Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Genet ; 14: 1320185, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38152652

RESUMO

Ovarian cancer is one of the female reproductive system tumors. Chemotherapy is used for advanced ovarian cancer patients; however, drug resistance is a pivotal cause of chemotherapeutic failure. Hence, it is critical to explore the molecular mechanisms of drug resistance of ovarian cancer cells and to ameliorate chemoresistance. Noncoding RNAs (ncRNAs) have been identified to critically participate in drug sensitivity in a variety of human cancers, including ovarian cancer. Among ncRNAs, circRNAs sponge miRNAs and prevent miRNAs from regulation of their target mRNAs. CircRNAs can interact with DNA or proteins to modulate gene expression. In this review, we briefly describe the biological functions of circRNAs in the development and progression of ovarian cancer. Moreover, we discuss the underneath regulatory molecular mechanisms of circRNAs on governing drug resistance in ovarian cancer. Furthermore, we mention the novel strategies to overcome drug resistance via targeting circRNAs in ovarian cancer. Due to that circRNAs play a key role in modulation of drug resistance in ovarian cancer, targeting circRNAs could be a novel approach for attenuation of chemoresistance in ovarian cancer.

2.
Cancer Control ; 30: 10732748231206929, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37924202

RESUMO

PURPOSE: This study aims to determine the optimal cut-off value of Ki-67 to better predict the recurrence of early low-risk endometrial cancer (EC). METHODS: Seven hundred and forty-eight patients diagnosed with low-risk EC from West China Second Hospital of Sichuan University and the First Affiliated Hospital of Chongqing Medical University were retrospectively analyzed. The receiver operating characteristic curve (ROC) and Youden index were used to calculate the optimal cut-off value of Ki-67 expression. The clinicopathological indexes between two groups divided by cut-off value of Ki-67 were compared. The univariate and multivariate regression analyses were performed to investigate risk factors connected to the recurrence of early low-risk EC. The survival analysis was shown in Kaplan-Meier curve. RESULT: Thirty-three patients were detected with tumor recurrence after primary surgery (4.4%); 33% was the optimal cut-off value of the Ki-67 index. A high Ki-67 was significantly associated with age (P = .002), myometrial invasion (P < .001), and the expression of P53 (P = .007). The multivariate regression analysis verified that Ki67 ≥ 33% was an independent prognostic factor for predicting recurrence. The recurrence-free survival (RFS) and the overall survival (OS) in high Ki-67 group was significantly lower than that in low Ki-67 group (P < .001 and P = .029, respectively). The prognostic values of ER, PR, and P53 in combination with Ki-67 were superior to each single predictor. CONCLUSIONS: The optimal cut-off value of Ki-67 for predicting recurrence is 33%, which quantitatively defines the specific value of Ki-67 that causes high-risk recurrence in early low-risk EC.


Assuntos
Neoplasias do Endométrio , Proteína Supressora de Tumor p53 , Humanos , Feminino , Antígeno Ki-67/metabolismo , Prognóstico , Estudos Retrospectivos , Recidiva Local de Neoplasia
3.
J Clin Med ; 12(3)2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36769874

RESUMO

BACKGROUND: Combining traditional clinical parameters with neuroendocrine markers to construct a nomogram model to predict the postoperative recurrence of neuroendocrine carcinoma of cervix (NECC). METHODS: A total of 257 patients were included in this study. Univariate and multivariate Cox regression analyses were used to establish a nomogram model in the training cohorts, which was further validated in the validation cohorts. The calibration curve was used to conduct the internal and external verification of the model. RESULTS: Overall, 41 relapse cases were observed in the training (23 cases) and validation (18 cases) cohorts. The univariate analysis preliminarily showed that FIGO stage, stromal invasion, nerve invasion, lymph vascular space invasion, lymph node involvement, cervical-uterine junction invasion and CgA were correlated with NECC recurrence. The multivariate analysis further confirmed that FIGO stage (p = 0.023), stromal invasion (p = 0.002), lymph vascular space invasion (p = 0.039) and lymph node involvement (p = 0.00) were independent risk factors for NECC recurrence, which were ultimately included in the nomogram model. In addition, superior consistency indices were demonstrated in the training (0.863, 95% CI 0.784-0.942) and validation (0.884, 95% CI 0.758-1.010) cohorts. CONCLUSIONS: The established nomogram model combining traditional clinical parameters with neuroendocrine markers can reliably and accurately predict the recurrence risks in NECC patients.

4.
Proc Natl Acad Sci U S A ; 117(46): 28763-28774, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33139573

RESUMO

The molecular mechanisms by which receptor tyrosine kinases (RTKs) and heterotrimeric G proteins, two major signaling hubs in eukaryotes, independently relay signals across the plasma membrane have been extensively characterized. How these hubs cross-talk has been a long-standing question, but answers remain elusive. Using linear ion-trap mass spectrometry in combination with biochemical, cellular, and computational approaches, we unravel a mechanism of activation of heterotrimeric G proteins by RTKs and chart the key steps that mediate such activation. Upon growth factor stimulation, the guanine-nucleotide exchange modulator dissociates Gαi•ßγ trimers, scaffolds monomeric Gαi with RTKs, and facilitates the phosphorylation on two tyrosines located within the interdomain cleft of Gαi. Phosphorylation triggers the activation of Gαi and inhibits second messengers (cAMP). Tumor-associated mutants reveal how constitutive activation of this pathway impacts cell's decision to "go" vs. "grow." These insights define a tyrosine-based G protein signaling paradigm and reveal its importance in eukaryotes.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Células COS , Chlorocebus aethiops , Receptores ErbB/metabolismo , Células HEK293 , Células HeLa , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Humanos , Fosforilação , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais , Tirosina/metabolismo
5.
Int J Med Sci ; 14(13): 1410-1417, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29200955

RESUMO

The present study evaluated the prognostic value of the epidermal growth factor receptor (EGFR) mutation status, and excision repair cross-complementation group 1 (ERCC1) and thymidylate synthase (TS) expression following intercalated tyrosine kinase inhibitor (TKI) therapy and platinum- and pemetrexed-based chemotherapies (subsequent second-line treatment) for patients with adenocarcinoma non-small-cell lung cancer (AC-NSCLC). In total, 131 patients with AC-NSCLC were enrolled. The EGFR mutation status and ERCC1 and TS expression were evaluated through direct DNA sequencing and immunohistochemical analyses, respectively. The EGFR mutation status and ERCC1 and TS expression were the significant predictors of clinical outcomes. The EGFR mutation status was the main outcome predictor for overall survival (OS) benefits in the overall population. Further exploratory ERCC1 and TS expression analyses were conducted to provide additional insights. Low TS expression was predictive of improved OS of patients with negative EGFR-mutated advanced AC-NSCLC, whereas high ERCC1 expression resulted in poor OS in patients with positive EGFR-mutated advanced AC-NSCLC. TS and ERCC1 expression levels were effective prognostic factors for negative and positive EGFR-mutated AC-NSCLC, respectively. In conclusion, the present results indicate that the EGFR mutation status and TS and ERCC1 expression can be used as the predictors of OS after subsequent second-line treatments for AC-NSCLC.


Assuntos
Adenocarcinoma/genética , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Timidilato Sintase/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Idoso , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico
6.
Int J Med Sci ; 14(8): 785-790, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28824314

RESUMO

Hypoxia is an important factor in tumor angiogenesis, metastasis, and resistance to chemotherapy or radiotherapy, and may be an indicator of poor prognosis. The transcription factor hypoxia-inducible factor 1 (HIF-1) is the key regulator of the hypoxic state. This study was designed to evaluate the prognostic value of HIF-1α expression in small cell lung cancer (SCLC). Forty-three paraffin-embedded biopsy materials were examined using immunohistochemistry. Our results indicated that the expression of HIF-1α was high in males, and patients with poor Eastern Cooperative Oncology Group (ECOG) performance status and metastases. To elucidate the prognostic value of HIF-1α expression, Kaplan-Meier analysis was carried out and the results showed that patients with high HIF-1α expression had a poorer prognosis than patients with low expression of HIF-1α. After adjusting clinical parameters by the Cox proportional hazards model, our results demonstrated that high HIF-1α expression is an independent prognostic factor for SCLC with a 39.2-fold risk of death (p<0.003). In conclusion, we have provided evidence that HIF-1α expression has significant value in predicting survival of patients with SCLC and is an independent prognostic factor beyond ECOG performance and metastasis status.


Assuntos
Biomarcadores Tumorais/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neovascularização Patológica/genética , Carcinoma de Pequenas Células do Pulmão/genética , Idoso , Idoso de 80 Anos ou mais , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Neovascularização Patológica/diagnóstico , Neovascularização Patológica/patologia , Prognóstico , Modelos de Riscos Proporcionais , Carcinoma de Pequenas Células do Pulmão/mortalidade , Carcinoma de Pequenas Células do Pulmão/patologia
7.
Mol Biol Cell ; 25(22): 3654-71, 2014 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-25187647

RESUMO

A long-standing issue in the field of signal transduction is to understand the cross-talk between receptor tyrosine kinases (RTKs) and heterotrimeric G proteins, two major and distinct signaling hubs that control eukaryotic cell behavior. Although stimulation of many RTKs leads to activation of trimeric G proteins, the molecular mechanisms behind this phenomenon remain elusive. We discovered a unifying mechanism that allows GIV/Girdin, a bona fide metastasis-related protein and a guanine-nucleotide exchange factor (GEF) for Gαi, to serve as a direct platform for multiple RTKs to activate Gαi proteins. Using a combination of homology modeling, protein-protein interaction, and kinase assays, we demonstrate that a stretch of ∼110 amino acids within GIV C-terminus displays structural plasticity that allows folding into a SH2-like domain in the presence of phosphotyrosine ligands. Using protein-protein interaction assays, we demonstrated that both SH2 and GEF domains of GIV are required for the formation of a ligand-activated ternary complex between GIV, Gαi, and growth factor receptors and for activation of Gαi after growth factor stimulation. Expression of a SH2-deficient GIV mutant (Arg 1745→Leu) that cannot bind RTKs impaired all previously demonstrated functions of GIV-Akt enhancement, actin remodeling, and cell migration. The mechanistic and structural insights gained here shed light on the long-standing questions surrounding RTK/G protein cross-talk, set a novel paradigm, and characterize a unique pharmacological target for uncoupling GIV-dependent signaling downstream of multiple oncogenic RTKs.


Assuntos
Receptores ErbB/química , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Proteínas dos Microfilamentos/química , Proteínas de Transporte Vesicular/química , Sequência de Aminoácidos , Animais , Movimento Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais , Homologia Estrutural de Proteína , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
8.
Tumour Biol ; 35(5): 4719-25, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24453031

RESUMO

The cytochrome P450 1B1 (CYP1B1) gene plays a key role in the metabolism of various carcinogens. The CYP1B1 Leu432Val polymorphism leads to leucine to valine substitution at codon 432. A lot of studies have shown that the CYP1B1 Leu432Val polymorphism was associated with urinary system cancers, especially prostate cancer. However, the results were still inconclusive. In this meta-analysis, by searching online databases and references of related reviews, we identified 17 eligible studies to assess the relationship between CYP1B1 Leu432Val polymorphism and urinary system cancers, including 7,783 cancer cases and 7,238 controls. By pooling all eligible studies, we found that the CYP1B1 Leu432Val polymorphism was not associated with overall urinary system cancers. However, in subgroup analyses, we found that the variant 432Val allele significantly increased the risk of prostate cancer (Val vs. Leu, odds ratio (OR) = 1.064, 95% confidence interval (CI) 0.981-1.154; Pheterogeneity = 0.002), while no association was found for bladder cancer (Val vs. Leu, OR = 0.942, 95% CI 0.853-1.041; Pheterogeneity = 0.504). No evidence of publication bias was found (Begg's test, P = 0.053; Egger's test, P = 0.073). In conclusion, based on 17 eligible studies, we found that the CYP1B1 Leu432Val polymorphism was associated with an increased risk of prostate cancer, while no association of bladder cancer was observed.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Predisposição Genética para Doença , Polimorfismo Genético , Neoplasias da Próstata/genética , Neoplasias da Bexiga Urinária/genética , Citocromo P-450 CYP1B1 , Humanos , Masculino , Risco
9.
Cancers (Basel) ; 5(3): 959-84, 2013 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-24202329

RESUMO

We report here that the Jun dimerization protein 2 (JDP2) plays a critical role as a cofactor for the transcription factors nuclear factor-erythroid 2-related factor 2 (Nrf2) and MafK in the regulation of the antioxidants and production of reactive oxygen species (ROS). JDP2 associates with Nrf2 and MafK (Nrf2-MafK) to increase the transcription of antioxidant response element-dependent genes. Oxidative-stress-inducing reagent led to an increase in the intracellular accumulation of ROS and cell proliferation in Jdp2 knock-out mouse embryonic fibroblasts. In Jdp2-Cre mice mated with reporter mice, the expression of JDP2 was restricted to granule cells in the brain cerebellum. The induced pluripotent stem cells (iPSC)-like cells were generated from DAOY medulloblastoma cell by introduction of JDP2, and the defined factor OCT4. iPSC-like cells expressed stem cell-like characteristics including alkaline phosphatase activity and some stem cell markers. However, such iPSC-like cells also proliferated rapidly, became neoplastic, and potentiated cell malignancy at a later stage in SCID mice. This study suggests that medulloblastoma cells can be reprogrammed successfully by JDP2 and OCT4 to become iPSC-like cells. These cells will be helpful for studying the generation of cancer stem cells and ROS homeostasis.

10.
Sci Signal ; 4(192): ra64, 2011 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-21954290

RESUMO

GIV (Gα-interacting vesicle-associated protein; also known as Girdin) enhances Akt activation downstream of multiple growth factor- and G protein (heterotrimeric guanosine 5'-triphosphate-binding protein)-coupled receptors to trigger cell migration and cancer invasion. We demonstrate that GIV is a tyrosine phosphoprotein that directly binds to and activates phosphoinositide 3-kinase (PI3K). Upon ligand stimulation of various receptors, GIV was phosphorylated at tyrosine-1764 and tyrosine-1798 by both receptor and non-receptor tyrosine kinases. These phosphorylation events enabled direct binding of GIV to the amino- and carboxyl-terminal Src homology 2 domains of p85α, a regulatory subunit of PI3K; stabilized receptor association with PI3K; and enhanced PI3K activity at the plasma membrane to trigger cell migration. Tyrosine phosphorylation of GIV and its association with p85α increased during metastatic progression of a breast carcinoma. These results suggest a mechanism by which multiple receptors activate PI3K through tyrosine phosphorylation of GIV, thereby making the GIV-PI3K interaction a potential therapeutic target within the PI3K-Akt pathway.


Assuntos
Movimento Celular/fisiologia , Ativação Enzimática/fisiologia , Proteínas dos Microfilamentos/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Tirosina/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Análise de Variância , Linhagem Celular Tumoral , Cromatografia Líquida , Imunofluorescência , Humanos , Imunoprecipitação , Modelos Moleculares , Fosforilação , Espectrometria de Massas em Tandem
11.
Cell Reprogram ; 12(3): 237-47, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20698766

RESUMO

Induced pluripotent stem (iPS) cells have been generated from human somatic cells by ectopic expression of defined transcription factors. Application of this approach in human cells may have enormous potential to generate patient-specific pluripotent stem cells. However, traditional methods of reprogramming in human somatic cells involve the use of oncogenes c-MYC and KLF4, which are not applicable to clinical translation. In the present study, we investigated whether human fetal gut mesentery-derived cells (hGMDCs) could be successfully reprogrammed into induced pluripotent stem (iPS) cells by OCT4, SOX2, and NANOG alone. We used lentiviruses to express OCT4, SOX2, NANOG, in hGMDCs, then generated iPS cells that were identified by morphology, presence of pluripotency markers, global gene expression profile, DNA methylation status, capacity to form embryoid bodies (EBs), and terotoma formation. iPS cells resulting from hGMDCs were similar to human embryonic stem (ES) cells in morphology, proliferation, surface markers, gene expression, and epigenetic status of pluripotent cell-specific genes. Furthermore, these cells were able to differentiate into cell types of all three germ layers both in vitro and in vivo, as shown by EB and teratoma formation assays. DNA fingerprinting showed that the human iPS cells were derived from the donor cells, and are not a result of contamination. Our results provide proof that hGMDCs can be reprogrammed into pluripotent cells by ectopic expression of three factors (OCT4, SOX2, and NANOG) without the use of oncogenes c-MYC and KLF4.


Assuntos
Proteínas de Homeodomínio/genética , Mesentério/citologia , Fator 3 de Transcrição de Octâmero/genética , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição SOXB1/genética , Animais , Diferenciação Celular , Linhagem Celular , Citometria de Fluxo , Humanos , Cariotipagem , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos SCID , Proteína Homeobox Nanog , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Biochem Biophys Res Commun ; 344(1): 438-45, 2006 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-16603127

RESUMO

Dendritic cells (DCs) and macrophages share some stages in the development and function of antigen presentation. But it is difficult to separate them from their precursors. We used one-way ANOVA (analysis of variances) on murine expression profilings of several hematopoietic cells associated with DCs and macrophages to find the genes with great differences across the cell groups. These groups were the DCs from spleen, cultivated DCs, DC precursors, DC progenitors, DC progenitor cell lines, hematopoietic stem cell (HSC), and bone marrow-derived macrophages. The data of expression profilings were all downloaded from GEO and ArrayExpress database. After the normalization of 11 housekeeping genes across 42 arrays, we got 39 genes (44 probesets) by analysis of one-way ANOVA (Bonferroni step-down) with p values cutoff of 0.05. These genes (probesets) could separate the hematopoietic cells well by the methods of unsupervised hierarchical clustering and principal component analysis (PCA). The class prediction also indicated that these genes could separate the precursors of DC and macrophages with 20 arrays composed of 5 cell types with the same normalization. The accuracy rate of class prediction was 90% (18/20). The genes selected by one-way ANOVA included those of MHC (major histocompatibility complex) and defense of immunity, cell adhesion, chemokine or its receptors, and transcription factors. The results indicated that these 39 genes could separate precursors of DC and macrophages very clearly. It was suggested that these genes might represent some important molecules that related with the precursors of DCs and macrophages, and were worthy for further study.


Assuntos
Diferenciação Celular/genética , Células Dendríticas/citologia , Perfilação da Expressão Gênica , Macrófagos/citologia , Células-Tronco/citologia , Animais , Células Cultivadas , Análise por Conglomerados , Células Dendríticas/metabolismo , Expressão Gênica , Imunidade/genética , Macrófagos/metabolismo , Camundongos , Células-Tronco/metabolismo
13.
Neuroreport ; 15(12): 1907-12, 2004 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-15305135

RESUMO

The hypothesis that stem cells may seed cancer has emerged from the cancer stem cells concept. However, the experimental systems necessary to provide more direct evidence to support the hypothesis have been lacking. We have used fetal neural progenitor cells (hNPC) transduced with the telomerase hTERT gene to investigate the neoplastic potential of hNPCs. The hTERT-transduced line, hNPCs-G3 lost normal diploid karyotype, showed loss of contact inhibition, anchorage independence, and formed neuroblastoma-like tumours in all of 10 mice. These data suggest that hNPCs have the potential for neoplastic transformation. These data have implications for providing a novel tool to test the feasibility of new anticancer treatment strategies and raise the possibility of a risk for the use of hNPCs in cell transplantation.


Assuntos
Transformação Celular Neoplásica , Feto/citologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Animais , Southern Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Contagem de Células/métodos , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , DNA/metabolismo , Proteínas de Ligação ao GTP , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imuno-Histoquímica/métodos , Proteínas de Filamentos Intermediários/genética , Cariotipagem/métodos , Camundongos , Camundongos Nus/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Nestina , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/biossíntese , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Coloração e Rotulagem , Telômero/metabolismo , Fatores de Tempo , Transdução Genética/métodos , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
14.
Neuroreport ; 15(2): 245-9, 2004 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-15076745

RESUMO

It is necessary to expand human neural progenitor cells in vitro to obtain large numbers for research purposes and cell transplantation. A potential obstacle to in vitro expansion, however, is that neural progenitor cells have a limited replication life-span and gradually lose their differentiation potential. We report here that ectopic expression of the catalytic subunit of human telomerase (hTERT) gene in neural progenitor cells could induce telomerase activity, stabilize telomeres and extend their replicative life-spans. The telomerase-immortalized cells (hNPC-TERT) maintained the normal diploid karyotype, expressed the markers of human neural progenitor cells and meanwhile held the differentiation potential in vitro for up to 120 population doublings. This study provides a new approach for obtaining unlimited quantities of normal phenotypic and homogeneous human neural progenitor cells in vitro.


Assuntos
Técnicas de Cultura de Células/métodos , Linhagem Celular Transformada/enzimologia , Neurônios/enzimologia , Células-Tronco/enzimologia , Telomerase/genética , Transdução Genética/métodos , Animais , Biomarcadores , Domínio Catalítico/genética , Diferenciação Celular/genética , Divisão Celular/genética , Linhagem Celular Transformada/citologia , Senescência Celular/genética , Vetores Genéticos/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Cariotipagem , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/citologia , Fenótipo , Células-Tronco/citologia , Telômero/genética , Transgenes/genética
15.
Beijing Da Xue Xue Bao Yi Xue Ban ; 35(3): 266-70, 2003 Jun 18.
Artigo em Chinês | MEDLINE | ID: mdl-12914242

RESUMO

OBJECTIVE: To investigate the ability of human GFAP positive neural progenitor cell line from the subventricular zone (SVZ) to differentiate into neurons. METHODS: Real-time RT-PCR, Western blot analysis and immunocytochemistry were used to examine the expression level of the neural stem cell marker and neuronal-specific marker before and after all-trans-retinoic acid (AT-RA) induction in the GFAP positive neural progenitor cell line. Immunocytochemistry was used to examine the expression of the neuronal-specific marker after transplantation the GFAP positive neural progenitor cell line into the animal model. RESULTS: After induction, in the GFAP positive neural progenitor cell line the expression levels of the neuronal-specific marker increased, while the neural stem cell marker decreased both in mRNA and protein levels. After transplantation into animal model, the GFAP positive neural progenitor cell line could differentiate into neurons. CONCLUSION: The GFAP positive neural progenitor cell line could be induced to differentiate into neurons both in vitro and in vivo.


Assuntos
Diferenciação Celular , Proteína Glial Fibrilar Ácida/análise , Proteínas do Tecido Nervoso , Neurônios/citologia , Células-Tronco/citologia , Linhagem Celular , Proteína Glial Fibrilar Ácida/genética , Humanos , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/análise , Proteínas de Filamentos Intermediários/genética , Nestina , Neurônios/química , Fosfoproteínas Fosfatases/análise , Fosfoproteínas Fosfatases/genética , RNA Mensageiro/análise , Células-Tronco/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA