de Proteínas/fisiología , Procesamiento Postranscripcional del ARN/fisiología , ARN Mensajero/metabolismo , Cola del Espermatozoide/fisiología , Espermátides/citología
18.
Gene Expr Patterns ; 14(1): 30-41, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24140495

RESUMEN

Nucleosomes are basic chromatin structural units that are formed by DNA sequences wrapping around histones. Global chromatin states in different cell types are specified by combinatorial effects of post-translational modifications of histones and the expression of histone variants. During mouse spermatogenesis, spermatogonial stem cells (SSCs) self-renew while undergo differentiation, events that occur in the company of constant re-modeling of chromatin structures. Previous studies have shown that testes contain highly expressed or specific histone variants to facilitate these epigenetic modifications. However, mechanisms of regulating the epigenetic changes and the specific histone compositions of spermatogenic cells are not fully understood. Using real time quantitative RT-PCR, we examined the dynamic expression of replication-dependent histone genes in post-natal mouse testes. It was found that distinct sets of histone genes are expressed in various spermatogenic cells at different stages during spermatogenesis. While gonocyte-enriched testes from mice at 2-dpp (days post partum) express pre-dominantly thirteen histone variant genes, SSC-stage testes at 9-dpp highly express a different set of eight histone genes. During differentiation stage when testes are occupied mostly by spermatocytes and spermatids, another twenty-two histone genes are expressed much higher than the rest, including previously known testis-specific hist1h1t, hist1h2ba and hist1h4c. In addition, histone genes that are pre-dominantly expressed in gonocytes and SSCs are also highly expressed in embryonic stem cells. Several of them were changed when embryoid bodies were formed from ES cells, suggesting their roles in regulating pluripotency of the cells. Further more, differentially expressed histone genes are specifically localized in either SSCs or spermatocytes and spermatids, as demonstrated by in situ hybridization using gene specific probes. Taken together, results presented here revealed that different combinations of histone variant genes are expressed in distinct spermatogenic cell types accompanying the progression of self-renewal and differentiation of SSCs, suggesting a systematic regulatory role histone variants play during spermatogenesis.


Asunto(s)
Cromatina/fisiología , Células Madre Embrionarias/metabolismo , Histonas/genética , Histonas/metabolismo , Espermatogénesis/genética , Espermatogonias/citología , Animales , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Células 3T3 NIH , Espermatogénesis/fisiología , Espermatogonias/fisiología , Testículo/metabolismo
19.
Genesis ; 51(2): 83-96, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23175476

RESUMEN

Mammalian spermatogenesis originates from spermatogonial stem cells (SSCs), which undergo mitosis, meiosis and spermiogenesis in order to generate mature spermatozoa. SSCs are adult stem cells that can both self-renew and differentiate. To maintain pluripotency, SSCs are regulated by both extrinsic factors secreted from surrounding somatic cells and intrinsic factors including specific gene expression programs. Using fluorescent labeled germ line stem cells, mouse gonocytes and SSCs were purified up to 97% by improved FACS method. Through microarray analyses, global gene expression profiles of gonocytes, SSCs, and differentiated cells were compared. A large number of distinctive genes were found to be enriched in respective cell populations, indicating different functional requirements of each cell type. Functional clustering analyses revealed that while gonocytes and SSCs preferentially express genes implicated in gene expression regulation and epigenetic modifications, differentiated cells including somatic cells are enriched with genes encoding proteins involved in various cellular activities. Further in situ hybridization and RT-PCR experiments confirmed SSC specific expression of several genes of which functions have not been characterized in SSCs. The comparative gene expression profiling provides a useful resource for gene discovery in relation to SSC regulation and opens new avenues for the study of molecular mechanisms underlying SSC self-renewal and differentiation.


Asunto(s)
Perfilación de la Expresión Génica , Espermatogonias/citología , Espermatogonias/fisiología , Células Madre/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Citometría de Flujo/métodos , Hibridación in Situ , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Espermatogénesis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...