Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Int J Surg ; 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38869973

RESUMEN

BACKGROUND: Cholecystectomy is the standard surgery for patients with gallbladder disease, but the impact of cholecystectomy on gastroesophageal reflux disease (GERD) is not clear. METHODS: We obtained genetic variants associated with cholecystectomy at a genome-wide significant level (P value<5 × 10-8) as instrumental variables (IVs) and performed Mendelian randomization (MR) to explore the relationship with GERD. RESULTS: The Inverse Variance Weighted analysis (IVW) showed that the risk of GERD in patients after cholecystectomy increased (OR=2.19; 95% CI: 1.18 - 4.09). At the same time, the analysis results of weighted median (OR=2.30; 95% CI: 1.51 - 3.48) and weighted mode (OR=2.21; 95% CI: 1.42 - 3.45) were also consistent with the direction of the IVW analysis and were statistically significant (P<0.05). CONCLUSIONS: This study shows that patients who have undergone cholecystectomy are a susceptible population of GERD.

2.
Int J Oncol ; 65(2)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38873997

RESUMEN

Non­small cell lung cancer (NSCLC) is one of the major causes of cancer­related death worldwide. Cisplatin is a front­line chemotherapeutic agent in NSCLC. Nevertheless, subsequent harsh side effects and drug resistance limit its further clinical application. Polydatin (PD) induces apoptosis in various cancer cells by generating reactive oxygen species (ROS). However, underlying molecular mechanisms of PD and its effects on cisplatin­mediated antitumor activity in NSCLC remains unknown. MTT, colony formation, wound healing analyses and flow cytometry was employed to investigate the cell phenotypic changes and ROS generation. Relative gene and protein expressions were evaluated by reverse transcription­quantitative PCR and western blot analyses. The antitumor effects of PD, cisplatin and their combination were evaluated by mouse xenograft model. In the present study, it was found that PD in combination with cisplatin synergistically enhances the antitumor activity in NSCLC by stimulating ROS­mediated endoplasmic reticulum stress, and the C­Jun­amino­terminal kinase and p38 mitogen­activated protein kinase signaling pathways. PD treatment elevated ROS generation by promoting expression of NADPH oxidase 5 (NOX5), and NOX5 knockdown attenuated ROS­mediated cytotoxicity of PD in NSCLC cells. Mice xenograft model further confirmed the synergistic antitumor efficacy of combined therapy with PD and cisplatin. The present study exhibited a superior therapeutic strategy for some patients with NSCLC by combining PD and cisplatin.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Cisplatino , Sinergismo Farmacológico , Glucósidos , Neoplasias Pulmonares , NADPH Oxidasa 5 , Estrés Oxidativo , Especies Reactivas de Oxígeno , Estilbenos , Ensayos Antitumor por Modelo de Xenoinjerto , Cisplatino/farmacología , Cisplatino/uso terapéutico , Glucósidos/farmacología , Glucósidos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Animales , Humanos , Estilbenos/farmacología , Estilbenos/uso terapéutico , Ratones , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Apoptosis/efectos de los fármacos , Células A549 , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proliferación Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino
3.
Oncogene ; 40(25): 4324-4337, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34079085

RESUMEN

Cholangiocarcinoma (CCA) is aggressive and has poor clinical outcomes because of typically delayed diagnosis and a lack of effective non-surgical therapeutic options. Recent studies have shown that plasmalemma vesicle-associated protein (PLVAP) is related to angiogenesis in various tumors, and in vivo PLVAP targeting therapy has been proven effective against hepatocellular carcinoma and pancreatic cancer. The goal of this study was to determine the potential therapeutic utility of targeting PLVAP and thus angiogenesis in CCA and explore the underlying molecular mechanisms. We found that the PLVAP expression levels were significantly higher in CCA tissues when compared with matched adjacent non-tumor tissues obtained from a total of 90 CCA patients; higher expression levels of PLVAP were associated with shorter overall survival of patients. In addition, overexpression of PLVAP was associated with higher micro-vessel density in CCA tissues. In a PLVAP overexpressing CCA patient-derived xenograft model, a novel humanized anti-PLVAP antibody in combination with Gemcitabine plus Cisplatin was significantly inhibited tumor growth. Molecular analysis of CCA cells co-cultured with human umbilical vascular endothelial cells or human hepatic sinusoidal endothelial cells showed that Dickkopf-related protein 1 (DKK1) secreted by CCA cells activated the PI3K/Akt pathway after binding to its receptor, cytoskeleton-associated protein 4 (CKAP4), resulting in the upregulation of PLVAP. Thus, CCA cells increased the angiogenic potency of endothelial cells in a paracrine fashion. Consistently, patients bearing CKAP4 and PLVAP overexpressing tumors had a poor prognosis. In conclusion, the DKK1/CKAP4/PI3K/PLVAP pathway increases angiogenesis in CCA and is therefore a potential anti-angiogenic target.


Asunto(s)
Colangiocarcinoma/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Patológica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/efectos de los fármacos , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Colangiocarcinoma/patología , Cisplatino/farmacología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/patología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones Desnudos , Persona de Mediana Edad , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Gemcitabina , Neoplasias Pancreáticas
4.
Arch Pharm Res ; 44(4): 414-426, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33759138

RESUMEN

Long non-coding RNAs (LncRNAs) have been implicated in the pathogenesis of various human diseases. In this study, we probed into the role and potential mechanisms of the antisense of IGF2R non-protein coding RNA (LncRNA AIRN) in the progression of hepatocellular carcinoma (HCC). Using a quantitative real-time polymerase chain reaction, we corroborated that LncRNA AIRN expression was raised in the HCC tissues and cells. The bioinformatic analysis revealed that a potential interaction between LncRNA AIRN and STAT1, which was verified by the RNA pull-down and RNA immunoprecipitation. In the cycloheximide-chase assay, the knockdown of LncRNA AIRN enhanced the stability of STAT1 protein. In the immunoprecipitation assay, the knockdown of LncRNA AIRN restrained the cullin 4A (CUL4A)-mediated ubiquitination of STAT1 protein. The cell transfection, MTT and flow cytometry assays expounded that the LncRNA AIRN/STAT1 axis was bound up with the regulation of the proliferation and apoptosis of HCC cells. The in vivo experiments corroborated that the knockdown of LncRNA AIRN restrained the tumor growth of HCC. Our data expounded that the knockdown of LncRNA AIRN restrained HCC cell proliferation and boosted cell apoptosis by restraining the CUL4A-mediated ubiquitination of STAT1 protein.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT1/metabolismo , Apoptosis , Carcinoma Hepatocelular/patología , Proliferación Celular , Femenino , Humanos , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , ARN Largo no Codificante/genética , Factor de Transcripción STAT1/genética , Ubiquitinación
5.
Cell Death Dis ; 12(2): 169, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568627

RESUMEN

Cisplatin is one of the most effective chemotherapy drugs and is widely used in the treatment of cancer, including hepatocellular carcinoma (HCC) and cervical cancer, but its therapeutic benefit is limited by the development of resistance. Our previous studies demonstrated that BCAT1 promoted cell proliferation and decreased cisplatin sensitivity in HCC cells. However, the exact role and mechanism of how BCAT1 is involved in cisplatin cytotoxicity remain undefined. In this study, we revealed that cisplatin triggered autophagy in cancer cells, with an increase in BCAT1 expression. The cisplatin-induced up-regulation of BCAT1 decreased the cisplatin sensitivity by regulating autophagy through the mTOR signaling pathway. In addition, branched-chain amino acids or leucine treatment inhibited cisplatin- or BCAT1-mediated autophagy and increased cisplatin sensitivity by activating mTOR signaling in cancer cells. Moreover, inhibition of autophagy by chloroquine increased cisplatin sensitivity in vivo. Also, the knockdown of BCAT1 or the administration of leucine activated mTOR signaling, inhibited autophagy, and increased cisplatin sensitivity in cancer cells in vivo. These findings demonstrate a new mechanism, revealing that BCAT1 decreases cisplatin sensitivity in cancer cells by inducing mTOR-mediated autophagy via branched-chain amino acid leucine metabolism, providing an attractive pharmacological target to improve the effectiveness of chemotherapy.


Asunto(s)
Aminoácidos de Cadena Ramificada/metabolismo , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Cisplatino/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/metabolismo , Transaminasas/metabolismo , Animales , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Cloroquina/farmacología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal , Transaminasas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Cell Mol Med ; 25(3): 1468-1479, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33393219

RESUMEN

ATP citrate lyase (ACLY), a key enzyme in the metabolic reprogramming of many cancers, is widely expressed in various mammalian tissues. This study aimed to evaluate the effects and mechanisms of ACLY and its inhibitor BMS-303141 on hepatocellular carcinoma (HCC). In this study, ACLY was highly expressed in HCC tissues, especially in HepG2 and Huh7 cells, but was down-regulated in Hep3B and HCC-LM3 cells. Besides, ACLY knockdown inhibited HepG2 proliferation and clone formation, while opposite result was noticed in HCC-LM3 cells with ACLY overexpression. Moreover, ACLY knockdown impeded the migration and invasion abilities of HepG2 cells. Similarly, BMS-303141 suppressed HepG2 and Huh-7 cell proliferation. The p-eIF2α, ATF4, CHOP p-IRE1α, sXBP1 and p-PERK were activated in HepG2 cells stimulated by BMS-303141. In cells where ER stress was induced, ATF4 was involved in BMS-303141-mediated cell death procession, and ATF4 knockdown reduced HCC cell apoptosis stimulated by BMS-303141. In a mouse xenograft model, combined treatment with BMS-303141 and sorafenib reduced HepG2 tumour volume and weight. In addition, ACLY expression was associated with HCC metastasis and tumour-node-metastases staging. Survival analysis and Cox proportional hazards regression model showed that overall survival was lower in HCC patients with high ACLY expression; AFP level, TNM staging, tumour size and ACLY expression level were independent risk factors affecting their overall survival. In conclusion, ACLY might represent a promising target in which BMS-303141 could induce ER stress and activate p-eIF2α/ATF4/CHOP axis to promote apoptosis of HCC cells, and synergized with sorafenib to enhance the efficacy of HCC treatment.


Asunto(s)
ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , Factor de Transcripción Activador 4/metabolismo , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor 2 Eucariótico de Iniciación/metabolismo , Factor de Transcripción CHOP/metabolismo , Adulto , Anciano , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Modelos de Riesgos Proporcionales , Transducción de Señal/efectos de los fármacos , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncol Lett ; 16(3): 3635-3641, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30127972

RESUMEN

Dynamin-1-like protein (DNM1L) encodes a member of the dynamin superfamily of GTPases. It mediates mitochondrial and peroxisomal division and is involved in the regulation of apoptosis. However, its role in gastric cancer remains unclear. MKN-45 gastric cancer cells were transfected with short hairpin RNA (shRNA) to suppress DNM1L expression. MTT, flow cytometry, and Transwell assays were used to detect the changes in cell proliferation, apoptosis, and invasion, respectively. Immunohistochemistry was used to detect DNM1L expression in gastric adenocarcinoma specimens, and the association of DNM1L expression with clinicopathological features and prognosis was analyzed. After the suppression of endogenous DNM1L expression in MKN-45 cells with shRNA, cell proliferation and invasion rates were significantly reduced, whereas apoptosis was significantly increased (all P<0.01). The expression of DNM1L was significantly higher in gastric adenocarcinoma specimens compared with that in pericarcinoma tissues (P<0.001). The expression of DNM1L increased with increasing infiltration depth, lymphatic metastasis, and higher tumor node metastasis stage (P<0.05). The expression of DNM1L associated negatively with prognosis (P<0.01). DNM1L plays a critical role in the proliferation, invasion and apoptosis of human gastric adenocarcinoma. DNM1L expression has prognostic significance for the survival of patients with gastric adenocarcinoma.

8.
Mol Med Rep ; 17(1): 300-306, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115604

RESUMEN

Regulatory and effector T cells possess immunological cytotoxicity for tumor cells in the tumor microenvironment during tumor progression and are the primary suppressors inhuman cancer therapy. Interleukin­2 (IL­2) is an anticancer cytokine, which triggers human innate and adaptive immunity by stimulating T cell propagation and lymphocyte infiltration into tumor sites. IL­2 has been used successfully for cancer therapy. Recombinant adenovirus expressing IL­2 (rAd­IL­2) injection is a gene therapy agent that may improve prognosis of hepatocellular carcinoma (HCC) patients. In the present study, the ability of IL­2 to stimulate an immune response and the ability of recombinant adenovirus to inhibit tumor cell growth in HCC was investigated in a HCC tumor model. It was demonstrated that the regulatory and effector cell­mediated tumor suppression by antitumor cluster of differentiation (CD)4+ and CD8+ T cells stimulated by rAd­IL­2 is tumor­specific. Furthermore, rAd­IL­2 significantly stimulated tumor­specific cytotoxic T lymphocyte responses, increased interferon­Î³ release and enhanced antitumor immunity by inducing CD4+ and CD8+ T cell recruitment into the tumor, and additionally induced memory to protect tumor­bearing mice against tumor challenge. Treatment with rAd­IL­2 led to tumor regression and long­term survival of mice in the 120­day treatment period. Tumor challenge experiments demonstrated that rAd­IL­2 induced memory, protecting against reinfection. In conclusion, rAd­IL­2 may promote tumor­associated effector and regulatory T cell expansion and may be a potential therapeutic agent for clinical immunotherapy application in the treatment of cancer.


Asunto(s)
Adenoviridae/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Vectores Genéticos/genética , Interleucina-2/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Animales , Carcinoma Hepatocelular/terapia , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Ingeniería Genética , Terapia Genética , Humanos , Memoria Inmunológica , Inmunoterapia , Neoplasias Hepáticas/terapia , Ratones , Transducción Genética
9.
Liver Int ; 36(12): 1836-1847, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27246112

RESUMEN

BACKGROUND & AIMS: BCAT1 initiates the catabolism of branched-chain amino acids. Here, we investigated the function of BCAT1 and its transcriptional regulatory mechanism in hepatocellular carcinoma (HCC). METHODS: RNASeq was used to evaluate BCAT1 mRNA levels in HCC and normal matched specimens. After the exogenous expression of BCAT1 in BEL-7404 cells and the suppression of endogenous BCAT1 expression with shRNA in HepG2 cells, the cell proliferation, clone-forming ability and cell-cycle changes were measured with MTT assay, colony-forming assay and flow cytometry respectively. A xenograft model was used to investigate the effect of BCAT1 on cancer growth in vivo. Chromatin immunoprecipitation and luciferase reporter technologies were used to confirm the transcriptional regulation of the BCAT1 gene by MYC. The expression of the BCAT1 and MYC proteins in 122 HCC tissues was determined with an immunohistochemical analysis. RESULTS: BCAT1 mRNA was clearly increased in HCC tissues and hepatomas. The ectopic expression of BCAT1 in BEL-7404 cells enhanced their proliferation, clone formation, tumourigenic properties, S-G2 /M phase transition and chemoresistance to cisplatin. The suppression of BCAT1 expression in HepG2 cells significantly inhibited their proliferation, clone formation, and S-G2 /M phase transition and caused their chemosensitization to cisplatin. MYC affected the transcriptional regulation of BCAT1. Clinical data showed that BCAT1 expression correlated with a significantly poorer prognosis. CONCLUSION: BCAT1 plays a pathogenic role in HCC by causing cell proliferation and chemoresistance. The MYC transcription factor is involved in regulating the transcriptional activity of BCAT1. BCAT1 expression has prognostic significance for the survival of patients with HCC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/genética , Cisplatino/farmacología , Resistencia a Antineoplásicos , Neoplasias Hepáticas/genética , Transaminasas/genética , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , China , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Pronóstico , Proteínas Proto-Oncogénicas c-myc/genética , ARN Interferente Pequeño/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
World J Surg Oncol ; 13: 52, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25890096

RESUMEN

BACKGROUND: Bone marrow-derived stem cells (BMSCs) are locally adjacent to the tumor tissues and may interact with tumor cells directly. The purpose of this study was to explore the effects of BMSCs on the proliferation and invasion of osteosarcoma cells in vitro and the possible mechanism involved. METHODS: BMSCs were co-cultured with osteosarcoma cells, and CCK-8 assay was used to measure cell proliferation. The ELISA method was used to determine the concentration of stromal cell-derived factor-1 (SDF-1) in the supernatants. Reverse transcription polymerase chain reaction (RT-PCR) was performed to detect the expression of CXCR4 in osteosarcoma cells and BMSCs. Matrigel invasion assay was performed to measure tumor cell invasion. RESULTS: SDF-1 was detected in the supernatants of BMSCs, but not in osteosarcoma cells. Higher CXCR4 mRNA levels were detected in the osteosarcoma cell lines compared to BMSCs. In addition, conditioned medium from BMSCs can promote the proliferation and invasion of osteosarcoma cells, and AMD3100, an antagonist for CXCR4, can significantly downregulate these growth-promoting effects. CONCLUSIONS: BMSCs can promote the proliferation and invasion of osteosarcoma cells, which may involve the SDF-1/CXCR4 axis.


Asunto(s)
Células de la Médula Ósea/patología , Neoplasias Óseas/patología , Movimiento Celular , Proliferación Celular , Células Madre Mesenquimatosas/patología , Osteosarcoma/patología , Apoptosis , Western Blotting , Células de la Médula Ósea/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Células Madre Mesenquimatosas/metabolismo , Osteosarcoma/genética , Osteosarcoma/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
11.
Int J Clin Exp Pathol ; 8(11): 15007-12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26823836

RESUMEN

CIP2A is highly expressed in a variety of malignancies. We determined the expression and clinical significance of CIP2A in patients with advanced gastric cancer. CIP2A protein was expressed in 25 of 37 cancer tissue specimens. There was no correlation between CIP2A and PGP, GST-π, Topo-II, and LRP expression. Survival analysis showed significant differences between the survival rate of the CIP2A protein-positive and -negative groups (χ(2)=4.509, P=0.034), but the degree of positive expression was unrelated to survival time (χ(2)=4.639, P=0.098). CIP2A expression may have no prospective value for optimizing chemotherapy regimens, but it can be an indicator for patient prognosis.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/biosíntesis , Adenocarcinoma/patología , Autoantígenos/biosíntesis , Biomarcadores de Tumor/análisis , Proteínas de la Membrana/biosíntesis , Neoplasias Gástricas/patología , Subfamilia B de Transportador de Casetes de Unión a ATP/análisis , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Adulto , Anciano , Autoantígenos/análisis , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Estimación de Kaplan-Meier , Masculino , Proteínas de la Membrana/análisis , Persona de Mediana Edad , Pronóstico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidad
12.
Prz Gastroenterol ; 9(1): 43-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24868298

RESUMEN

INTRODUCTION: Acute pancreatitis (AP) protease release induces lung parenchymal destruction via inflammatory mediators. Ginkgo biloba has been reported to have anti-inflammatory effects. AIM: To evaluate the effect of ginkgo biloba extract on experimental acute pancreatitis-associated lung injury in the rat and to investigate the underlying mechanisms. MATERIAL AND METHODS: Acute pancreatitis was induced in rats by injection of 5% sodium taurocholate into the biliary pancreatic duct. Ginkgo biloba extract (GBE) was administered and pancreas and lung injury were assessed by histological examination. Alveolar macrophages were harvested by bronchoalveolar lavage. Specificity fluorescent probe DAF-FM-DA was applied to observe nitric oxide (NO) bioavailability in alveolar macrophage. The expression of tumour necrosis factor α (TNF-α) and macrophage migration inhibitory factor (MIF) protein in alveolar macrophage was studied by ELISA. RESULTS: In sodium taurocholate-induced acute pancreatitis, treatment with GBE significantly protected rats against lung injury associated with pancreatitis in histological sections. Ginkgo biloba extract had a tendency to down-regulate NO bioavailability compared with the AP group, but without statistical significance. Moreover, TNF-α and MIF at protein levels in alveolar macrophage with GBE treatment were decreased compared with the AP group. CONCLUSIONS: These results suggest that GBE could effectively protect rats against acute pancreatitis-associated lung injury. The GBE may inhibit excessive activation of alveolar macrophages from acute pancreatitis-associated lung injury through down-regulation of generation of NO, TNF-α and MIF. These findings suggest that ginkgo biloba extract is a suitable candidate as an effective strategy against acute pancreatitis-associated lung injury.

13.
Phytother Res ; 28(9): 1342-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24596136

RESUMEN

The therapeutic potential of baicalein against hepatoma cells was evaluated in vitro and in vivo. In cell viability assays, baicalein showed significant cytotoxicity against the hepatocellular carcinoma cell lines H22, Bel-7404, and Hep G2 and moderate cytotoxicity against immortalized human hepatocytes. Baicalein induced G0/G1-phase arrest in hepatocellular carcinoma cells, inhibited AKT, and promoted the degradation of ß-catenin and cyclin D1 without activation of GSK-3ß. Furthermore, baicalein significantly inhibited H22 xenograft tumor growth without causing obvious adverse effects on weight or liver and spleen weight indexes in ICR mice. Immunohistochemical analysis showed that the inhibition of tumor growth in baicalein-treated mice was associated with decreased AKT, ß-catenin, and cyclin D1 expression ex vivo. Our data indicate that baicalein might regulate cyclin D1 transcription via a ß-catenin-dependent mechanism, leading to cell cycle arrest at G0/G1 phase and impaired cancer cell proliferation. These results suggest that baicalein is a potential candidate for the treatment of hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/patología , Flavanonas/farmacología , Neoplasias Hepáticas/patología , Animales , Carcinoma Hepatocelular/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones Endogámicos ICR , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
14.
Hepat Mon ; 13(4): e6496, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23805156

RESUMEN

CONTEXT: Raising the chemotherapy-induced HBV reactivation is parallel to the increment of chemotherapy treatments in breast cancer patients. This meta-analysis aims to evaluate the efficacy of prophylactic use of lamivudine in breast cancer patients with HBsAg positive during chemotherapy. EVIDENCE ACQUISITION: MEDLINE, Pubmed, Ovid and Embase were used to search for clinical studies comparing with or without prophylactic use of lamivudine for HBV reactivation in breast cancer patients receiving chemotherapy. Outcomes of interest were the rate of HBV reactivation, incidence of hepatitis and incidence of hepatitis attributable to HBV reactivation, severity of hepatitis and severity of hepatitis attributable to HBV reactivation, the rate of chemotherapy disruption, and the rate of chemotherapy disruption attributable to HBV reactivation, overall mortality, and mortality attributable to HBV reactivation. RESULTS: Four studies with 285 patients were included in this meta-analysis. The rate of HBV reactivation, incidence of hepatitis and incidence of hepatitis related to HBV reactivation were reduced by use of prophylactic lamivudine compared to control group. Pooled Odds Ratios (ORs) were 0.09 (95% confidence intervals [CI] 0.03-0.26; P < 0.0001), 0.23 (95% CI 0.06-0.92; P = 0.04), and 0.10 (95% CI 0.03-0.32; P < 0.0001) respectively. There was a reduction in chemotherapy disruption related to HBV reactivation by use of prophylactic lamivudine (pooled OR = 0.11; 95% CI 0.02-0.58; P = 0.01). Chemotherapy disruption, overall mortality, and mortality attributable to HBV reactivation were not significantly different between two groups. Pooled ORs were 0.42 (95% CI 0.11-1.58; P = 0.20), 0.37 (95% CI 0.07-2.04; P = 0.25), and 0.25 (95% CI 0.01-6.82; P = 0.41) respectively. Lamivudine was well-tolerated, and no additional toxicity was observed. CONCLUSIONS: Use of prophylactic lamivudine may have positive effect on the outcome of breast cancer patients with HBsAg positive during chemotherapy.

15.
Med Oncol ; 30(2): 512, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23435990

RESUMEN

The outcome is variable for unresectable locally advanced non-small-cell lung cancer (ULANSCLC) patients treated with radio(chemo)therapy. The aim of this study is to investigate whether single-nucleotide polymorphisms (SNPs) in the transforming growth factor-beta1 (TGF-ß1) gene are associated with overall survival (OS) in ULANSCLC patients treated with definitive radio(chemo)therapy. A total of 109 patients who had available blood samples and complete clinical and follow-up information were enrolled. DNA from blood was genotyped for two SNPs: TGF-ß1 C-509T and T+869C. Kaplan-Meier survival analysis, log-rank test, and Cox's proportional hazard model were used to evaluate associations between genotypes and OS. Log-rank test showed that TGF-ß1 C-509T significantly correlated with OS (pooled P = 0.017). Both univariate and multivariate analyses showed that TGF-ß1 C-509T CC genotype was significantly associated with better OS than CT or TT genotypes. These results indicate that TGF-ß1 C-509T CC genotype is significantly associated with better OS in ULANSCLC patients treated with radio(chemo)therapy as a potential independent survival predictor.


Asunto(s)
Pueblo Asiatico/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Quimioradioterapia , Neoplasias Pulmonares/genética , Polimorfismo de Nucleótido Simple/genética , Factor de Crecimiento Transformador beta1/genética , Adulto , Anciano , Pueblo Asiatico/etnología , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/etnología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Quimioradioterapia/tendencias , Estudios de Cohortes , Femenino , Estudios de Asociación Genética/tendencias , Predisposición Genética a la Enfermedad/etnología , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/etnología , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Vigilancia de la Población , Resultado del Tratamiento
16.
World J Gastroenterol ; 18(28): 3696-704, 2012 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-22851861

RESUMEN

AIM: To investigate the mechanism of interleukin (IL)-6 secretion through blocking the IL-17A/IL-17A receptor (IL-17RA) signaling pathway with a short hairpin RNA (shRNA) in hepatic stellate cells (HSCs) in vitro. METHODS: HSCs were derived from the livers of adult male Sprague-Dawley rats. IL-6 expression was evaluated using real-time quantitative polymerase chain reaction and enzyme linked immunosorbent assay. The phosphorylation activity of p38 mitogen activated protein kinases (MAPK) and extracellular regulated protein kinases (ERK) 1/2 upon induction by IL-17A and suppression by IL-17RA shRNA were examined using Western blotting. RESULTS: IL-6 expression induced by IL-17A was significantly increased compared to control in HSCs (P < 0.01 in a dose-dependent manner). Suppression of IL-17RA using lentiviral-mediated shRNA inhibited IL-6 expression induced by IL-17A compared to group with only IL-17A treatment (1.44 ± 0.17 vs 4.07 ± 0.43, P < 0.01). IL-17A induced rapid phosphorylation of p38 MAPK and ERK1/2 after 5 min exposure, and showed the strongest levels of phosphorylation of p38 MAPK and ERK1/2 at 15 min in IL-17A-treated HSCs. IL-6 mRNA expression induced by IL-17A (100 ng/mL) for 3 h exposure was inhibited by preincubation with specific inhibitors of p38 MAPK (SB-203580) and ERK1/2 (PD-98059) compared to groups without inhibitors preincubation (1.67 ± 0.24, 2.01 ± 0.10 vs 4.08 ± 0.59, P < 0.01). Moreover, Lentiviral-mediated IL-17RA shRNA 1 inhibited IL-17A-induced IL-6 mRNA expression compared to random shRNA in HSCs (1.44 ± 0.17 vs 3.98 ± 0.68, P < 0.01). Lentiviral-mediated IL-17RA shRNA 1 inhibited phosphorylation of p38 MAPK and ERK1/2 induced by 15 min IL-17A (100 ng/mL) exposure. CONCLUSION: Down-regulation of the IL-17RA receptor by shRNA decreased IL-6 expression induced by IL-17A via p38 MAPK and ERK1/2 phosphorylation in HSCs. Suppression of IL-17RA expression may be a strategy to reduce the inflammatory response induced by IL-17A in the liver.


Asunto(s)
Regulación de la Expresión Génica , Células Estrelladas Hepáticas/citología , Interleucina-6/metabolismo , Lentivirus/genética , Receptores de Interleucina-17/metabolismo , Animales , Secuencia de Bases , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Vectores Genéticos , Inflamación , Cirrosis Hepática/patología , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Fosforilación , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Hepatogastroenterology ; 59(120): 2602-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22626880

RESUMEN

BACKGROUND/AIMS: Liver resection is the most effective treatment for selected patients with primary or metastatic hepatic tumor and many liver resection techniques have tried to minimize blood loss. The objective of the present study was to examine whether TissueLink dissection sealer (DS) was superior to clamp-crushing (CC) technique for liver transection or not. METHODOLOGY: MEDLINE, Pubmed, Ovid and Cochrane Library electronic databases were used to search for studies without language and time period restrictions. RESULTS: Four clinical trials with 276 patients were involved. We evaluated intraoperative blood loss, postoperative morbidity, postoperative biliary leakage, transfusion rate, operation time,hospital stay duration, postoperative mortality,transection time, blood loss in liver transection and pertran section area, transection speed, AST and TBIL and found no statistical differences between the DS and CC groups. Sensitive analysis showed transection time was longer in the former group. In addition, there was no apparent publication bias concerning intraoperative blood loss. CONCLUSIONS: In summary, we could not draw a firm conclusion that DS is superior to CC in liver resection of transection and the advantage of TissueLink dissecting sealer should be evaluated


Asunto(s)
Pérdida de Sangre Quirúrgica/prevención & control , Hemostasis Quirúrgica/instrumentación , Hepatectomía/instrumentación , Hemorragia Posoperatoria/prevención & control , Equipo Quirúrgico , Adulto , Anciano , Aspartato Aminotransferasas/sangre , Bilirrubina/sangre , Biomarcadores/sangre , Pérdida de Sangre Quirúrgica/mortalidad , Transfusión Sanguínea , Distribución de Chi-Cuadrado , Constricción , Diseño de Equipo , Femenino , Hemostasis Quirúrgica/efectos adversos , Hemostasis Quirúrgica/mortalidad , Hepatectomía/efectos adversos , Hepatectomía/mortalidad , Humanos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Hemorragia Posoperatoria/sangre , Hemorragia Posoperatoria/mortalidad , Factores de Tiempo , Resultado del Tratamiento
18.
Hepatogastroenterology ; 59(116): 1174-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22057375

RESUMEN

BACKGROUND/AIMS: NF-κB protein family members act as transcription facts and play a key role in regulating the immune response to infection and inflammatory signals. We proposed to determine the role of NF-κB in the development of trauma-associated liver damage and inflammation. METHODOLOGY: NF-κB DNA-binding activity was inhibited using double-stranded oligodeoxynucleotides (ODN). A total of 288 Wistar rats were randomly divided into four groups: control (C), traumatic inflammation (T), traumatic inflammation plus NF-κB decoy (ODN) and traumatic inflammation plus mutant NF-κB decoy ODN (mODN). RESULTS: Our data shows that inhibition of NF-κB activation significantly reduces liver tissue damage as evidenced by serum ALT levels and histological changes using both light microscopy and transmission electron microscopy. Furthermore, EMSA results showed that NF-κB activation was reduced in Group ODN rats compared to Group T and Group mODN rats. Expression of TNF-a and IL-6 protein in Group ODN rats were also reduced compared to Group T and Group mODN rats. We demonstrated that NF-κB plays an important role in trauma-associated inflammation and liver tissue damage. CONCLUSIONS: Suppressing NF-κB activation effectively reduces the release of the pro-inflammatory cytokines TNF-a and IL-6 following liver trauma.


Asunto(s)
Hepatopatías/etiología , FN-kappa B/fisiología , Heridas y Lesiones/complicaciones , Alanina Transaminasa/sangre , Animales , Inflamación/etiología , Inflamación/prevención & control , Interleucina-6/sangre , Hígado/patología , Hepatopatías/prevención & control , FN-kappa B/antagonistas & inhibidores , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/sangre
19.
J Cancer Res Clin Oncol ; 137(12): 1813-22, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21922327

RESUMEN

PURPOSE: Gemcitabine plus cisplatin (GP) is a first-line treatment for advanced non-small-cell lung cancer (NSCLC). In this study, we evaluated the efficacy and safety of a combined treatment consisting of CT-guided percutaneous (125)I seed implantation with GP chemotherapy for advanced NSCLC. METHODS: Fifty-three patients with advanced NSCLC were enrolled in a nonrandomized, two-armed clinical trial. Of these patients, 24 received a combination treatment of CT-guided percutaneous (125) I seed implantation and GP (the combo group), while 29 were treated with GP only (the control group). RESULTS: Patients in the combo group received (125)I seed implantation with prescription dose of 100-140 Gy and a total of 55 cycles of GP, and patients in the control group received a total of 73 cycles of GP. The overall response rate was 79.2% in the combo group and 41.4% in the control group. The median overall survival time was 13.5 ± 1.5 months in the combo group and 9.0 ± 1.8 months in the control group. The progression-free survival time was 8.0 ± 1.2 months in the combo group and 5.0 ± 0.8 months in the control group. The 1- and 2-year survival rates were 62.5 and 16.7% in the combo group, respectively, and 41.4 and 13.8% in the control group. The interventional complications in the combo group included 5 cases of pneumothorax and 4 cases of hemoptysis. There were no complications due to radiation pneumonia or radiation esophagitis in the combo group, and no patients had lethal hemoptysis or esophagotracheal fistula. Chemotherapy treatment-related toxicities, including Grade 3/4 myelosuppression and Grade 3 gastrointestinal toxicity, were similar in both groups. CONCLUSIONS: Our initial experience showed that combined CT-guided (125)I radioactive seed implantation and GP chemotherapy are effective and safe for treating advanced NCSLC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Braquiterapia/métodos , Carcinoma de Pulmón de Células no Pequeñas/terapia , Radioisótopos de Yodo/uso terapéutico , Neoplasias Pulmonares/terapia , Radioterapia Guiada por Imagen , Tomografía Computarizada por Rayos X/métodos , Adulto , Anciano , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Quimioradioterapia , Cisplatino/administración & dosificación , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Radiocirugia , Dosificación Radioterapéutica , Gemcitabina
20.
Oncol Lett ; 2(4): 753-758, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22848261

RESUMEN

Adoptive immunotherapy with T cells expressing CD20-specific chimeric T-cell receptors is a promising approach to lymphoma therapy. However, modification of the cellular signaling pathways in target tumor cells by treatment with engineered CD20-specific T cells has yet to be fully elucidated. In this study, the non-Hodgkin's lymphoma Raji cell line was co-cultured with T cells that were genetically modified with anti-CD20scFvFc/CD28/CD3ζ or anti-CD20scFvFc gene. The cytolytic activity of engineered CD20-specific T cells and IL-10 secretion was quantitated by Cytotoxicity and ELISA assays, respectively. The engineered CD20-specific T cells and Raji cells were sorted using flow cytomety for the Western blot analysis. Treatment of Raji cells with T cells genetically modified with anti-CD20scFvFc/CD28/CD3ζ chimera (compared to anti-CD20scFvFc) yielded a higher cytotoxicity against Raji cells in vitro. Additionally, we found that engineered CD20-specific T cells caused a decrease in IL-10 secretion and inhibition of phosphor-STAT3 and Bcl-2 expression in Raji cells, possibly through the down-regulation of p38 MAPK and NF-κB activity. These results indicate that the treatment of Raji cells with engineered CD20-specific T cells inhibited the cellular p38 MAPK signaling pathways, which enhanced its antitumor activities against CD20-positive tumor cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA