Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Biochem ; 120(6): 9831-9838, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30618186

RESUMEN

Nuclear paraspeckle assembly transcript 1 (NEAT1) has been found to be dysregulated and associated with clinical progression in various human cancers. The clinical and prognostic value of NEAT1 in nasopharyngeal carcinoma (NPC) was still controversial. The aim of our study was to provide more sufficient evidence that NEAT1 expression is correlated with overall survival in patients with NPC. NEAT1 expression was detected in NPC tissue samples, and the relationship between NEAT1 expression and clinical parameters, including prognosis, was analyzed. The meta-analysis was performed to further assess the prognostic significance of NEAT1 expression in patients with NPC. In our study, we found that the levels of NEAT1 expression were increased in NPC clinical tissue specimens, and associated with advanced M classification and clinical stages. Moreover, the Kaplan-Meier analysis suggested that the levels of NEAT1 expression were negatively associated with the overall survival of patients with NPC. Furthermore, univariate and multivariate Cox regression analyses showed that NEAT1 high-expression was an independent unfavorable prognostic factor in patients with NPC. Finally, we conducted a meta-analysis including 297 patients with NPC from the three studies, and found the pooled HR (95% confidence interval [CI]) was 1.64 (95% CI: 0.68-3.93) for the random effects model and 2.04 (95% CI: 1.42-2.95) for the fixed effect model. In conclusion, NEAT1 is a potential prognostic biomarker for NPC, but more studies are needed to further verify the prognostic value of NEAT1 in patients with NPC.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Regulación Neoplásica de la Expresión Génica , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , ARN Largo no Codificante/biosíntesis , ARN Neoplásico/biosíntesis , Adulto , Anciano , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Metaanálisis como Asunto , Persona de Mediana Edad , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/mortalidad , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/mortalidad , Tasa de Supervivencia
2.
J Transl Med ; 13: 105, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25884389

RESUMEN

BACKGROUND: Let-7a has been shown to play important roles in nasopharyngeal carcinoma (NPC) cell proliferation and apoptosis, but little is known about the function and mechanism of let-7a in nasopharyngeal carcinoma metastasis. We aimed to investigate the function and mechanism of let-7a in nasopharyngeal carcinoma metastasis and clarified the regulation of high mobility group A2 (HMGA2) by let-7a. METHODS: The expression levels of let-7a and HMGA2 were examined in NPC clinical specimens using quantitative reverse transcription-PCR (RT-qPCR). HMGA2 was confirmed as a target of let-7a through luciferase reporter assays, RT-qPCR, and Western blotting. Furthermore, the roles of let-7a and HMGA2 in regulating NPC cells biological properties including proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) process were analyzed with let-7a mimics and si-HMGA2 transfected cells. RESULTS: Our study demonstrated that let-7a was downregulated and inversely associated with the clinical stage, T classification and N classification, and HMGA2 was upregulated and directly associated with the clinical stage and N classification in patients with NPC. Moreover, there was an inverse correlation between let-7a expression and HMGA2 expression in NPC patient. In addition, HMGA2 was negatively regulated at the posttranscriptional level by let-7a via a binding site of HMGA2-3'UTR. In addition, synthetic let-7a mimics suppressed NPC cells migration, invasion and EMT process and knockdown of HMGA2 was consistent with the effects of let-7a in NPC cells. CONCLUSION: Let-7a directly downregulates HMGA2 protein expression, which suppress NPC cell migration, invasion and EMT process. Let-7a could serve as a potential diagnostic marker and therapeutic target for NPC.


Asunto(s)
Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Proteína HMGA2/metabolismo , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Anciano , Secuencia de Bases , Carcinoma , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo/genética , Retroalimentación Fisiológica , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Datos de Secuencia Molecular , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Invasividad Neoplásica , Unión Proteica/genética , Regulación hacia Arriba/genética
3.
Mol Cell Biochem ; 409(1-2): 155-62, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26183485

RESUMEN

Our previous study showed that knockdown of high-mobility group A2 (HMGA2) could suppress nasopharyngeal carcinoma (NPC) cell migration, invasion, and epithelial-mesenchymal transition (EMT) process, and HMGA2 is a direct functional target of let-7 to regulate NPC cell migration, invasion, and EMT process. However, little is known about the clinical and prognostic significance of HMGA2 protein in NPC patients. The purpose of this study is to identify the clinical and prognostic roles of HMGA2 in NPC patients. We initially analyzed the microarray data and verified mRNA and protein levels of HMGA2 in NPC tissues. Immunohistochemical staining for HMGA2 protein was performed in 116 NPC patients. The associations between HMGA2 protein expression and clinicopathologic features and its prognostic significance were analyzed. In our results, we found mRNA and protein expressions of HMGA2 were upregulated in NPC tissues and cell lines. In 116 NPC tissue samples, we observed that HMGA2 protein overexpression was associated with clinical stage, lymph node metastasis, and distant metastasis. Moreover, NPC patients with high levels of HMGA2 protein expression had shorter overall survival in comparison to patients with low levels of HMGA2 protein. In multivariate analysis, HMGA2 protein overexpression was an unfavorable prognostic factor for NPC patients. In conclusion, HMGA2 is an important biomarker to predicting NPC patient's survival time.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteína HMGA2/biosíntesis , Neoplasias Nasofaríngeas/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Proteína HMGA2/genética , Humanos , Metástasis Linfática/genética , Metástasis Linfática/patología , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/mortalidad , Pronóstico , ARN Mensajero/biosíntesis , Regulación hacia Arriba , Adulto Joven
4.
Tumour Biol ; 35(10): 9815-21, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24986569

RESUMEN

Matrix metalloproteinase-14 (MMP-14) has been demonstrated to play an important role in tumor progression. The aim of this study was to analyze the correlation between MMP-14 expression and clinicopathologic features and its prognostic significance in non-small cell lung cancer (NSCLC). Immunohistochemical staining for MMP-14 protein was performed in 104 patients with NSCLC. High levels of MMP-14 protein were positively correlated with the status of clinical stage (I-II vs. III-IV; P < 0.001), N classification (N0-N1 vs. N2-N3; P < 0.001), distant metastasis (no vs. yes; P = 0.014), and differentiated degree (high vs. low or undifferentiated; P = 0.001). The patients with higher MMP-14 expression of protein had shorter survival time than patients with low MMP-14 expression. Multivariate analysis indicated that the level of MMP-14 expression was an independent prognostic indicator (P < 0.001) for the survival of patients with NSCLC. In conclusion, MMP-14 is a potential unfavorable prognostic factor for patients with NSCLC.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Metaloproteinasa 14 de la Matriz/biosíntesis , Adulto , Anciano , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Masculino , Metaloproteinasa 14 de la Matriz/análisis , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Regulación hacia Arriba
5.
BMC Cancer ; 14: 274, 2014 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-24751144

RESUMEN

BACKGROUND: CDK4 is a protein kinase in the CDK family important for G1/S phase cell cycle progression. However, the roles and molecular mechanisms of CDK4 triggering nasopharynx carcinogenesis are still unclear. METHODS: Lentiviral-vector mediated shRNA was used to suppress CDK4 expression and examine its molecular mechanisms. Using immunohistochemistry, we analyzed CDK4 protein expression in clinicopathologically characterized nasopharyngeal carcinoma (NPC) cases and nasopharyngeal tissues (NPs). Survival curves were plotted by the Kaplan-Meier method and compared using the log-rank test. RESULTS: In this investigation, we knocked down CDK4 expression and observed that NPC cell growth and cell cycle progression were significantly blocked by suppressing expression of CCND1, CDK6, and E2F1 as well as elevated p21 expression. Further, we found that reduced CDK4 expression elevated the expression of let-7c, a tumor-suppressive miRNA modulated by E2F1. We found that let-7c was markedly downregulated in NPC tissues compared to NPs and suppressed cell growth and cell cycle progression by modulating p15/p16/CDK4/E2F1 pathway. Finally, CDK4 protein was observed to be overexpressed in NPC tissues and could be considered an unfavorable prognosis factor for NPC patients although its independent prognostic value did not reach statistical significance (p = 0.087). CONCLUSIONS: Our results demonstrated that overexpressed CDK4 is an unfavorable prognostic factor which suppresses the expression of tumor suppressive-factor let-7c through p21/CCND1/CDK6/E2F1 signaling, and inhibits cell proliferation by p15/p16/CDK4/E2F1 feedback signaling in NPC.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , MicroARNs/biosíntesis , Neoplasias Nasofaríngeas/genética , Transducción de Señal/genética , Carcinoma , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , MicroARNs/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , Pronóstico
6.
World J Surg Oncol ; 12: 95, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24731479

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) is a special subtype of breast cancer that is characterized by poor prognosis, strong tumor invasion and a high pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC). The pCR rate is a prognostic factor for TNBC. We aimed to evaluate the relationship between pCR and TNBC after NAC and originally tried to identify factors related to achieving pCR for TNBC using a meta-analysis. METHODS: We systematically searched the literature for pCR and breast cancer after NAC and carefully identified eligibility criteria. The association between pCR and breast cancer subtypes was estimated using Review Manager, while pCR rates for TNBC and non-TNBC were determined using Meta-Analyst. RESULTS: This analysis included a total of 9,460 cases from 27 studies. The summary odds ratio estimating the relationship between pCR and breast cancer subtypes (TNBC vs non-TNBC) was 3.02 (95% confidence interval (CI), 2.66 to 3.42). The TNBC pCR rate was 28.9% (95% CI, 27.0 to 30.8%) and the non-TNBC was 12.5% (95% CI, 11.7 to 13.4%). From subgroup analyses, we identified the factors associated with the highest pCR rates for TNBC. CONCLUSIONS: TNBC has a higher pCR rate than non-TNBC. In the NAC setting, these factors of platinum-containing, more than six cycles, four kinds of drugs, 16 weeks' treatment duration and sequential chemotherapy may contribute to increasing the pCR rate.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Terapia Neoadyuvante , Compuestos Organoplatinos/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/patología , Ensayos Clínicos como Asunto , Femenino , Humanos , Metástasis de la Neoplasia , Pronóstico , Inducción de Remisión , Neoplasias de la Mama Triple Negativas/clasificación , Neoplasias de la Mama Triple Negativas/patología
7.
Aging (Albany NY) ; 16(8): 7217-7248, 2024 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-38656880

RESUMEN

AIM: In 2019, to examine the functions of METTL3 in liver and underlying mechanisms, we generated mice with hepatocyte-specific METTL3 homozygous knockout (METTL3Δhep) by simultaneously crossing METTL3fl/fl mice with Alb-iCre mice (GPT) or Alb-Cre mice (JAX), respectively. In this study, we explored the potential reasons why hepatocyte-specific METTL3 homozygous disruption by Alb-iCre mice (GPT), but not by Alb-Cre mice (JAX), resulted in acute liver failure (ALF) and then postnatal lethality. MAIN METHODS: Mice with hepatocyte-specific METTL3 knockout were generated by simultaneously crossing METTL3fl/fl mice with Alb-iCre mice (GPT; Strain No. T003814) purchased from the GemPharmatech Co., Ltd., (Nanjing, China) or with Alb-Cre mice (JAX; Strain No. 003574) obtained from The Jackson Laboratory, followed by combined-phenotype analysis. The publicly available RNA-sequencing data deposited in the NCBI Gene Expression Omnibus (GEO) database under the accession No.: GSE198512 (postnatal lethality), GSE197800 (postnatal survival) and GSE176113 (postnatal survival) were mined to explore the potential reasons why hepatocyte-specific METTL3 homozygous deletion by Alb-iCre mice (GPT), but not by Alb-Cre mice (JAX), leads to ALF and then postnatal lethality. KEY FINDINGS: Firstly, we observed that hepatocyte-specific METTL3 homozygous deficiency by Alb-iCre mice (GPT) or by Alb-Cre mice (JAX) caused liver injury, abnormal lipid accumulation and apoptosis. Secondly, we are surprised to find that hepatocyte-specific METTL3 homozygous deletion by Alb-iCre mice (GPT), but not by Alb-Cre mice (JAX), led to ALF and then postnatal lethality. Our findings clearly demonstrated that METTL3Δhep mice (GPT), which are about to die, exhibited the severe destruction of liver histological structure, suggesting that METTL3Δhep mice (GPT) nearly lose normal liver function, which subsequently contributes to ALF, followed by postnatal lethality. Finally, we unexpectedly found that as the compensatory growth responses of hepatocytes to liver injury induced by METTL3Δhep (GPT), the proliferation of METTL3Δhep hepatocytes (GPT), unlike METTL3Δhep hepatocytes (JAX), was not evidenced by the significant increase of Ki67-positive hepatocytes, not accompanied by upregulation of cell-cycle-related genes. Moreover, GO analysis revealed that upregulated genes in METTL3Δhep livers (GPT), unlike METTL3Δhep livers (JAX), are not functionally enriched in terms associated with cell cycle, cell division, mitosis, microtubule cytoskeleton organization, spindle organization, chromatin segregation and organization, and nuclear division, consistent with the loss of compensatory proliferation of METTL3Δhep hepatocytes (GPT) observed in vivo. Thus, obviously, the loss of the compensatory growth capacity of METTL3Δhep hepatocytes (GPT) in response to liver injury might contribute to, at least partially, ALF and subsequently postnatal lethality of METTL3Δhep mice (GPT). SIGNIFICANCE: These findings from this study and other labs provide strong evidence that these phenotypes (i.e., ALF and postnatal lethality) of METTL3Δhep mice (GPT) might be not the real functions of METTL3, and closely related with Alb-iCre mice (GPT), suggesting that we should remind researchers to use Alb-iCre mice (GPT) with caution to knockout gene in hepatocytes in vivo.


Asunto(s)
Hepatocitos , Fallo Hepático Agudo , Metiltransferasas , Animales , Ratones , Hepatocitos/metabolismo , Hepatocitos/patología , Hígado/patología , Hígado/metabolismo , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/patología , Fallo Hepático Agudo/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones Noqueados
8.
Aging (Albany NY) ; 15(10): 4391-4410, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37219449

RESUMEN

B-cell-specific Moloney murine leukemia virus integration site 1 (Bmi-1) is overexpressed in various cancer types. We found that Bmi-1 mRNA levels were elevated in nasopharyngeal carcinoma (NPC) cell lines. In immunohistochemical analyses, high Bmi-1 levels were observed in not only 5 of 38 non-cancerous nasopharyngeal squamous epithelial biopsies, but also in 66 of 98 NPC specimens (67.3%). High Bmi-1 levels were detected more frequently in T3-T4, N2-N3 and stage III-IV NPC biopsies than in T1-T2, N0-N1 and stage I-II NPC samples, indicating that Bmi-1 is upregulated in advanced NPC. In 5-8F and SUNE1 NPC cells, stable depletion of Bmi-1 using lentiviral RNA interference greatly suppressed cell proliferation, induced G1-phase cell cycle arrest, reduced cell stemness and suppressed cell migration and invasion. Likewise, knocking down Bmi-1 inhibited NPC cell growth in nude mice. Both chromatin immunoprecipitation and Western blotting assays demonstrated that Hairy gene homolog (HRY) upregulated Bmi-1 by binding to its promoter, thereby increasing the stemness of NPC cells. Immunohistochemistry and quantitative real-time PCR analyses revealed that HRY expression correlated positively with Bmi-1 expression in a cohort of NPC biopsies. These findings suggested that HRY promotes NPC cell stemness by upregulating Bmi-1, and that silencing Bmi-1 can suppress NPC progression.


Asunto(s)
Neoplasias Nasofaríngeas , Animales , Ratones , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/patología , Ratones Desnudos , Línea Celular Tumoral , Nasofaringe/patología , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética
9.
Aging (Albany NY) ; 15(12): 5550-5568, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37335109

RESUMEN

AIMS: N6-methyladenosine (m6A), the most abundant and conserved epigenetic modification of mRNA, participates in various physiological and pathological processes. However, the roles of m6A modification in liver lipid metabolism have yet to be understood entirely. We aimed to investigate the roles of the m6A "writer" protein methyltransferase-like 3 (Mettl3) in liver lipid metabolism and the underlying mechanisms. MAIN METHODS: We assessed the expression of Mettl3 in liver tissues of diabetes (db/db) mice, obese (ob/ob) mice, high saturated fat-, cholesterol-, and fructose-induced non-alcoholic fatty liver disease (NAFLD) mice, and alcohol abuse and alcoholism (NIAAA) mice by quantitative reverse-transcriptase PCR (qRT-PCR). Hepatocyte-specific Mettl3 knockout mice were used to evaluate the effects of Mettl3 deficiency in mouse liver. The molecular mechanisms underlying the roles of Mettl3 deletion in liver lipid metabolism were explored by multi-omics joint analysis of public data from the Gene Expression Omnibus database and further validated by qRT-PCR and Western blot. KEY FINDINGS: Significantly decreased Mettl3 expression was associated with NAFLD progression. Hepatocyte-specific knockout of Mettl3 resulted in significant lipid accumulation in the liver, increased serum total cholesterol levels, and progressive liver damage in mice. Mechanistically, loss of Mettl3 significantly downregulated the expression levels of multiple m6A-modified mRNAs related to lipid metabolism, including Adh7, Cpt1a, and Cyp7a1, further promoting lipid metabolism disorders and liver injury in mice. SIGNIFICANCE: In summary, our findings demonstrate that the expression alteration of genes related to lipid metabolism by Mettl3-mediated m6A modification contributes to the development of NAFLD.


Asunto(s)
Trastornos del Metabolismo de los Lípidos , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Metiltransferasas/genética , Metiltransferasas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Metabolismo de los Lípidos/genética , Expresión Génica
10.
Histopathology ; 61(1): 113-22, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22486228

RESUMEN

AIM: To investigate whether the neoplastic spindle cells in nasopharyngeal carcinoma (NPC) are associated with the process of epithelial-mesenchymal transition (EMT). METHODS AND RESULTS: We used immunohistochemistry to analyse the expression of cytokeratin, E-cadherin, ß-catenin, vimentin, fibronectin, Snail1, Slug and aldehyde dehydrogenase 1 (ALDH1) in 115 cases of NPC in which there were neoplastic spindle cells; in 47 cases a neoplastic squamous cell component was also present. There was no significant difference in the expression of cytokeratin observed in the neoplastic spindle cells (P = 0.644), compared to the squamous component whereas E-cadherin expression was reduced. By contrast, the expression of ß-catenin, vimentin, fibronectin, Snail1, Slug and ALDH1 was up-regulated in the spindle cells (all P = 0.000). Furthermore, E-cadherin expression was associated negatively with ß-catenin (P < 0.001), vimentin (P < 0.001), fibronectin (P < 0.001), Slug (P < 0.001) and ALDH1 (P < 0.001) in neoplastic spindle cells, but did not correlate with Snail1 expression (P = 0.093). CONCLUSIONS: Our findings demonstrate for the first time that EMT might play an important role in the development of neoplastic spindle cells in NPC.


Asunto(s)
Carcinoma de Células Escamosas/patología , Transición Epitelial-Mesenquimal , Fibroblastos/patología , Neoplasias Nasofaríngeas/patología , Adolescente , Adulto , Anciano , Familia de Aldehído Deshidrogenasa 1 , Biomarcadores de Tumor/metabolismo , Cadherinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Progresión de la Enfermedad , Femenino , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Humanos , Inmunohistoquímica/métodos , Isoenzimas/metabolismo , Queratinas/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias Nasofaríngeas/metabolismo , Invasividad Neoplásica/patología , Retinal-Deshidrogenasa/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Vimentina/metabolismo , Adulto Joven , beta Catenina/metabolismo
11.
Histopathology ; 61(2): 237-46, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22385354

RESUMEN

AIMS: To investigate the aberrant expression of N-cadherin in nasopharyngeal carcinoma (NPC) and its prognostic significance. METHODS AND RESULTS: Immunohistochemical staining for N-cadherin protein was performed on tissue microarray (TMA) from 122 NPC patients. Cytoplasmic N-cadherin was observed in 42.6% and nuclear N-cadherin in 45.1% of NPC tissues. High expression of cytoplasmic and nuclear N-cadherin was associated with a majority of the clinicopathological variables, including lymph node metastasis, distant metastasis and clinical stage. Cytoplasmic N-cadherin was associated positively with nuclear N-cadherin expression (P = 0.000). In univariate analysis, cytoplasmic N-cadherin showed no significant impact on patient prognosis. In contrast, the overall survival was significantly shorter in patients with high nuclear N-cadherin than those with low levels of staining (P = 0.002). A high expression of nuclear N-cadherin predicted poorer survival in patients with late stage disease (P = 0.033), but not those with early tumour stage. In addition, multivariate analysis showed nuclear N-cadherin to bean independent prognostic marker for NPC patients (P = 0.024). CONCLUSIONS: Nuclear N-cadherin expression may represent a valuable prognostic marker in NPC patients, especially those with late stage disease.


Asunto(s)
Antígenos CD/metabolismo , Biomarcadores de Tumor/metabolismo , Cadherinas/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Adolescente , Adulto , Anciano , Carcinoma , Núcleo Celular/metabolismo , China/epidemiología , Citoplasma/metabolismo , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis Multivariante , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/mortalidad , Neoplasias Nasofaríngeas/patología , Estadificación de Neoplasias , Pronóstico , Adulto Joven
12.
Acta Biochim Biophys Sin (Shanghai) ; 44(6): 503-12, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22551584

RESUMEN

The aim of the present study was to analyze the expression of DNA-binding protein inhibitor 2 (ID2) in nasopharyngeal carcinoma (NPC) and its correlation with clinicopathological features. It was found that the expression of ID2 was significantly increased in NPC cells when compared with that in NP69 cell line. Similar level of ID2 cytoplasmic expression was observed in NPC when compared with that in non-cancerous nasopharynx tissues. However, the level of ID2 in nucleus was increased in NPC when compared with that in normal nasopharynx tissues. Furthermore, the higher expression level of nuclear ID2 was significantly associated with tumor size (T classification), lymph node metastasis (N classification), and clinical stage. Patients with increased ID2 expression level had poorer overall survival rates than those with low ID2 levels. The inhibition of ID2 expression in NPC cell line SUNE1 by lentiviral-mediated short hairpin RNA could suppress cell proliferation and colony formation, but did not disrupt cell migration. Knocking down the expression of ID2 by RNA interference could down-regulate the expression of Snail, suggesting that ID2-promoted cell growth, partially attributing to the regulation of Snail activity in NPC. Our study demonstrated that over-expression of ID2 protein is an unfavorable prognostic factor which promotes cell proliferation in NPC.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proteína 2 Inhibidora de la Diferenciación/biosíntesis , Neoplasias Nasofaríngeas/metabolismo , Adulto , Anciano , Carcinoma , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/diagnóstico , Neoplasias Nasofaríngeas/patología , Nasofaringe/metabolismo , Pronóstico , ARN Mensajero/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/biosíntesis
14.
J Cancer ; 13(3): 987-997, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154464

RESUMEN

The current study aimed to investigate the function of the Hedgehog pathway and its association with epithelial-mesenchymal transition (EMT) in epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) resistance in non-small cell lung cancer (NSCLC). Lung tumor tissue specimens from EGFR TKI-resistant patients, including those with brain metastases, had hyperactive Hedgehog signaling compared with those from TKI-sensitive patients. SHH stimulation promoted GLI1 activation as well as cell motility in parental PC9 cells while suppressing gefitinib-induced apoptosis in gefitinib-resistant cells. SHH also promoted EMT in parental PC9 cells via E-cadherin suppression and N-cadherin and vimentin upregulation. The knockdown of GLI1 exhibited the opposite effects. Besides, SHH induced, whereas GLI1 knockdown reversed gefitinib resistance in xenograft tumors. The Hedgehog pathway inhibitor GDC-0449 synergized with gefitinib to increase xenograft tumor sensitivity to chemotherapy and extend survival in tumor-bearing animals. These results suggest the Hedgehog pathway mediates EGFR TKI resistance and induces EMT in NSCLC, representing a potential therapeutic target to defeat TKI resistance.

15.
J Transl Med ; 9: 38, 2011 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-21477379

RESUMEN

BACKGROUND: The aim of the present study was to analyze the expression of Cyclin-dependent kinase 4 (CDK4) in lung cancer and its correlation with clinicopathologic features. Furthermore, the involvement of CDK4-mediated cell cycle progression and its molecular basis were investigated in the pathogenesis of lung cancer. METHODS: Using immunohistochemistry analysis, we analyzed CDK4 protein expression in 89 clinicopathologically characterized lung cancer patients (59 males and 30 females) with ages ranging from 36 to 78 years and compared them to 23 normal lung tissues. Cases with cytoplasmic and nuclear CDK4 immunostaining score values greater than or equal to 7 were regarded as high expression while scores less than 7 were considered low expression. The correlation between the expression level of CDK4 and clinical features was analyzed. Furthermore, we used lentiviral-mediated shRNA to suppress the expression of CDK4 and investigate its function and molecular mechanism for mediating cell cycle progression. RESULTS: The expression level of CDK4 protein was significantly increased in lung cancer tissues compared to normal tissues (P < 0.001). In addition, high levels of CDK4 protein were positively correlated with the status of pathology classification (P = 0.047), lymph node metastasis (P = 0.007), and clinical stage (P = 0.004) of lung cancer patients. Patients with higher CDK4 expression had a markedly shorter overall survival time than patients with low CDK4 expression. Multivariate analysis suggested the level of CDK4 expression was an independent prognostic indicator (P < 0.001) for the survival of patients with lung cancer. Use of lentiviral-mediated shRNA to inhibit the expression of CDK4 in lung cancer cell line A549 not only inhibited cell cycle progression, but also dramatically suppressed cell proliferation, colony formation, and migration. Furthermore, suppressing CDK4 expression also significantly elevated the expression of cell cycle regulator p21 CONCLUSION: Overexpressed CDK4 is a potential unfavorable prognostic factor and mediates cell cycle progression by regulating the expression of p21 in lung cancer.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Pulmón/enzimología , Pulmón/patología , Neoplasias Pulmonares/genética , Masculino , Análisis Multivariante , Modelos de Riesgos Proporcionales , Análisis de Supervivencia
16.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 32(10): 1247-1250, 2020 Oct.
Artículo en Zh | MEDLINE | ID: mdl-33198873

RESUMEN

OBJECTIVE: To explore the effect of hydrogen sulfide (H2S) on expression of phosphatidylinositol 3 kinase/protein kinase B (PI3K/Akt) signal pathway in rats with intestinal ischemia/reperfusion (IRI) injury. METHODS: Thirty male Wistar rats were divided into sham operation group (Sham group), IRI group, and H2S precursor sodium hydrosuphide (NaHS) intervention group (IRI+NaHS group) by random number table method, with 10 rats in each group. The animal model of IRI was established by 60 minutes superior mesenteric artery (SMA) blockage with non-invasive vascular clamp and 120 minutes reflow. SMA was dissociated and peritoneum cavity was closed in Sham group. The rats in IRI+NaHS group was received NaHS (100 µmol/kg bolus+1.07 mmol×kg-1×h-1 infusion) 10 minutes prior to the onset of reperfusion, while the rats in IRI group and Sham group were received equal volume of normal sodium. Blood in vena cava was collected. H2S was detected by sensitive sulfide electrode. Rats were sacrificed after blood collection. Histopathology change was observed by hematoxylin-eosin (HE) staining, ileal pathological score was studied by Chiu score. The protein expressions of phosphated Akt (p-Akt), Akt, PI3K, cleaved caspase-9, mammalian target of rapamycin (mTOR) were determined by Western Blot. RESULTS: Compared with the Sham group, there was intestinal mucosa structure disorder edema and shedding villous fracture in the IRI group. Ileal pathological score in IRI group was significantly increased (4.21±0.15 vs. 0.15±0.03, P < 0.01), while plasma H2S in IRI group was significantly decreased (µmol/L: 26.72±3.17 vs. 38.34±5.24, P < 0.01). Ileal p-Akt, PI3K, caspase-9 and mTOR protein in IRI group were significantly increased (p-Akt/GAPDH: 2.67±0.12 vs. 0.24±0.05, PI3K/GAPDH: 1.42±0.07 vs. 0.57±0.08, caspase-9/GAPDH: 4.23±0.61 vs. 0.13±0.02, mTOR/GAPDH: 2.17±0.23 vs. 0.23±0.02, all P < 0.01). Compared with the IRI group, pathological changes of intestinal mucosa in the IRI+NaHS group was improved, ileal pathological score was significantly decreased (1.56±0.02 vs. 4.21±0.15, P < 0.01), plasma H2S was significantly increased (µmol/L: 32.36±2.45 vs. 26.72±3.17, P < 0.01) and ileal p-Akt, PI3K were significantly increased (p-Akt/GAPDH: 5.12±0.08 vs. 2.67±0.12, PI3K/GAPDH: 3.14±0.05 vs. 1.42±0.07, both P < 0.01), while caspase-9, mTOR in IRI+NaHS group were significantly decreased (caspase-9/GAPDH: 2.12±0.24 vs. 4.23±0.61, mTOR/GAPDH: 1.37±0.28 vs. 2.17±0.23, both P < 0.01). CONCLUSIONS: H2S attenuates intestinal injury in IRI rats by up-regulating PI3K/Akt signal pathway and down-regulating caspase-9 and mTOR.


Asunto(s)
Daño por Reperfusión , Animales , Sulfuro de Hidrógeno , Masculino , Fosfatidilinositol 3-Quinasa , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Transducción de Señal
17.
J BUON ; 25(3): 1490-1496, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32862595

RESUMEN

PURPOSE: To compare the efficacy and safety of etoposide combined with lobaplatin or cisplatin in the first-line treatment of extensive-stage small cell lung cancer (SCLC). METHODS: A total of 98 extensive-stage SCLC patients treated at the Oncology Department from March 2015 to March 2017 were enrolled and divided into etoposide + lobaplatin group (EL group, n=49) and etoposide + cisplatin group (EP group, n=49) using a random number table. The clinical data of all patients were collected, and the short-term effective rate, changes in the levels of serum tumor markers carcinoembryonic antigen (CEA), cytokeratin 19 fragment (CYFRA21-1) and neurone specific enolase (NSE) before and after chemotherapy and adverse reactions were compared between the two groups. Moreover, the patients were followed up, and the overall survival (OS) and progression-free survival (PFS) were recorded. RESULTS: In EL group and EP group, the level of serum NSE significantly declined after treatment compared with that before treatment, but the levels of serum CEA and CYFRA21-1 were not significantly decreased after chemotherapy compared with those before chemotherapy. The incidence rate of leukopenia, erythropenia and thrombocytopenia was 71.4%, 44.9% and 40.8%, respectively, in EL group, and 85.7%, 30.6% and 24.5%, respectively, in EP group, and the degree I-II decline was more common in both groups. The proportion of gastrointestinal reactions was 14.3% and 59.2%, respectively, in EL group and EP group, with significant difference between the two groups. During follow-up, the 1-year OS was 59.2% (29/49) and 51.9% (25/49), respectively, and the 2-year OS was 26.5% (13/49) and 20.4% (10/49), respectively, in EL group and EP group. The survival curves of were plotted using the Kaplan-Meier method and log-rank test showed no statistically significant differences in the OS and PFS between the two groups. CONCLUSIONS: The short-term efficacy of EL and EP regimens is equivalent in the first-line treatment of extensive-stage SCLC, both OS and PFS are similar, and the adverse reactions can be tolerated. The EL regimen produced mild gastrointestinal reactions, and is worthy of clinical popularization.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/uso terapéutico , Ciclobutanos/uso terapéutico , Etopósido/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Compuestos Organoplatinos/uso terapéutico , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Supervivencia sin Enfermedad , Epirrubicina/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Carcinoma Pulmonar de Células Pequeñas/metabolismo
18.
Life Sci ; 235: 116798, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31472149

RESUMEN

Lung cancer is the leading cause for cancer death due to refractory nature to current treatment strategies, understanding the regulatory mechanism of therapy resistance of lung cancer is important for lung cancer therapy. Here, we aimed to study the role of SHCBP1 in lung cancer cisplatin resistance, we found SHCBP1 was upregulated in lung cancer tissues and cells, patients with high SHCBP1 had poor prognosis. SHC binding and spindle associated 1 (SHCBP1) overexpression promoted cisplatin induced apoptosis resistance, migration and invasion determined by apoptosis assay and transwell assay with or without Matrigel, while SHCBP1 knockdown inhibited cisplatin induced apoptosis resistance, migration and invasion. Wnt pathway promoted lung cancer progression, we found SHCBP1 activated Wnt pathway, characterized by promoting ß-catenin nuclear translocation. Inhibition of Wnt pathway in SHCBP1 overexpression cells reversed the effect of SHCBP1 overexpression, confirming SHCBP1 promoted lung cancer progression through activating Wnt pathway. We also found SHCBP1 expression was positively corrected with Wnt pathway activity in lung cancer samples. In summary, we found SHCBP1 promoted cisplatin induced apoptosis resistance, migration and invasion through activating Wnt pathway, providing a potential target for lung cancer therapy.


Asunto(s)
Apoptosis , Cisplatino/farmacología , Resistencia a Antineoplásicos , Neoplasias Pulmonares/patología , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Invasividad Neoplásica , Pronóstico , Proteínas Adaptadoras de la Señalización Shc/genética , Tasa de Supervivencia , Proteínas Wnt/genética , Vía de Señalización Wnt , beta Catenina/genética
19.
Mol Ther Nucleic Acids ; 17: 563-577, 2019 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-31382188

RESUMEN

Long non-coding RNAs and microRNAs (miRNAs) have been reported to participate in the progression of non-small-cell lung cancer (NSCLC). Long intergenic non-protein-coding RNA 472 (LINC00472), miR-149-3p, and miR-4270 were found to be involved in tumor activities, suggesting potential roles in NSCLC. Thus, this study aimed to examine the ability of LINC00472 to influence the progression of NSCLC with the involvement of miR-149-3p and miR-4270. Initially, differentially expressed long non-coding RNAs (lncRNAs), downstream regulatory miRNAs, and genes related to NSCLC were identified. Next, the interaction among LINC00472, miR-149-3p and miR-4270, and KLLN and the p53-signaling pathway was determined. The effect of LINC00472 on the expression of E-cadherin, N-cadherin, and Vimentin was examined through gain-of-function and loss-of-function experiments. Lastly, the effects of LINC00472 on NSCLC tumor growth were assessed in vivo. LINC00472 and KLLN were found to exhibit low levels, while miR-149-3p and miR-4270 were highly expressed in NSCLC. In addition, the overexpression of LINC00472 was observed to upregulate KLLN and activate the p53-signaling pathway, which ultimately inhibited the invasion, migration, and EMT of NSCLC cells via miR-149-3p and miR-4270, corresponding to decreased N-cadherin and Vimentin and increased E-cadherin. The overexpression of LINC00472 exerted an inhibitory effect on tumor growth in vivo. Taken together, the key evidence suggests that the overexpression of LINC00472 can downregulate miR-149-3p and miR-4270 to upregulate KLLN and activate the p53-signaling pathway, thus inhibiting the development of NSCLC. This study highlights the potential of LINC00472 as a promising therapeutic target for NSCLC treatment.

20.
Medicine (Baltimore) ; 97(44): e13014, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30383657

RESUMEN

To examine the outcomes of concurrent versus sequential whole-brain radiotherapy (WBRT) and epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) in nonsmall cell lung cancer (NSCLC) patients with EGFR mutation.Retrospectively 105 patients with NSCLC, brain metastasis, and EGFR mutation (Affiliated Hospital of Guangdong Medical University, 01/2011 to 12/2014) were grouped as: EGFR-TKIs alone (n = 39, group A), EGFR-TKIs + concurrent radiotherapy (n = 34, group B), and radiotherapy followed by EGFR-TKIs (n = 32, group C).The intracranial objective response rates of groups A, B, and C were 66.7%, 85.3%, and 75%, respectively (P < .05). The median intracranial progression-free survival of groups A, B, and C were 6.8, 12.4, and 9.1 months, respectively (P < .05). The median extracranial progression-free survival of groups A, B, and C were 7.8, 9.4, and 8.3 months, respectively (P > .05).EGFR-TKIs and WBRT by simultaneous application improved the short- and long-term benefits to patients with NSCLC brain metastasis carrying EGFR mutation compared to concurrent application or EGFR-TKIs alone without additional adverse events.


Asunto(s)
Neoplasias Encefálicas/terapia , Carcinoma de Pulmón de Células no Pequeñas/patología , Irradiación Craneana/métodos , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/administración & dosificación , Anciano , Encéfalo/patología , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Terapia Combinada/métodos , Irradiación Craneana/efectos adversos , Receptores ErbB/genética , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/efectos adversos , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA