Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Epigenetics ; 13(1): 28, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33541399

RESUMO

BACKGROUND: Patients suffering from the BCR-ABL1-negative myeloproliferative disease prefibrotic primary myelofibrosis (pre-PMF) have a certain risk for progression to myelofibrosis. Accurate risk estimation for this fibrotic progression is of prognostic importance and clinically relevant. Commonly applied risk scores are based on clinical, cytogenetic, and genetic data but do not include epigenetic modifications. Therefore, we evaluated the assessment of genome-wide DNA methylation patterns for their ability to predict fibrotic progression in PMF patients. RESULTS: For this purpose, the DNA methylation profile was analyzed genome-wide in a training set of 22 bone marrow trephines from patients with either fibrotic progression (n = 12) or stable disease over several years (n = 10) using the 850 k EPIC array from Illumina. The DNA methylation classifier constructed from this data set was validated in an independently measured test set of additional 11 bone marrow trephines (7 with stable disease, 4 with fibrotic progress). Hierarchical clustering of methylation ß-values and linear discriminant classification yielded very good discrimination between both patient groups. By gene ontology analysis, the most differentially methylated CpG sites are primarily associated with genes involved in cell-cell and cell-matrix interactions. CONCLUSIONS: In conclusion, we could show that genome-wide DNA methylation profiling of bone marrow trephines is feasible under routine diagnostic conditions and, more importantly, is able to predict fibrotic progression in pre-fibrotic primary myelofibrosis with high accuracy.


Assuntos
Impressões Digitais de DNA/métodos , Fibrose/genética , Estudo de Associação Genômica Ampla/métodos , Mielofibrose Primária/genética , Experimentação Animal , Medula Óssea/metabolismo , Competição entre as Células/genética , Técnicas de Reprogramação Celular/métodos , Ilhas de CpG/genética , Metilação de DNA , Progressão da Doença , Células Germinativas Embrionárias/metabolismo , Epigenômica/métodos , Feminino , Fibrose/patologia , Proteínas de Fusão bcr-abl/genética , Ontologia Genética , Humanos , Masculino , Valor Preditivo dos Testes , Mielofibrose Primária/patologia , Prognóstico , Fatores de Risco
2.
PLoS One ; 15(4): e0232047, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32339196

RESUMO

Spontaneous testicular teratomas (STTs) derived from primordial germ cells (PGCs) in the mouse embryonic testes predominantly develop in the 129 family inbred strain. Ter (spontaneous mutation) is a single nucleotide polymorphism that generates a premature stop codon of Dead end1 (Dnd1) and increases the incidence of STTs in the 129 genetic background. We previously found that DND1 interacts with NANOS2 or NANOS3 and that these complexes play a vital role in male embryonic germ cells and adult spermatogonia. However, the following are unclear: (a) whether DND1 works with NANOS2 or NANOS3 to regulate teratoma incidence, and (b) whether Ter simply causes Dnd1 loss or produces a short mutant DND1 protein. In the current study, we newly established a conventional Dnd1-knockout mouse line and found that these mice showed phenotypes similar to those of Ter mutant mice in spermatogenesis, oogenesis, and teratoma incidence, with a slight difference in spermiogenesis. In addition, we found that the amount of DND1 in Dnd1+/Ter embryos decreased to half of that in wild-type embryos, while the expression of the short mutant DND1 was not detected. We also found that double mutants for Dnd1 and Nanos2 or Nanos3 showed synergistic increase in the incidence of STTs. These data support the idea that Ter causes Dnd1 loss, leading to an increase in STT incidence, and that DND1 acts with NANOS2 and NANOS3 to regulate the development of teratoma from PGCs in the 129 genetic background. Thus, our results clarify the role of Dnd1 in the development of STTs and provide a novel insight into its pathogenic mechanism.


Assuntos
Células Germinativas Embrionárias/patologia , Proteínas de Neoplasias/fisiologia , Proteínas de Ligação a RNA/metabolismo , Teratoma/etiologia , Neoplasias Testiculares/etiologia , Testículo/patologia , Animais , Células Germinativas Embrionárias/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Oogênese , Proteínas de Ligação a RNA/genética , Espermatogênese , Teratoma/metabolismo , Teratoma/patologia , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia , Testículo/metabolismo
3.
Dev Growth Differ ; 61(6): 357-364, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31199000

RESUMO

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.


Assuntos
Reprogramação Celular/fisiologia , Ciclina D1/metabolismo , Células Germinativas Embrionárias/citologia , Células Germinativas Embrionárias/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Fase G1 , Fase G2 , Histonas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Ativação Enzimática , Fase G1/fisiologia , Fase G2/fisiologia , Masculino , Metilação , Camundongos , Camundongos Transgênicos , Transdução de Sinais
4.
Dev Biol ; 445(1): 103-112, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30439356

RESUMO

Dead end is a vertebrate-specific RNA-binding protein implicated in germ cell development. We have previously shown that mouse Dead end1 (DND1) is expressed in male embryonic germ cells and directly interacts with NANOS2 to cooperatively promote sexual differentiation of fetal germ cells. In addition, we have also reported that NANOS2 is expressed in self-renewing spermatogonial stem cells and is required for the maintenance of the stem cell state. However, it remains to be determined whether DND1 works with NANOS2 in the spermatogonia. Here, we show that DND1 is expressed in a subpopulation of differentiating spermatogonia and undifferentiated spermatogonia, including NANOS2-positive spermatogonia. Conditional disruption of DND1 depleted both differentiating and undifferentiated spermatogonia; however, the numbers of Asingle and Apaired spermatogonia were preferentially decreased as compared with those of Aaligned spermatogonia. Finally, we found that postnatal DND1 associates with NANOS2 in vivo, independently of RNA, and interacts with some of NANOS2-target mRNAs. These data not only suggest that DND1 is a partner of NANOS2 in undifferentiated spermatogonia as well as in male embryonic germ cells, but also show that DND1 plays an essential role in the survival of differentiating spermatogonia.


Assuntos
Proteínas de Neoplasias/metabolismo , Espermatogônias/metabolismo , Animais , Diferenciação Celular , Células Germinativas Embrionárias/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/fisiologia , RNA/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/fisiologia , Reprodução/fisiologia , Espermatogênese/genética , Espermatogônias/citologia , Espermatozoides/metabolismo , Células-Tronco/citologia , Testículo/metabolismo
5.
PLoS One ; 13(10): e0205004, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30286177

RESUMO

Primordial germ cells (PGCs) are precursors of eggs and sperm. Although PGCs are unipotent cells in vivo, they are reprogrammed into pluripotent stem cells (PSCs), also known as embryonic germ cells (EGCs), in the presence of leukemia inhibitory factor and basic fibroblast growth factor (bFGF) in vitro. However, the molecular mechanisms responsible for their reprogramming are not fully understood. Here we show identification of transcription factors that mediate PGC reprogramming. We selected genes encoding transcription factors or epigenetic regulatory factors whose expression was significantly different between PGCs and PSCs with in silico analysis and RT-qPCR. Among the candidate genes, over-expression (OE) of Bcl3 or Klf9 significantly enhanced PGC reprogramming. Notably, EGC formation was stimulated by Klf9-OE even without bFGF. G-protein-coupled receptor signaling-related pathways, which are involved in PGC reprogramming, were enriched among genes down-regulated by Klf9-OE, and forskolin which activate adenylate cyclase, rescued repressed EGC formation by knock-down of Klf9, suggesting a molecular linkage between KLF9 and such signaling.


Assuntos
Reprogramação Celular , Células Germinativas Embrionárias/citologia , Fatores de Transcrição Kruppel-Like/metabolismo , Óvulo/citologia , Proteínas Proto-Oncogênicas/metabolismo , Espermatozoides/citologia , Fatores de Transcrição/metabolismo , Animais , Proteína 3 do Linfoma de Células B , AMP Cíclico/metabolismo , Células Germinativas Embrionárias/metabolismo , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
Sci Rep ; 8(1): 10955, 2018 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-30026469

RESUMO

Patient-specific induced pluripotent stem cells (iPSCs) have the potential to be useful in the treatment of human diseases. While prior studies have reported multiple methods to generate iPSCs, DNA methylation continues to limit the totipotency and reprogramming efficiency of iPSCs. Here, we first show the competency of embryonic germ cells (EGCs) as a reprogramming catalyst capable of effectively promoting reprogramming induced by four defined factors, including Oct4, Sox2, Klf4 and c-Myc. Combining EGC extracts with these four factors resulted in formation of more embryonic stem cell-like colonies than did factors alone. Notably, expression of imprinted genes was higher with combined induction than with factors alone. Moreover, iPSCs derived from the combined inductors tended to have more global hypomethylation. Our research not only provides evidence that EGC extracts could activate DNA demethylation and reprogram imprinted genes, but also establishes a new way to enhance reprogramming of iPSCs, which remains a critical safety concern for potential use of iPSCs in regenerative medicine.


Assuntos
Células Germinativas Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Células Cultivadas , Reprogramação Celular , Metilação de DNA , Células Germinativas Embrionárias/metabolismo , Feminino , Impressão Genômica , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-myc , Medicina Regenerativa
7.
Toxicol Appl Pharmacol ; 352: 38-45, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29800640

RESUMO

Mice lacking the modifier subunit of glutamate cysteine ligase (Gclm), the rate-limiting enzyme in glutathione (GSH) synthesis, have decreased tissue GSH. We previously showed that Gclm-/- embryos have increased sensitivity to the prenatal in vivo ovarian toxicity of the polycyclic aromatic hydrocarbon benzo[a]pyrene (BaP) compared with Gclm+/+ littermates. We also showed that BaP-induced germ cell death in cultured wild type embryonic ovaries is caspase-dependent. Here, we hypothesized that GSH deficiency increases sensitivity of cultured embryonic ovaries to BaP-induced germ cell death. 13.5 days post coitum (dpc) embryonic ovaries of all Gclm genotypes were fixed immediately or cultured for 24 h in media supplemented with DMSO vehicle or 500 ng/ml BaP. The percentage of activated caspase-3 positive germ cells varied significantly among groups. Within each genotype, DMSO and BaP-treated groups had increased germ cell caspase-3 activation compared to uncultured. Gclm+/- ovaries had significantly increased caspase-3 activation with BaP treatment compared to DMSO, and caspase-3 activation increased non-significantly in Gclm-/- ovaries treated with BaP compared to DMSO. There was no statistically significant effect of BaP treatment on germ cell numbers at 24 h, consistent with our prior observations in wild type ovaries, but Gclm-/- ovaries in both cultured groups had lower germ cell numbers than Gclm+/+ ovaries. There were no statistically significant BaP-treatment or genotype-related differences among groups in lipid peroxidation and germ cell proliferation. These data indicate that Gclm heterozygous or homozygous deletion sensitizes embryonic ovaries to BaP- and tissue culture-induced germ cell apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Benzo(a)pireno/toxicidade , Células Germinativas Embrionárias/efeitos dos fármacos , Glutationa/deficiência , Ovário/efeitos dos fármacos , Animais , Citoproteção , Células Germinativas Embrionárias/metabolismo , Células Germinativas Embrionárias/patologia , Feminino , Idade Gestacional , Glutamato-Cisteína Ligase/deficiência , Glutamato-Cisteína Ligase/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovário/embriologia , Ovário/metabolismo , Técnicas de Cultura de Tecidos
8.
Development ; 145(6)2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29545285

RESUMO

Testicular teratomas result from anomalies in embryonic germ cell development. In 129 inbred mice, teratoma initiation coincides with germ cell sex-specific differentiation and the mitotic-meiotic switch: XX and XY germ cells repress pluripotency, XX germ cells initiate meiosis, and XY germ cells activate male-specific differentiation and mitotic arrest. Here, we report that expression of Nanos2, a gene that is crucial to male sex specification, is delayed in teratoma-susceptible germ cells. Decreased expression of Nanos2 was found to be due, in part, to the Nanos2 allele present in 129 mice. In teratoma-susceptible germ cells, diminished expression of genes downstream of Nanos2 disrupted processes that were crucial to male germ cell differentiation. Deficiency for Nanos2 increased teratoma incidence in 129 mice and induced developmental abnormalities associated with tumor initiation in teratoma-resistant germ cells. Finally, in the absence of commitment to the male germ cell fate, we discovered that a subpopulation of teratoma-susceptible germ cells transition into embryonal carcinoma (EC) cells with primed pluripotent features. We conclude that delayed male germ cell sex-specification facilitates the transformation of germ cells with naïve pluripotent features into primed pluripotent EC cells.


Assuntos
Células-Tronco de Carcinoma Embrionário/metabolismo , Células Germinativas Embrionárias/metabolismo , Teratoma/metabolismo , Neoplasias Testiculares/metabolismo , Animais , Técnicas de Cultura de Células , Diferenciação Celular/genética , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos da Linhagem 129 , Polimorfismo de Nucleotídeo Único , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Processos de Determinação Sexual/genética
9.
FEBS Lett ; 592(6): 852-877, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28834535

RESUMO

Estrogen-related receptor b (Esrrb) is part of a family of three orphan nuclear receptors with broad expression profiles and a generic function in regulating energy metabolism in mammals. However, Esrrb performs specific functions during early mouse development, in pluripotent and multipotent populations of the embryo as well as in primordial germ cells. Moreover, Esrrb also impinges upon the control of self-renewal in embryo-derived stem cells and enhances reprogramming. Here, we review the function of Esrrb with special emphasis on its role in pluripotency. Esrrb activity at crucial regulatory elements of the pluripotency network, coupled with its role as a mitotic bookmarking factor and the ability to reset cellular metabolism, might explain its potent functions in ensuring the stability of pluripotency and driving the late stages of reprogramming. Hence, we argue that Esrrb represents a key addition to the pantheon of transcription factors sustaining pluripotent stem cell identity in mice. Understanding the mechanisms governing the interplay between different estrogen-related receptors (ERRs) and their specificity of action may clarify the role these factors play during preimplantation development and in pluripotent cells in both mouse and humans.


Assuntos
Blastocisto/metabolismo , Reprogramação Celular/fisiologia , Células Germinativas Embrionárias/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Blastocisto/citologia , Células Germinativas Embrionárias/citologia , Humanos , Camundongos
10.
J Biol Chem ; 291(19): 10318-31, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26945064

RESUMO

The embryonic myosin isoform is expressed during fetal development and rapidly down-regulated after birth. Freeman-Sheldon syndrome (FSS) is a disease associated with missense mutations in the motor domain of this myosin. It is the most severe form of distal arthrogryposis, leading to overcontraction of the hands, feet, and orofacial muscles and other joints of the body. Availability of human embryonic muscle tissue has been a limiting factor in investigating the properties of this isoform and its mutations. Using a recombinant expression system, we have studied homogeneous samples of human motors for the WT and three of the most common FSS mutants: R672H, R672C, and T178I. Our data suggest that the WT embryonic myosin motor is similar in contractile speed to the slow type I/ß cardiac based on the rate constant for ADP release and ADP affinity for actin-myosin. All three FSS mutations show dramatic changes in kinetic properties, most notably the slowing of the apparent ATP hydrolysis step (reduced 5-9-fold), leading to a longer lived detached state and a slowed Vmax of the ATPase (2-35-fold), indicating a slower cycling time. These mutations therefore seriously disrupt myosin function.


Assuntos
Trifosfato de Adenosina/metabolismo , Disostose Craniofacial/genética , Disostose Craniofacial/patologia , Proteínas do Citoesqueleto/genética , Contração Muscular/fisiologia , Mutação/genética , Subfragmentos de Miosina/genética , Adenosina Trifosfatases/metabolismo , Células Cultivadas , Disostose Craniofacial/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Germinativas Embrionárias/citologia , Células Germinativas Embrionárias/metabolismo , Humanos , Hidrólise , Subfragmentos de Miosina/metabolismo , Isoformas de Proteínas
11.
Cell Cycle ; 15(7): 919-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26901436

RESUMO

Testicular teratomas result from anomalies in embryonic germ cell development. In the 129 family of inbred mouse strains, teratomas arise during the same developmental period that male germ cells normally enter G1/G0 mitotic arrest and female germ cells initiate meiosis (the mitotic:meiotic switch). Dysregulation of this switch associates with teratoma susceptibility and involves three germ cell developmental abnormalities seemingly critical for tumor initiation: delayed G1/G0 mitotic arrest, retention of pluripotency, and misexpression of genes normally restricted to embryonic female and adult male germ cells. One misexpressed gene, cyclin D1 (Ccnd1), is a known regulator of cell cycle progression and an oncogene in many tissues. Here, we investigated whether Ccnd1 misexpression in embryonic germ cells is a determinant of teratoma susceptibility in mice. We found that CCND1 localizes to teratoma-susceptible germ cells that fail to enter G1/G0 arrest during the mitotic:meiotic switch and is the only D-type cyclin misexpressed during this critical developmental time frame. We discovered that Ccnd1 deficiency in teratoma-susceptible mice significantly reduced teratoma incidence and suppressed the germ cell proliferation and pluripotency abnormalities associated with tumor initiation. Importantly, Ccnd1 expression was dispensable for somatic cell development and male germ cell specification and maturation in tumor-susceptible mice, implying that the mechanisms by which Ccnd1 deficiency reduced teratoma incidence were germ cell autonomous and specific to tumorigenesis. We conclude that misexpression of Ccnd1 in male germ cells is a key component of a larger pro-proliferative program that disrupts the mitotic:meiotic switch and predisposes 129 inbred mice to testicular teratocarcinogenesis.


Assuntos
Ciclina D1/genética , Células Germinativas Embrionárias/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular , Teratoma/etiologia , Neoplasias Testiculares/etiologia , Animais , Proliferação de Células , Ciclina D1/metabolismo , Feminino , Expressão Gênica , Predisposição Genética para Doença , Células Intersticiais do Testículo/metabolismo , Masculino , Meiose , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Mitose , Células de Sertoli/metabolismo , Teratoma/genética , Teratoma/metabolismo , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo
12.
Cell Tissue Res ; 364(2): 429-41, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26613602

RESUMO

Primordial germ cells (PGCs) have the ability to be reprogrammed into embryonic germ cells (EGCs) in vitro and are an alternative source of embryonic stem cells. Other than for the mouse, the systematic characterization of mammalian PGCs is still lacking, especially the process by which PGCs convert to pluripotency. This hampers the understanding of germ cell development and the derivation of authenticated EGCs from other species. We observed the morphological development of the genital ridge from Bama miniature pigs and found primary sexual differentiation in the E28 porcine embryo, coinciding with Blimp1 nuclear exclusion in PGCs. To explore molecular events involved in porcine PGC reprogramming, transcriptome data of porcine EGCs and fetal fibroblasts (FFs) were assembled and 1169 differentially expressed genes were used for Gene Ontology analysis. These genes were significantly enriched in cell-surface receptor-linked signal transduction, in agreement with the activation of LIF/Stat3 signaling and FGF signaling during the derivation of porcine EG-like cells. Using a growth-factor-defined culture system, we explored the effects of bFGF on the process and found that bFGF not only functioned at the very beginning of PGC dedifferentiation by impeding Blimp1 nuclear expression via a PI3K/AKT-dependent pathway but also maintained the viability of cultured PGCs thereafter. These results provide further insights into the development of germ cells from livestock and the mechanism of porcine PGC reprogramming.


Assuntos
Reprogramação Celular/fisiologia , Células Germinativas Embrionárias/citologia , Fatores de Crescimento de Fibroblastos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Porco Miniatura/embriologia , Animais , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Células Germinativas Embrionárias/metabolismo , Fator Inibidor de Leucemia/metabolismo , Gado/embriologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Suínos/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA