Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Commun ; 10(1): 967, 2019 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-30814500

RESUMO

The stem cell pluripotency factor Oct4 serves a critical protective role during atherosclerotic plaque development by promoting smooth muscle cell (SMC) investment. Here, we show using Myh11-CreERT2 lineage-tracing with inducible SMC and pericyte (SMC-P) knockout of Oct4 that Oct4 regulates perivascular cell migration and recruitment during angiogenesis. Knockout of Oct4 in perivascular cells significantly impairs perivascular cell migration, increases perivascular cell death, delays endothelial cell migration, and promotes vascular leakage following corneal angiogenic stimulus. Knockout of Oct4 in perivascular cells also impairs perfusion recovery and decreases angiogenesis following hindlimb ischemia. Transcriptomic analyses demonstrate that expression of the migratory gene Slit3 is reduced following loss of Oct4 in cultured SMCs, and in Oct4-deficient perivascular cells in ischemic hindlimb muscle. Together, these results provide evidence that Oct4 plays an essential role within perivascular cells in injury- and hypoxia-induced angiogenesis.


Assuntos
Neovascularização Fisiológica , Fator 3 de Transcrição de Octâmero/deficiência , Células-Tronco Pluripotentes/metabolismo , Animais , Morte Celular , Linhagem da Célula , Movimento Celular , Células Cultivadas , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Feminino , Membro Posterior , Isquemia/metabolismo , Isquemia/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/metabolismo , Neovascularização Patológica , Fator 3 de Transcrição de Octâmero/genética , Pericitos/metabolismo , Pericitos/patologia , Células-Tronco Pluripotentes/patologia
2.
Med Sci Monit ; 25: 1214-1219, 2019 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-30763293

RESUMO

BACKGROUND POU5F1B, serving as a carcinogen, participates in radiosensitivity of several tumors. However, in esophageal cancer, its potential mechanism and function in regulating radiosensitivity remain unclear. MATERIAL AND METHODS The expression level of POU5F1B was detected in plasma of esophageal tumor patients and cancer cell lines. The effect of POU5F1B knockdown on cell proliferation and colony formation was determined using CCK-8 assay and colony formation assay. Cell apoptosis rate was detected by flow cytometry. RESULTS POU5F1B expression level declined after radiotherapy in the plasma of esophageal cancer patients (p=0.025). Compared with HEEPIC, the level of POU5F1B was upregulated in ECA109 (p<0.01), ECA9706 (p<0.01), KYSE410 (p<0.01), and KYSE510 (p=0.036). The silencing of POU5F1B played a role in inhibiting colony formation. After radiotherapy, the apoptosis rates in the ECA109 with 4Gy si-POU5F1B group and 4Gy si-NC group were 39.1±0.1% and 35.3±0.1%, respectively (p=0.0193). The rate was 21.00±0.1 and 29.1±0.1% (p<0.0072) in the si-NC group and si-POU5F1B group, respectively. For proliferation rate, 4Gy si-POU5F1B ECA109 performed better than 4Gy si-NC. CONCLUSIONS Radiotherapy contributed to the decline in the expression level of POU5F1B in plasma, which was upregulated in ECA109, ECA9706, KYSE410, and KYSE510, but not in HEEPIC. The knockdown of POU5F1B increased the radiosensitivity of esophageal cancer cell lines.


Assuntos
Neoplasias Esofágicas/radioterapia , Fator 3 de Transcrição de Octâmero/deficiência , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Movimento Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Regulação para Baixo/efeitos da radiação , Neoplasias Esofágicas/sangue , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Técnicas de Silenciamento de Genes/métodos , Genes myc , Humanos , Fator 3 de Transcrição de Octâmero/sangue , Fator 3 de Transcrição de Octâmero/genética , RNA Longo não Codificante/genética , Tolerância a Radiação , Regulação para Cima/efeitos da radiação
3.
Sci Rep ; 7: 46312, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28406185

RESUMO

Oct4A is a master regulator of self-renewal and pluripotency in embryonic stem cells. It is a well-established marker for cancer stem cell (CSC) in malignancies. Recently, using a loss of function studies, we have demonstrated key roles for Oct4A in tumor cell survival, metastasis and chemoresistance in in vitro and in vivo models of ovarian cancer. In an effort to understand the regulatory role of Oct4A in tumor biology, we employed the use of an ovarian cancer shRNA Oct4A knockdown cell line (HEY Oct4A KD) and a global mass spectrometry (MS)-based proteomic analysis to investigate novel biological targets of Oct4A in HEY samples (cell lysates, secretomes and mouse tumor xenografts). Based on significant differential expression, pathway and protein network analyses, and comprehensive literature search we identified key proteins involved with biologically relevant functions of Oct4A in tumor biology. Across all preparations of HEY Oct4A KD samples significant alterations in protein networks associated with cytoskeleton, extracellular matrix (ECM), proliferation, adhesion, metabolism, epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs) and drug resistance was observed. This comprehensive proteomics study for the first time presents the Oct4A associated proteome and expands our understanding on the biological role of this stem cell regulator in carcinomas.


Assuntos
Reprogramação Celular/genética , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/deficiência , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Animais , Biomarcadores , Linhagem Celular Tumoral , Biologia Computacional/métodos , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Ovarianas/patologia , Proteoma/metabolismo , Proteômica/métodos , Reprodutibilidade dos Testes , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nature ; 540(7631): 114-118, 2016 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-27905446

RESUMO

Germ-cell tumours (GCTs) are derived from germ cells and occur most frequently in the testes. GCTs are histologically heterogeneous and distinctly curable with chemotherapy. Gains of chromosome arm 12p and aneuploidy are nearly universal in GCTs, but specific somatic genomic features driving tumour initiation, chemosensitivity and progression are incompletely characterized. Here, using clinical whole-exome and transcriptome sequencing of precursor, primary (testicular and mediastinal) and chemoresistant metastatic human GCTs, we show that the primary somatic feature of GCTs is highly recurrent chromosome arm level amplifications and reciprocal deletions (reciprocal loss of heterozygosity), variations that are significantly enriched in GCTs compared to 19 other cancer types. These tumours also acquire KRAS mutations during the development from precursor to primary disease, and primary testicular GCTs (TGCTs) are uniformly wild type for TP53. In addition, by functional measurement of apoptotic signalling (BH3 profiling) of fresh tumour and adjacent tissue, we find that primary TGCTs have high mitochondrial priming that facilitates chemotherapy-induced apoptosis. Finally, by phylogenetic analysis of serial TGCTs that emerge with chemotherapy resistance, we show how TGCTs gain additional reciprocal loss of heterozygosity and that this is associated with loss of pluripotency markers (NANOG and POU5F1) in chemoresistant teratomas or transformed carcinomas. Our results demonstrate the distinct genomic features underlying the origins of this disease and associated with the chemosensitivity phenotype, as well as the rare progression to chemoresistance. These results identify the convergence of cancer genomics, mitochondrial priming and GCT evolution, and may provide insights into chemosensitivity and resistance in other cancers.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Genoma Humano/genética , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/genética , Apoptose , Progressão da Doença , Evolução Molecular , Exoma/genética , Genômica , Humanos , Perda de Heterozigosidade , Masculino , Mitocôndrias/metabolismo , Mutação , Proteína Homeobox Nanog/deficiência , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Neoplasias Embrionárias de Células Germinativas/metabolismo , Neoplasias Embrionárias de Células Germinativas/patologia , Fator 3 de Transcrição de Octâmero/deficiência , Filogenia , Proteínas Proto-Oncogênicas p21(ras)/genética , Teratoma/genética , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia , Transcriptoma/genética , Proteína Supressora de Tumor p53/genética
5.
PLoS One ; 7(8): e43628, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22928007

RESUMO

The cancer stem cell (CSC) model posits the presence of a small number of CSCs in the heterogeneous cancer cell population that are ultimately responsible for tumor initiation, as well as cancer recurrence and metastasis. CSCs have been isolated from a variety of human cancers and are able to generate a hierarchical and heterogeneous cancer cell population. CSCs are also resistant to conventional chemo- and radio-therapies. Here we report that ionizing radiation can induce stem cell-like properties in heterogeneous cancer cells. Exposure of non-stem cancer cells to ionizing radiation enhanced spherogenesis, and this was accompanied by upregulation of the pluripotency genes Sox2 and Oct3/4. Knockdown of Sox2 or Oct3/4 inhibited radiation-induced spherogenesis and increased cellular sensitivity to radiation. These data demonstrate that ionizing radiation can activate stemness pathways in heterogeneous cancer cells, resulting in the enrichment of a CSC subpopulation with higher resistance to radiotherapy.


Assuntos
Raios gama , Células-Tronco Neoplásicas/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/deficiência , Fator 3 de Transcrição de Octâmero/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética
6.
Histochem Cell Biol ; 134(2): 197-204, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20532795

RESUMO

The biological basis for manifestation of chemotherapy resistance in metastatic testicular germ cell tumors (GCT) remains obscure and is of particular clinical interest. In nonseminomatous GCT (NSGCT) the pluripotent embryonal carcinoma (EC) cells are the precursors of the manifold differentiated structures but also drive the malignant growth. They are known to be hypersensitive towards DNA-damaging agents and to express the embryonal transcription factor OCT4. We recently characterized EC cells that lack OCT4 expression and show cisplatin resistance. In the present, immunohistochemical study we analyzed the composition of NSGCT with the focus on such OCT4-negative EC cells using a NSGCT xenograft model as well as patient-derived NSGCT samples. In the xenograft model, the cisplatin-sensitive cell line H12.1 gives rise to xenografts where EC structures are mainly composed of OCT4-positive cells, whereas xenografts from the resistant cell line 1411HP exclusively comprise OCT4-negative EC areas. We found that post-chemotherapy residual metastatic tumors of patients can be comprised of exclusively OCT4-negative EC, whereas the matched testicular primary tumor harbors OCT4-positive EC. Thorough histological analyses revealed a few examples of such OCT4-negative EC cells also in the testicular primary tumor as well as in xenografts from the cisplatin-sensitive NSGCT-cell line. For these cells we propose an identity as early extraembryonal progenitor cells directly derived from OCT4-expressing EC cells. This challenges the use of the term EC cell. The data also support our hypothesis that malignant growth of resistant NSGCT may be driven by this cell type.


Assuntos
Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Células-Tronco de Carcinoma Embrionário/patologia , Neoplasias Embrionárias de Células Germinativas/patologia , Fator 3 de Transcrição de Octâmero/deficiência , Animais , Linhagem Celular Tumoral , Feminino , Histologia , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Células-Tronco , Transplante Heterólogo/patologia
7.
Blood ; 114(1): 60-3, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-19321862

RESUMO

In embryonic stem cells, Oct-4 concentration is critical in determining the development of endoderm, mesoderm, and trophectoderm. Although Oct-4 expression is essential for mesoderm development, it is unclear whether it has a role in the development of specific mesodermal tissues. In this study, we have examined the importance of Oct-4 in the generation of hematopoietic cells using an inducible Oct-4 ESC line. We demonstrate that Oct-4 has a role in supporting hematopoiesis after specifying brachyury-positive mesoderm. When we suppressed Oct-4 expression before or after mesoderm specification, no hematopoietic cells are detected. However, hematopoiesis can be rescued in the absence of Oct-4 after mesoderm specification if the essential hematopoietic transcription factor stem cell leukemia is expressed. Our results suggest that, for hematopoiesis to occur, Oct-4 is required for the initial specification of mesoderm and subsequently is required for the development of hematopoietic cells from uncommitted mesoderm.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Hematopoese/fisiologia , Fator 3 de Transcrição de Octâmero/deficiência , Proteínas Proto-Oncogênicas/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Hematopoese/genética , Mesoderma/embriologia , Mesoderma/fisiologia , Camundongos , Fator 3 de Transcrição de Octâmero/antagonistas & inibidores , Fator 3 de Transcrição de Octâmero/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Transfecção
8.
Dev Biol ; 315(1): 1-17, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18215655

RESUMO

Pou5f1/Oct4 is a transcription factor required for pluripotency of embryonic stem cells in mammals. Zebrafish pou5f1 deficient maternal and zygotic spiel ohne grenzen (MZspg) mutant embryos develop severe gastrulation defects, are dorsalized, and defective in endoderm formation. Here we analyze in detail gastrulation defects, which are manifested by a severe delay in epiboly progression. All three embryonic lineages in MZspg embryos behave abnormally during epiboly: the yolk cell forms an altered array of cortical microtubules and F-Actin, with large patches of microtubule free areas; the enveloping layer (EVL) is delayed in the coordinated cell shape changes of marginal cells, that may be mediated by F-Actin; the deep layer cells (DEL), forming the embryo proper, are non-autonomously affected in their motility and do not enter the space opening by epiboly of the EVL. Analysis of adhesiveness as well as high resolution in vivo time lapse image analysis of DEL cells suggests changed adhesive properties and inability to migrate properly on EVL and yolk syncytial layer (YSL) surfaces. Our data further reveal that during epiboly the EVL may actively probe the YSL by filopodia formation, rather than just being passively pulled vegetalwards. Our findings on the effect of Pou5f1 on cell behavior may be relevant to understand stem cell behavior and tumorigenesis involving Pou5f1.


Assuntos
Linhagem da Célula , Citoesqueleto/fisiologia , Mutação , Fator 3 de Transcrição de Octâmero/deficiência , Peixe-Zebra/embriologia , Actinas/metabolismo , Animais , Adesão Celular/fisiologia , Agregação Celular , Movimento Celular , Tamanho Celular , Transplante de Células , Embrião não Mamífero , Feminino , Gástrula/anormalidades , Proteínas de Fluorescência Verde/metabolismo , Microinjeções , Microtúbulos/metabolismo , Fator 3 de Transcrição de Octâmero/genética , RNA Mensageiro/genética , Transcrição Gênica , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
9.
FASEB J ; 21(9): 2020-32, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17356004

RESUMO

Stem cells convert graded stimuli into all-or-nothing cell-fate responses. We investigated how embryonic stem cells (ESCs) convert leukemia inhibitory factor (LIF) concentration into an all-or-nothing cell-fate decision (self-renewal). Using a combined experimental/computational approach we demonstrate unexpected switch-like (on/off) signaling in response to LIF. This behavior emerges over time due to a positive feedback loop controlling transcriptional expression of LIF signaling pathway components. The autoregulatory loop maintains robust pathway responsiveness ("on") at sufficient concentrations of exogenous LIF, while autocrine signaling and low concentrations of exogenous LIF cause ESCs to adopt the weakly responsive ("off") state of differentiated cells. We demonstrate that loss of ligand responsiveness is reversible and precedes loss of the ESC transcription factors Oct4 and Nanog, suggesting an early step in the hierarchical control of differentiation. While endogenously produced ligands were insufficient to sustain the "on" state, they buffer it, influencing the timing of differentiation. These results demonstrate a novel switch-like behavior, which establishes the LIF threshold for ESC self-renewal.


Assuntos
Comunicação Autócrina/fisiologia , Células-Tronco Embrionárias/citologia , Retroalimentação Fisiológica/fisiologia , Fator Inibidor de Leucemia/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Receptor gp130 de Citocina/biossíntese , Receptor gp130 de Citocina/genética , Proteínas de Ligação a DNA/fisiologia , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/fisiologia , Fator Inibidor de Leucemia/fisiologia , Camundongos , Modelos Biológicos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/deficiência , Fator 3 de Transcrição de Octâmero/fisiologia , Receptores de OSM-LIF/biossíntese , Receptores de OSM-LIF/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA