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.
Dev Neurobiol ; 79(6): 559-577, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31177638

RESUMO

A large number of studies have focused on the generation of dopaminergic neurons from pluripotent cells. Differentiation of stem cells into distinct cell types is influenced by tissue-specific microenvironment. Since, central nervous system undergoes further development during postnatal life, in the present study neonatal rat brain tissue extract (NRBE) was applied to direct the differentiation of embryonal carcinoma stem cell line, P19 into dopaminergic (DA) phenotypes. Additionally, a neuroprotective drug, deprenyl was used alone or in combination with the extract. Results from morphological, immunofluorescence, and qPCR analyses showed that during a period of one to three weeks, a large percentage of stem cells were differentiated into neural cells. The results also indicated the greater effect of NRBE on the differentiation of the cells into tyrosine hydroxylase-expressing cells. MS analysis of NRBE showed the enrichment of gene ontology terms related to cell differentiation and neurogenesis. Network analysis of the studied genes and some DA markers resulted in the suggestion of potential regulatory candidates such as AVP, ACHE, LHFPL5, and DLK1 genes. In conclusion, NRBE as a natural native inducer was apparently able to simulate the brain microenvironment and support neural differentiation of P19 cells.


Assuntos
Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/enzimologia , Regulação Enzimológica da Expressão Gênica , Selegilina/farmacologia , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Animais Recém-Nascidos , Células Cultivadas , Inibidores da Monoaminoxidase/farmacologia , Ratos , Ratos Wistar , Tirosina 3-Mono-Oxigenase/genética
2.
J Biochem ; 158(5): 373-84, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26002960

RESUMO

The Mediator complex (Mediator) plays key roles in transcription and functions as the nexus for integration of various transcriptional signals. Previously, we screened for Mediator cyclin-dependent kinase (CDK)-interacting factors and identified three proteins related to chromatin regulation. One of them, SUZ12 is required for both stability and activity of Polycomb Repressive Complex 2 (PRC2). PRC2 primarily suppresses gene expression through histone H3 lysine 27 trimethylation, resulting in stem cell maintenance and differentiation; perturbation of this process leads to oncogenesis. Recent work showed that Mediator contributes to the embryonic stem cell state through DNA loop formation, which is strongly associated with chromatin architecture; however, it remains unclear how Mediator regulates gene expression in cooperation with chromatin regulators (i.e. writers, readers and remodelers). We found that Mediator CDKs interact directly with the PRC2 subunit EZH2, as well as SUZ12. Known PRC2 target genes were deregulated by Mediator CDK knockdown during neuronal differentiation, and both Mediator and PRC2 complexes co-occupied the promoters of developmental genes regulated by retinoic acid. Our results provide a mechanistic link between Mediator and PRC2 during neuronal differentiation.


Assuntos
Quinase 8 Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Neurogênese , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Transcrição Gênica , Tretinoína/metabolismo , Animais , Linhagem Celular , Quinase 8 Dependente de Ciclina/antagonistas & inibidores , Quinase 8 Dependente de Ciclina/genética , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Células-Tronco de Carcinoma Embrionário/citologia , Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste , Deleção de Genes , Humanos , Camundongos , Proteínas de Neoplasias , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Complexo Repressor Polycomb 2/genética , Proteínas Quinases/metabolismo , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Polimerase II/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição
3.
Metallomics ; 5(5): 484-91, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23536109

RESUMO

Organotin compounds such as tributyltin (TBT) are known to cause various forms of cytotoxicity, including developmental toxicity and neurotoxicity. However, the molecular target of the toxicity induced by nanomolar levels of TBT has not been identified. In the present study, we found that exposure to 100 nM TBT induced growth arrest in human pluripotent embryonic carcinoma cell line NT2/D1. Since glucose provides metabolic energy, we focused on the glycolytic system. We found that exposure to TBT reduced the levels of both glucose-6-phosphate and fructose-6-phosphate. To investigate the effect of TBT exposure on glycolysis, we examined glucose transporter (GLUT) activity. TBT exposure inhibited glucose uptake via a decrease in the level of cell surface-bound GLUT1. Furthermore, we examined the effect of AMP-activated protein kinase (AMPK), which is known to regulate glucose transport by facilitating GLUT translocation. Treatment with the potent AMPK activator, AICAR, restored the TBT-induced reduction in cell surface-bound GLUT1 and glucose uptake. In conclusion, these results suggest that exposure to nanomolar levels of TBT causes growth arrest by targeting glycolytic systems in human embryonic carcinoma cells. Thus, understanding the energy metabolism may provide new insights into the mechanisms of metal-induced cytotoxicity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Células-Tronco de Carcinoma Embrionário/metabolismo , Glucose/metabolismo , Compostos de Trialquitina/toxicidade , Transporte Biológico/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco de Carcinoma Embrionário/patologia , Transportador de Glucose Tipo 1/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia
4.
PLoS One ; 7(12): e51563, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251575

RESUMO

Testicular Germ Cell Tumors (TGCT) and patient-derived cell lines are extremely sensitive to cisplatin and other interstrand cross-link (ICL) inducing agents. Nevertheless, a subset of TGCTs are either innately resistant or acquire resistance to cisplatin during treatment. Understanding the mechanisms underlying TGCT sensitivity/resistance to cisplatin as well as the identification of novel strategies to target cisplatin-resistant TGCTs have major clinical implications. Herein, we have examined the proficiency of five embryonal carcinoma (EC) cell lines to repair cisplatin-induced ICLs. Using γH2AX staining as a marker of double strand break formation, we found that EC cell lines were either incapable of or had a reduced ability to repair ICL-induced damage. The defect correlated with reduced Homologous Recombination (HR) repair, as demonstrated by the reduction of RAD51 foci formation and by direct evaluation of HR efficiency using a GFP-reporter substrate. HR-defective tumors cells are known to be sensitive to the treatment with poly(ADP-ribose) polymerase (PARP) inhibitor. In line with this observation, we found that EC cell lines were also sensitive to PARP inhibitor monotherapy. The magnitude of sensitivity correlated with HR-repair reduced proficiency and with the expression levels and activity of PARP1 protein. In addition, we found that PARP inhibition strongly enhanced the response of the most resistant EC cells to cisplatin, by reducing their ability to overcome the damage. These results point to a reduced proficiency of HR repair as a source of sensitivity of ECs to ICL-inducing agents and PARP inhibitor monotherapy, and suggest that pharmacological inhibition of PARP can be exploited to target the stem cell component of the TGCTs (namely ECs) and to enhance the sensitivity of cisplatin-resistant TGCTs to standard treatments.


Assuntos
Cisplatino/farmacologia , Células-Tronco de Carcinoma Embrionário/enzimologia , Inibidores Enzimáticos/farmacologia , Recombinação Homóloga/efeitos dos fármacos , Neoplasias Embrionárias de Células Germinativas/enzimologia , Inibidores de Poli(ADP-Ribose) Polimerases , Neoplasias Testiculares/enzimologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Células-Tronco de Carcinoma Embrionário/patologia , Endonucleases/metabolismo , Inibidores Enzimáticos/uso terapêutico , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Proteínas de Neoplasias/metabolismo , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Ftalazinas/farmacologia , Ftalazinas/uso terapêutico , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Poli(ADP-Ribose) Polimerases/metabolismo , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
5.
PLoS One ; 6(11): e26484, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22140430

RESUMO

BACKGROUND: The Rho kinase pathway plays a key role in many early cell/tissue determination events that take place in embryogenesis. Rho and its downstream effector Rho kinase (ROCK) play pivotal roles in cell migration, apoptosis (membrane blebbing), cell proliferation/cell cycle, cell-cell adhesion and gene regulation. We and others have previously demonstrated that inhibition of ROCK blocks endoderm differentiation in embryonal carcinoma stem cells, however, the effect of ROCK inhibition on mesoderm and ectoderm specification has not been fully examined. In this study, the role of ROCK within the specification and differentiation of all three germ layers was examined. METHODOLOGY/PRINCIPAL FINDINGS: P19 cells were treated with the specific ROCK inhibitor Y-27623, and increase in differentiation efficiency into neuro-ectodermal and mesodermal lineages was observed. However, as expected a dramatic decrease in early endodermal markers was observed when ROCK was inhibited. Interestingly, within these ROCK-inhibited RA treated cultures, increased levels of mesodermal or ectodermal markers were not observed, instead it was found that the pluripotent markers SSEA-1 and Oct-4 remained up-regulated similar to that seen in undifferentiated cultures. Using standard and widely accepted methods for reproducible P19 differentiation into all three germ layers, an enhancement of mesoderm and ectoderm differentiation with a concurrent loss of endoderm lineage specification was observed with Y-27632 treatment. Evidence would suggest that this effect is in part mediated through TGF-ß and SMAD signaling as ROCK-inhibited cells displayed aberrant SMAD activation and did not return to a 'ground' state after the inhibition had been removed. CONCLUSIONS/SIGNIFICANCE: Given this data and the fact that only a partial rescue of normal differentiation capacity occurred when ROCK inhibition was alleviated, the effect of ROCK inhibition on the differentiation capacity of pluripotent cell populations should be further examined to elucidate the role of the Rho-ROCK pathway in early cellular 'fate' decision making processes.


Assuntos
Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco de Carcinoma Embrionário/patologia , Quinases Associadas a rho/antagonistas & inibidores , Amidas/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Endoderma/efeitos dos fármacos , Endoderma/patologia , Humanos , Camundongos , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/patologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tretinoína/farmacologia , Quinases Associadas a rho/metabolismo
6.
J Cell Physiol ; 226(12): 3190-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21321941

RESUMO

The identity of embryonic stem cells (ESCs) is controlled by a set of pluripotency genes, including Oct4, Sox2, Nanog, and Fgf4. How their expression is repressed during differentiation and reactivated during reprogramming is largely unknown. Here, using mouse ESCs as well as F9 and P19 cells (mouse embryonal carcinoma cell lines, P19 being considered further differentiated than F9 cells) as models, we found that HDAC inhibitors elevated Fgf4 expression in P19 cells, but reduced it in F9 cells. We also observed that HDAC inhibitors enhanced the expression of Fgf4 and a subset of pluripotency genes in differentiated ESCs, but reduced their expression in undifferentiated and less differentiated ESCs. Mechanistically, we observed more HDAC1 recruitment and a weaker association of histone 4 lysine 5 acetylation at the Fgf4 enhancer in P19 cells compared to F9 cells. Additionally, we demonstrated the interaction between Sox2 and HDAC1 both in vitro and in vivo, implicating a possible role for Sox2 in the recruitment of HDAC1 to the Fgf4 enhancer. We also found that Nanog bound to the Fgf4 enhancer, and this binding was stronger in F9 cells, indicating the involvement of Nanog in the regulation of Fgf4 expression in undifferentiated and less differentiated pluripotent stem cells. This study uncovers an important role of HDAC1 and histone modifications in the repression of Fgf4 and perhaps other pluripotency genes during ESC differentiation. Our results also suggest that HDAC inhibitors may promote reprogramming partially through activating pluripotency genes at some intermediate stages.


Assuntos
Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Fator 4 de Crescimento de Fibroblastos/metabolismo , Histona Desacetilase 1/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Acetilação , Animais , Sítios de Ligação , Linhagem Celular , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco Embrionárias/enzimologia , Elementos Facilitadores Genéticos , Fator 4 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Ácidos Hidroxâmicos/farmacologia , Camundongos , Proteína Homeobox Nanog , Células-Tronco Pluripotentes/enzimologia , Interferência de RNA , Fatores de Transcrição SOXB1/metabolismo , Fatores de Tempo , Transfecção , Ácido Valproico/farmacologia
7.
Arch Pharm Res ; 33(7): 1117-25, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20661723

RESUMO

Nanog is a key determinant that maintains self-renewal and pluripotency of embryonic stem cells and represses their differentiation to endoderm. In this study, we examined the regulation of Nanog expression by phosphoinositide-3-kinase (PI3K)/Akt pathway during retinoic acid (RA)-induced differentiation of F9 embryonic carcinoma cells. Nanog protein expression was transiently upregulated up to 6 h after RA treatment and then declined. In agreement, a murine Nanog promoter reporter assay revealed that promoter activity increased during early stage of differentiation, but decreased when F9 cells became fully differentiated. RA treatment of F9 cells also led to a transient and parallel increase in both Akt and glycogen synthase kinase 3beta phosphorylations. Nanog expression was diminished in the early stage by LY294002, a PI3K inhibitor, but was not affected in the late stage despite considerable inhibition of Akt phosphorylation and endoderm marker expression by the inhibitor. These data suggest that RA-induced PI3K/Akt activation in the early stage of differentiation is required for Nanog expression, which becomes independent of PI3K/Akt signaling once the differentiation is established. Thus, Nanog expression appears to be differently regulated by the PI3K/Akt pathway depending on differentiation stage.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco de Carcinoma Embrionário/enzimologia , Proteínas de Homeodomínio/biossíntese , Fosfatidilinositol 3-Quinase/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Cromonas/farmacologia , Células-Tronco de Carcinoma Embrionário/citologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Camundongos , Morfolinas/farmacologia , Proteína Homeobox Nanog , Inibidores de Fosfoinositídeo-3 Quinase , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Tretinoína/farmacologia
8.
Cell Biochem Funct ; 27(7): 468-72, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19711443

RESUMO

While analyzing the role of c-Jun NH(2)-terminal kinase (JNK) in neurogenesis in P19 embryonal carcinoma cells, we noticed that treatment with SP600125, a JNK inhibitor, increased the cell size markedly. SP600125-induced enlargement of P19 cells was time- and dose-dependent. The increased cell size in response to SP600125 was also detected in B6mt-1 embryonic stem cells. SP600125 treatment inhibited cell growth and increased DNA contents, indicating the inhibition of cell proliferation resulting from endoreduplication. Concurrently, the gene expression of p21, a regulator of G2/M arrest as well as G1 arrest, was increased in cells treated with SP600125. The increased cell size in response to SP600125 was detected even in P19 cells treated with colcemide, an inhibitor of cell cycle progression at the metaphase. The present study suggests that treatment with SP600125 progresses the cell cycle, skipping cytokinesis in P19 cells.


Assuntos
Antracenos/farmacologia , Crescimento Celular/efeitos dos fármacos , Citocinese/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco de Carcinoma Embrionário/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Citocinese/genética , Demecolcina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Camundongos
9.
Am J Physiol Heart Circ Physiol ; 295(2): H708-16, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18552163

RESUMO

TNNI3K is a new cardiac-specific MAP kinase whose gene is localized to 1p31.1 and that belongs to a tyrosine kinase-like branch in the kinase tree of the human genome. In the present study we investigated the role of TNNI3K in the cardiac myogenesis process and in the repair of ischemic injury. Pluripotent P19CL6 cells with or without transfection by pcDNA6-TNNI3K plasmid were used to induce differentiation into beating cardiomyocytes. TNNI3K promoted the differentiation process, judging from the increasing beating mass and increased number of alpha-actinin-positive cells. TNNI3K improved cardiac function by enhancing beating frequency and increasing the contractile force and epinephrine response of spontaneous action potentials without an increase of the single-cell size. TNNI3K suppressed phosphorylation of cardiac troponin I, annexin-V(+) cells, Bax protein, and p38/JNK-mediated apoptosis. Intramyocardial administration of TNNI3K-overexpressing P19CL6 cells in mice with myocardial infarction improved cardiac performance and attenuated ventricular remodeling compared with injection of wild-type P19CL6 cells. In conclusion, our study clearly indicates that TNNI3K promotes cardiomyogenesis, enhances cardiac performance, and protects the myocardium from ischemic injury by suppressing p38/JNK-mediated apoptosis. Therefore, modulation of TNNI3K activity would be a useful therapeutic approach for ischemic cardiac disease.


Assuntos
Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Desenvolvimento Muscular , Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/enzimologia , Células-Tronco Pluripotentes/enzimologia , Actinina/metabolismo , Potenciais de Ação , Animais , Anexina A5/metabolismo , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Células-Tronco de Carcinoma Embrionário/patologia , Células-Tronco de Carcinoma Embrionário/transplante , Epinefrina/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Contração Miocárdica , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/transplante , Fosforilação , Células-Tronco Pluripotentes/patologia , Células-Tronco Pluripotentes/transplante , Proteínas Serina-Treonina Quinases , Transplante de Células-Tronco , Transfecção , Troponina I/metabolismo , Função Ventricular Esquerda , Remodelação Ventricular , Proteína X Associada a bcl-2/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Mol Biol Cell ; 19(4): 1548-60, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18256293

RESUMO

The DNA repair enzyme telomerase maintains chromosome stability by ensuring that telomeres regenerate each time the cell divides, protecting chromosome ends. During onset of neuroectodermal differentiation in P19 embryonal carcinoma (EC) cells three independent techniques (Southern blotting, Q-FISH, and Q-PCR) revealed a catastrophic reduction in telomere length in nestin-expressing neuronal precursors even though telomerase activity remained high. Overexpressing telomerase protein (mTERT) prevented telomere collapse and the neuroepithelial precursors produced continued to divide, but deaggregated and died. Addition of FGF-2 prevented deaggregation, protected the precursors from the apoptotic event that normally accompanies onset of terminal neuronal differentiation, allowed them to evade senescence, and enabled completion of morphological differentiation. Similarly, primary embryonic stem (ES) cells overexpressing mTERT also initiated neuroectodermal differentiation efficiently, acquiring markers of neuronal precursors and mature neurons. ES precursors are normally cultured with FGF-2, and overexpression of mTERT alone was sufficient to allow them to evade senescence. However, when FGF-2 was removed in order for differentiation to be completed most neural precursors underwent apoptosis indicating that in ES cells mTERT is not sufficient allow terminal differentiation of ES neural precursors in vitro. The results demonstrate that telomerase can potentiate the transition between pluripotent stem cell and committed neuron in both EC and ES cells.


Assuntos
Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco de Carcinoma Embrionário/patologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Neurônios/citologia , Neurônios/enzimologia , Telomerase/metabolismo , Animais , Apoptose , Sequência de Bases , Agregação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/enzimologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Telomerase/genética , Telômero/genética
11.
Int J Biochem Cell Biol ; 40(5): 1043-54, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18191611

RESUMO

The Oct-4 gene encodes a transcription factor that plays an important role in maintaining the pluripotent state of embryonic stem cells and may prevent expression of genes activated during differentiation. Although its role in maintaining embryonic stem cell pluripotency is well established, there is still little known about the binding partners that regulate its function. To identify proteins that control Oct-4 function, we used affinity chromatography on immobilized Oct-4 (POU) together with MALDI-TOF (matrix-assisted laser-desorption ionization-time-of-flight) MS (mass spectrometry) and isolated a novel Oct-4-interacting protein, pyruvate kinase type M2 (PKM2 or M2-PK). PKM2 is an isozyme of pyruvate kinase that is specifically expressed in proliferating cells, such as embryonic stem cells, embryonic carcinoma cells, as well as cancer cells. Oct-4 and PKM2 were co-affinity precipitated from cell extracts, and glutathione S-transferase pull-down assays revealed that the POU DNA binding domain of Oct-4 was required for interaction with PKM2. In addition, the C-terminal domain of PKM2 (amino acids 307-531) was involved in binding to Oct-4. Moreover, ectopic expression of the PKM2 enhanced Oct-4-mediated transcription. These observations indicate that the transactivation potential of the Oct-4 transcription factor is positively modulated by PKM2.


Assuntos
Fator 3 de Transcrição de Octâmero/metabolismo , Piruvato Quinase/metabolismo , Ativação Transcricional , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/metabolismo , Humanos , Camundongos , Fator 3 de Transcrição de Octâmero/química , Estrutura Terciária de Proteína , Piruvato Quinase/química
12.
Cell Signal ; 20(3): 506-17, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18160257

RESUMO

Epithelial-to-mesenchymal transitions (EMTs) play key roles in the normal development of an organism as well as its demise following the metastasis of a malignant tumour. An EMT during early mouse development results in the differentiation of primitive endoderm into the parietal endoderm that forms part of the parietal yolk sac. In the embryo, primitive endoderm develops from cells in the inner cell mass, but the signals that instruct these cells to become specified and adopt an epithelial fate are poorly understood. The mouse F9 teratocarcinoma cell line, a model that can recapitulate the in vivo primitive to parietal endoderm EMT, has been used extensively to elucidate the signalling cascades involved in extraembryonic endoderm differentiation. Here, we identified Wnt6 as a gene up-regulated in F9 cells in response to RA and show that Wnt6 expressing cells or cells exposed to Wnt6 conditioned media form primitive endoderm. Wnt6 induction of primitive endoderm is accompanied by beta-catenin and Snail1 translocation to the nucleus and the appearance of cytokeratin intermediate filaments. Attenuating glycogen synthase kinase 3 activity using LiCl gave similar results, but the fact that cells de-differentiate when LiCl is removed reveals that other signalling pathways are required to maintain cells as primitive endoderm. Finally, Wnt6-induced primitive endodermal cells were tested to determine their competency to complete the EMT and differentiate into parietal endoderm. Towards that end, results show that up-regulating protein kinase A activity is sufficient to induce markers of parietal endoderm. Together, these findings indicate that undifferentiated F9 cells are responsive to canonical Wnt signalling, which negatively regulates glycogen synthase kinase 3 activity leading to the epithelialization and specification of primitive endoderm competent to receive additional signals required for EMT. Considering the ability of F9 cells to mimic an in vivo EMT, the identification of this Wnt6-beta-catenin-Snail signalling cascade has broad implications for understanding EMT mechanisms in embryogenesis and metastasis.


Assuntos
Padronização Corporal , Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/metabolismo , Endoderma/metabolismo , Células Epiteliais/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Padronização Corporal/efeitos dos fármacos , Bucladesina/farmacologia , Células COS , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Chlorocebus aethiops , Meios de Cultivo Condicionados/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário/enzimologia , Células-Tronco de Carcinoma Embrionário/patologia , Endoderma/efeitos dos fármacos , Endoderma/enzimologia , Endoderma/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Regulação da Expressão Gênica no Desenvolvimento , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Filamentos Intermediários/metabolismo , Cloreto de Lítio/farmacologia , Camundongos , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , Transfecção , Tretinoína/farmacologia , Regulação para Cima , Proteínas Wnt/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...