Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Front Cell Dev Biol ; 12: 1410177, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38911025

RESUMEN

Mammalian germ cells are derived from primordial germ cells (PGCs) and ensure species continuity through generations. Unlike irreversible committed mature germ cells, migratory PGCs exhibit a latent pluripotency characterized by the ability to derive embryonic germ cells (EGCs) and form teratoma. Here, we show that inhibition of p38 mitogen-activated protein kinase (MAPK) by chemical compounds in mouse migratory PGCs enables derivation of chemically induced Embryonic Germ-like Cells (cEGLCs) that do not require conventional growth factors like LIF and FGF2/Activin-A, and possess unique naïve pluripotent-like characteristics with epiblast features and chimera formation potential. Furthermore, cEGLCs are regulated by a unique PI3K-Akt signaling pathway, distinct from conventional naïve pluripotent stem cells described previously. Consistent with this notion, we show by performing ex vivo analysis that inhibition of p38 MAPK in organ culture supports the survival and proliferation of PGCs and also potentially reprograms PGCs to acquire indefinite proliferative capabilities, marking these cells as putative teratoma-producing cells. These findings highlight the utility of our ex vivo model in mimicking in vivo teratoma formation, thereby providing valuable insights into the cellular mechanisms underlying tumorigenesis. Taken together, our research underscores a key role of p38 MAPK in germ cell development, maintaining proper cell fate by preventing unscheduled pluripotency and teratoma formation with a balance between proliferation and differentiation.

2.
Environ Epigenet ; 9(1): dvad008, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38094661

RESUMEN

Growing evidence suggests that epigenetic changes through various parental environmental factors alter the phenotypes of descendants in various organisms. Environmental factors, including exposure to chemicals, stress and abnormal nutrition, affect the epigenome in parental germ cells by different epigenetic mechanisms, such as DNA methylation, histone modification as well as small RNAs via metabolites. Some current remaining questions are the causal relationship between environment-induced epigenetic changes in germ cells and altered phenotypes of descendants, and the molecular basis of how the abnormal epigenetic changes escape reprogramming in germ cells. In this review, we introduce representative examples of intergenerational and transgenerational inheritance of phenotypic changes through parental environmental factors and the accompanied epigenetic and metabolic changes, with a focus on animal species. We also discuss the molecular mechanisms of epigenomic inheritance and their possible biological significance.

3.
EMBO Rep ; 24(11): e56845, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37842859

RESUMEN

Fate determination of primordial germ cells (PGCs) is regulated in a multi-layered manner, involving signaling pathways, epigenetic mechanisms, and transcriptional control. Chemical modification of macromolecules, including epigenetics, is expected to be closely related with metabolic mechanisms but the detailed molecular machinery linking these two layers remains poorly understood. Here, we show that the hexosamine biosynthetic pathway controls PGC fate determination via O-linked ß-N-acetylglucosamine (O-GlcNAc) modification. Consistent with this model, reduction of carbohydrate metabolism via a maternal ketogenic diet that decreases O-GlcNAcylation levels causes repression of PGC formation in vivo. Moreover, maternal ketogenic diet intake until mid-gestation affects the number of ovarian germ cells in newborn pups. Taken together, we show that nutritional and metabolic mechanisms play a previously unappreciated role in PGC fate determination.


Asunto(s)
Acetilglucosamina , Transducción de Señal , Recién Nacido , Humanos , Transducción de Señal/fisiología , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Regulación de la Expresión Génica , Epigénesis Genética , Células Germinativas/metabolismo , Procesamiento Proteico-Postraduccional
4.
Sex Dev ; 16(5-6): 388-403, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35086109

RESUMEN

BACKGROUND: The germ cell lineage involves dynamic epigenetic changes during its formation and differentiation that are completely different from those of the somatic cell lineage. Metabolites and metabolic pathways have been reported as key factors related to the regulation of epigenetics as cofactors and substrates. However, our knowledge about the metabolic characteristics of germ cells, especially during the fetal stage, and their transition during differentiation is quite limited due to the rarity of the cells. Nevertheless, recent developments in omics technologies have made it possible to extract comprehensive metabolomic features of germ cells. SUMMARY: In this review, we present the latest researches on the metabolic properties of germ cells in 4 stages: primordial germ cell specification, fetal germ cell differentiation, spermatogenesis, and oogenesis. At every stage, extensive published data has been accumulated on energy metabolism, and it is possible to describe its changes during germ cell differentiation in detail. As pluripotent stem cells differentiate into germ cells, energy metabolism shifts from glycolysis to oxidative phosphorylation; however, in spermatogenesis, glycolytic pathways are also temporarily dominant in spermatogonial stem cells. Although the significance of metabolic pathways other than energy metabolism in germ cell differentiation is largely unknown, the relation of the pentose phosphate pathway and Ser-Gly-one-carbon metabolism with germ cell properties has been suggested at various stages. We further discuss the relationship between these characteristic metabolic pathways and epigenetic regulation during germ cell specification and differentiation. Finally, the relevance of dietary and supplemental interventions on germ cell function and epigenomic regulation is also discussed. KEY MESSAGES: Comprehensive elucidation of metabolic features and metabolism-epigenome crosstalk in germ cells is important to reveal how the characteristic metabolic pathways are involved in the germ cell regulation. The accumulation of such insights would lead to suggestions for optimal diets and supplements to maintain reproductive health through modulating metabolic and epigenetic status of germ cells.

5.
J Reprod Dev ; 68(2): 96-103, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-34955463

RESUMEN

Metabolism is an important cellular process necessary not only for producing energy and building blocks for cells, but also for regulating various cell functions, including intracellular signaling, epigenomic effects, and transcription. The regulatory roles of metabolism have been extensively studied in somatic cells, including stem cells and cancer cells, but data regarding germ cells are limited. Because germ cells produce individuals of subsequent generations, understanding the role of metabolism and its regulatory functions in germ cells is important. Although limited information concerning the specific role of metabolism in germ cells is available, recent advances in related research have revealed specific metabolic states of undifferentiated germ cells in embryos as well as in germ cells undergoing oogenesis and spermatogenesis. Studies have also elucidated the functions of some metabolic pathways associated with germ cell development and the non-genomic heritable machinery of germ cells. In this review, we summarized all the available knowledge on the characteristic metabolic pathways in germ cells, focusing on their regulatory functions, while discussing the issues that need to be addressed to enhance the understanding of germ cell metabolism.


Asunto(s)
Células Germinativas , Espermatogénesis , Diferenciación Celular , Células Germinativas/metabolismo , Humanos , Masculino , Redes y Vías Metabólicas , Oogénesis
6.
STAR Protoc ; 3(4): 101912, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36595925

RESUMEN

It is now recognized that maternal environmental factors, including chemical exposure and nutritional conditions, alter DNA methylation patterns in fetal germ cells, subsequently affecting germ cell development as well as offspring phenotypes. Here, we describe steps for detecting DNA methylation changes in mouse germ cells isolated from both embryonic and spermatogenic stages after maternal exposure to a chemical compound. For complete details on the use and execution of this protocol, please refer to Tando et al. (2021).1.


Asunto(s)
Metilación de ADN , Exposición Materna , Humanos , Animales , Ratones , Femenino , Masculino , Exposición Materna/efectos adversos , Espermatogénesis/genética , Células Germinativas , Reproducción
8.
Elife ; 102021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34319233

RESUMEN

Exposure to environmental factors during fetal development may lead to epigenomic modifications in fetal germ cells, altering gene expression and promoting diseases in successive generations. In mouse, maternal exposure to di(2-ethylhexyl) phthalate (DEHP) is known to induce defects in spermatogenesis in successive generations, but the mechanism(s) of impaired spermatogenesis are unclear. Here, we showed that maternal DEHP exposure results in DNA hypermethylation of promoters of spermatogenesis-related genes in fetal testicular germ cells in F1 mice, and hypermethylation of Hist1h2ba, Sycp1, and Taf7l, which are crucial for spermatogenesis, persisted from fetal testicular cells to adult spermatogonia, resulting in the downregulation of expression of these genes. Forced methylation of these gene promoters silenced expression of these loci in a reporter assay. These results suggested that maternal DEHP exposure-induced hypermethylation of Hist1h2ba, Sycp1, and Taf7l results in downregulation of these genes in spermatogonia and subsequent defects in spermatogenesis, at least in the F1 generation.


Asunto(s)
Dietilhexil Ftalato/farmacología , Exposición Materna/efectos adversos , Mutación , Ácidos Ftálicos/efectos adversos , Efectos Tardíos de la Exposición Prenatal/genética , Espermatogénesis/efectos de los fármacos , Espermatogénesis/genética , Animales , Metilación de ADN , Regulación hacia Abajo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ácidos Ftálicos/química , Plastificantes/efectos adversos , Embarazo , Espermatogonias/efectos de los fármacos , Testículo/citología , Testículo/efectos de los fármacos
9.
Biol Reprod ; 105(1): 64-75, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-33824958

RESUMEN

Fetal ovarian germ cells show characteristic energy metabolism status, such as enhanced mitochondrial metabolism as well as glycolysis, but their roles in early folliculogenesis are unclear. We show here that inhibition of pyruvate uptake to mitochondria by UK5099 in organ cultures of fetal mouse ovaries resulted in repressed early folliculogenesis without affecting energy production, survival of oocytes, or meiosis. In addition, the abnormal folliculogenesis by UK5099 was partially rescued by α-ketoglutarate and succinate, intermediate metabolites in the TCA cycle, suggesting the importance of those metabolites. The expression of TGFß-related genes Gdf9 and Bmp15 in ovarian germ cells, which are crucial for folliculogenesis, was downregulated by UK5099, and the addition of recombinant GDF9 partially rescued the abnormal folliculogenesis induced by UK5099. We also found that early folliculogenesis was similarly repressed, as in the culture, in the ovaries of a germ cell-specific knockout of Mpc2, which encodes a mitochondria pyruvate carrier that is targeted by UK5099. These results suggest that insufficient Gdf9 expression induced by abnormal pyruvate metabolism in oocytes results in early follicular dysgenesis, which is a possible cause of defective folliculogenesis in humans.


Asunto(s)
Acrilatos/farmacología , Proteína Morfogenética Ósea 15/genética , Factor 9 de Diferenciación de Crecimiento/genética , Oocitos/efectos de los fármacos , Folículo Ovárico/crecimiento & desarrollo , Ácido Pirúvico/metabolismo , Animales , Transporte Biológico , Proteína Morfogenética Ósea 15/metabolismo , Ciclo del Ácido Cítrico , Femenino , Regulación de la Expresión Génica , Factor 9 de Diferenciación de Crecimiento/metabolismo , Ratones , Mitocondrias/metabolismo , Oocitos/metabolismo
10.
EMBO Rep ; 22(2): e51524, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33399271

RESUMEN

Advanced paternal age can have deleterious effects on various traits in the next generation. Here, we establish a paternal-aging model in mice to understand the molecular mechanisms of transgenerational epigenetics. Whole-genome target DNA methylome analyses of sperm from aged mice reveal more hypo-methylated genomic regions enriched in REST/NRSF binding motifs. Gene set enrichment analyses also reveal the upregulation of REST/NRSF target genes in the forebrain of embryos from aged fathers. Offspring derived from young mice administrated with a DNA de-methylation drug phenocopy the abnormal vocal communication of pups derived from aged fathers. In conclusion, hypo-methylation of sperm DNA can be a key molecular feature modulating neurodevelopmental programs in offspring by causing fluctuations in the expression of REST/NRSF target genes.


Asunto(s)
Metilación de ADN , Edad Paterna , Animales , Epigénesis Genética , Padre , Humanos , Masculino , Ratones , Espermatozoides/metabolismo
11.
Biol Reprod ; 103(4): 717-735, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32627815

RESUMEN

Regulatory mechanisms of germline differentiation have generally been explained via the function of signaling pathways, transcription factors, and epigenetic regulation; however, little is known regarding proteomic and metabolomic regulation and their contribution to germ cell development. Here, we conducted integrated proteomic and metabolomic analyses of fetal germ cells in mice on embryonic day (E)13.5 and E18.5 and demonstrate sex- and developmental stage-dependent changes in these processes. In male germ cells, RNA processing, translation, oxidative phosphorylation, and nucleotide synthesis are dominant in E13.5 and then decline until E18.5, which corresponds to the prolonged cell division and more enhanced hyper-transcription/translation in male primordial germ cells and their subsequent repression. Tricarboxylic acid cycle and one-carbon pathway are consistently upregulated in fetal male germ cells, suggesting their involvement in epigenetic changes preceding in males. Increased protein stability and oxidative phosphorylation during female germ cell differentiation suggests an upregulation of aerobic energy metabolism, which likely contributes to the proteostasis required for oocyte maturation in subsequent stages. The features elucidated in this study shed light on the unrevealed mechanisms of germ cell development.


Asunto(s)
Diferenciación Celular/fisiología , Células Germinales Embrionarias/fisiología , Metabolómica , Proteómica , Animales , ADN/genética , ADN/metabolismo , Metilación de ADN , Embrión de Mamíferos/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Estudio de Asociación del Genoma Completo , Masculino , Ratones , Ratones Transgénicos , Diferenciación Sexual , Factores Sexuales , Factores de Transcripción
12.
Sci Rep ; 9(1): 11799, 2019 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-31395923

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
Mol Cell Biol ; 39(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31208979

RESUMEN

The cancer/testis antigen (CTA) genes were identified as human genes preferentially expressed in cancer cells and testis, but the contribution of CTAs to cancer and male germ cell development is unclear. In this study, we comprehensively examined mouse CTA functions and found that the majority of CTAs are involved in growth and/or survival of cancer cells. We focused on one mouse CTA gene, Tekt5, for its detailed functional analysis. Tekt5 knockdown (KD) in ovarian cancer cells caused G1 arrest and apoptosis, and p27kip1 was concomitantly upregulated. Tekt5 KD also resulted in decreased levels of acetylated α-tubulin and subsequent fragmentation of ß-III-tubulin, upregulation of HDAC6 that deacetylates α-tubulin, and nuclear accumulation of SMAD3 that induces p27kip1 expression. Because depolymerization of tubulin is known to cause translocation of SMAD3 to the nucleus, these results together suggested that TEKT5 negatively regulates Hdac6 expression and consequently maintains cell cycle via stabilization of tubulin. We also found that the number of spermatids was significantly decreased and acetylated α-tubulin levels were decreased in vivo by KD of Tekt5 in testis. Because acetylated α-tubulin is required for sperm morphogenesis, these results suggest that TEKT5 is necessary for spermiogenesis via maintenance of acetylated α-tubulin levels.


Asunto(s)
Proteínas de Microtúbulos/genética , Neoplasias Ováricas/genética , Espermatogénesis , Espermatozoides/citología , Tubulina (Proteína)/metabolismo , Acetilación , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 6/metabolismo , Masculino , Ratones , Estabilidad Proteica , Proteína smad3/metabolismo , Espermatozoides/metabolismo , Tubulina (Proteína)/química
15.
Dev Growth Differ ; 61(6): 357-364, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31199000

RESUMEN

Primordial germ cells (PGCs) are reprogrammed into pluripotent embryonic germ cells (EGCs) under specific culture conditions, but the detailed mechanisms of PGC reprogramming have not yet been fully clarified. Previous studies have demonstrated that AKT, an important intracellular signaling molecule, promotes reprogramming of PGCs into EGCs. Because AKT likely inhibits p53 functions to enhance PGC reprogramming, and p53 negatively regulates cell cycle progression, we analyzed cell cycle changes in PGCs following AKT activation and found that the ratio of PGCs in the G1/G0 phase was decreased while that of PGCs in the G2/M phase was increased after AKT activation. We also showed that the expression of the CDK inhibitor p27kip1, which prevents the G1­S transition and is transcriptionally activated by p53, was significantly downregulated by AKT activation. The results suggested that the characteristic cell cycle changes of PGCs by AKT activation are, at least in part, due to decreased expression of p27kip1 . We also investigated changes in histone H3K27 tri-methylation (H3K27me3) by AKT activation in PGCs, because we previously found that decreased H3K27me3 was involved in PGC reprogramming via upregulation of cyclin D1. We observed that AKT activation in PGCs resulted in H3K27 hypomethylation. In addition, DZNeP, an inhibitor of the H3K27 trimethyl transferase Ezh2, stimulated EGC formation. These results together suggested that AKT activation promotes G1-S transition and downregulates H3K27me3 to enhance PGC reprogramming.


Asunto(s)
Reprogramación Celular/fisiología , Ciclina D1/metabolismo , Células Germinales Embrionarias/citología , Células Germinales Embrionarias/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Fase G1 , Fase G2 , Histonas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Activación Enzimática , Fase G1/fisiología , Fase G2/fisiología , Masculino , Metilación , Ratones , Ratones Transgénicos , Transducción de Señal
16.
Sci Rep ; 9(1): 6132, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992461

RESUMEN

Many flatworms can alternate between asexual and sexual reproduction. This is a powerful reproductive strategy enabling them to benefit from the features of the two reproductive modes, namely, rapid multiplication and genetic shuffling. The two reproductive modes are enabled by the presence of pluripotent adult stem cells (neoblasts), by generating any type of tissue in the asexual mode, and producing and maintaining germ cells in the sexual mode. In the current study, RNA sequencing (RNA-seq) was used to compare the transcriptomes of two phenotypes of the planarian Dugesia ryukyuensis: an asexual OH strain and an experimentally sexualized OH strain. Pathway enrichment analysis revealed striking differences in amino acid metabolism in the two worm types. Further, the analysis identified serotonin as a new bioactive substance that induced the planarian ovary de novo in a postembryonic manner. These findings suggest that different metabolic states and physiological conditions evoked by sex-inducing substances likely modulate stem cell behavior, depending on their different function in the asexual and sexual reproductive modes. The combination of RNA-seq and a feeding assay in D. ryukyuensis is a powerful tool for studying the alternation of reproductive modes, disentangling the relationship between gene expression and chemical signaling molecules.


Asunto(s)
Aminoácidos/metabolismo , Ovario/crecimiento & desarrollo , Planarias/fisiología , Reproducción Asexuada/genética , Procesos de Determinación del Sexo/genética , Animales , Bioensayo , Femenino , Regulación de la Expresión Génica/fisiología , Masculino , Redes y Vías Metabólicas/genética , Ovario/metabolismo , Células Madre Pluripotentes/metabolismo , RNA-Seq , Serotonina/metabolismo
17.
PLoS One ; 14(2): e0211739, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30707741

RESUMEN

MicroRNAs (miRNAs) play a critical role in multiple aspects of biology. Dicer, an RNase III endonuclease, is essential for the biogenesis of miRNAs, and the germ cell-specific Dicer1 knockout mouse shows severe defects in gametogenesis. How miRNAs regulate germ cell development is still not fully understood. In this study, we identified germ cell-specific miRNAs (miR-741-3p, miR-871-3p, miR-880-3p) by analyzing published RNA-seq data of mouse. These miRNA genes are contiguously located on the X chromosome near other miRNA genes. We named them X chromosome-linked miRNAs (XmiRs). To elucidate the functions of XmiRs, we generated knockout mice of these miRNA genes using the CRISPR/Cas9-mediated genome editing system. Although no histological abnormalities were observed in testes of F0 mice in which each miRNA gene was disrupted, a deletion covering miR-871 and miR-880 or covering all XmiRs (ΔXmiRs) resulted in arrested spermatogenesis in meiosis in a few seminiferous tubules, indicating their redundant functions in spermatogenesis. Among candidate targets of XmiRs, we found increased expression of a gene encoding a WNT receptor, FZD4, in ΔXmiRs testis compared with that in wildtype testis. miR-871-3p and miR-880-3p repressed the expression of Fzd4 via the 3'-untranslated region of its mRNA. In addition, downstream genes of the WNT/ß-catenin pathway were upregulated in ΔXmiRs testis. We also found that miR-871, miR-880, and Fzd4 were expressed in spermatogonia, spermatocytes and spermatids, and overexpression of miR-871 and miR-880 in germ stem cells in culture repressed their increase in number and Fzd4 expression. Previous studies indicated that the WNT/ß-catenin pathway enhances and represses proliferation and differentiation of spermatogonia, respectively, and our results consistently showed that stable ß-catenin enhanced GSC number. In addition, stable ß-catenin partially rescued reduced GSC number by overexpression of miR-871 and miR-880. The results together suggest that miR-871 and miR-880 cooperatively regulate the WNT/ß-catenin pathway during testicular germ cell development.


Asunto(s)
MicroARNs/genética , Espermatogénesis/genética , Cromosoma X/genética , Animales , Sistemas CRISPR-Cas , Diferenciación Celular/genética , Proliferación Celular , Células Germinativas , Masculino , Meiosis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/clasificación , Espermátides/metabolismo , Espermatocitos/metabolismo , Espermatogonias/metabolismo , Testículo/metabolismo
18.
Epigenomics ; 11(5): 543-561, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30667280

RESUMEN

AIMS: Epigenetic signatures of germline cells are dynamically reprogrammed to induce appropriate differentiation, development and sex specification. We investigated sex-specific epigenetic changes in mouse fetal germ cells (FGCs) and neonatal germ cells. MATERIALS & METHODS: Six histone marks in mouse E13.5 FGCs and P1 neonatal germ cells were analyzed by chromatin immunoprecipitation and sequencing. These datasets were compared with transposase-accessible chromatin sites, DNA methylation and transcriptome. RESULTS: Different patterns of each histone mark were detected in female and male FGCs, and H3K4me3/H3K27me3 bivalent marks were enriched in different chromosomal regions of female and male FGCs. CONCLUSION: Our results suggest that histone modifications may affect FGC gene expression following DNA methylation erasure, contributing to the differentiation into female and male germ cells.


Asunto(s)
Células Germinativas/metabolismo , Histonas/metabolismo , Animales , Animales Recién Nacidos , Epigenómica , Femenino , Feto/citología , Células Germinativas/citología , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Transcriptoma
19.
Dev Biol ; 446(1): 43-55, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30529251

RESUMEN

Teratomas are tumors consisting of components of the three germ layers that differentiate from pluripotent stem cells derived from germ cells. In the normal mouse testis, teratomas rarely form, but a deficiency in Dead-end1 (Dnd1) in mice with a 129/Sv genetic background greatly enhances teratoma formation. Thus, DND1 is crucial for suppression of teratoma development from germ cells. In the Dnd1 mutant testis, nascent teratoma cells emerge at E15.5. To understand the nature of early teratoma cells, we established cell lines in the presence of serum and leukemia inhibitory factor (LIF) from teratoma-forming cells in neonatal Dnd1 mutant testis. These cells, which we designated cultured Dnd1 mutant germ cells (CDGCs), were morphologically similar to embryonic stem cells (ESCs) and could be maintained in the naïve pluripotent condition. In addition, the cells expressed pluripotency genes including Oct4, Nanog, and Sox2; differentiated into cells of the three germ layers in culture; and contributed to chimeric mice. The expression levels of pluripotency genes and global transcriptomes in CDGCs as well as these cells' adaption to culture conditions for primed pluripotency suggested that their pluripotent status is intermediate between naïve and primed pluripotency. In addition, the teratoma-forming cells in the neonatal testis from which CDGCs were derived also showed gene expression profiles intermediate between naïve and primed pluripotency. The results suggested that germ cells in embryonic testes of Dnd1 mutants acquire the intermediate pluripotent status during the course of conversion into teratoma cells.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias de Ratones/metabolismo , Proteínas de Neoplasias/genética , Células Madre Pluripotentes/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Proteínas de Neoplasias/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Testículo/citología , Testículo/embriología , Testículo/metabolismo
20.
Methods Mol Biol ; 2045: 259-269, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29790096

RESUMEN

Primordial germ cells (PGCs), the precursors of gametes, are the only cells capable of acquiring totipotency upon fertilization, but the molecular mechanisms regulating germ cell characteristics have not been fully elucidated. Although intracellular metabolic status and regulation are responsible for the control of cell function and differentiation, little is known about the metabolic features of PGCs. Here, we describe use of an integrated metabolomic, proteomic, and energy metabolic analysis method to comprehensively elucidate the metabolic characteristics of PGCs using mass spectrometry.


Asunto(s)
Células Germinales Embrionarias/metabolismo , Metaboloma/fisiología , Metabolómica/métodos , Proteoma/metabolismo , Proteómica/métodos , Animales , Cromatografía Liquida , Células Germinales Embrionarias/citología , Células Germinales Embrionarias/efectos de los fármacos , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Metaboloma/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Proteoma/efectos de los fármacos , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA