Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell ; 84(4): 760-775.e7, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38215751

RESUMEN

Apart from the canonical serotonin (5-hydroxytryptamine [5-HT])-receptor signaling transduction pattern, 5-HT-involved post-translational serotonylation has recently been noted. Here, we report a glyceraldehyde-3-phosphate dehydrogenase (GAPDH) serotonylation system that promotes the glycolytic metabolism and antitumor immune activity of CD8+ T cells. Tissue transglutaminase 2 (TGM2) transfers 5-HT to GAPDH glutamine 262 and catalyzes the serotonylation reaction. Serotonylation supports the cytoplasmic localization of GAPDH, which induces a glycolytic metabolic shift in CD8+ T cells and contributes to antitumor immunity. CD8+ T cells accumulate intracellular 5-HT for serotonylation through both synthesis by tryptophan hydroxylase 1 (TPH1) and uptake from the extracellular compartment via serotonin transporter (SERT). Monoamine oxidase A (MAOA) degrades 5-HT and acts as an intrinsic negative regulator of CD8+ T cells. The adoptive transfer of 5-HT-producing TPH1-overexpressing chimeric antigen receptor T (CAR-T) cells induced a robust antitumor response. Our findings expand the known range of neuroimmune interaction patterns by providing evidence of receptor-independent serotonylation post-translational modification.


Asunto(s)
Linfocitos T CD8-positivos , Serotonina , Linfocitos T CD8-positivos/metabolismo , Serotonina/metabolismo , Serotonina/farmacología , Procesamiento Proteico-Postraduccional , Transducción de Señal
3.
JCI Insight ; 7(14)2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35708906

RESUMEN

Although macrophages are undoubtedly attractive therapeutic targets for acute kidney injury (AKI) because of their critical roles in renal inflammation and repair, the underlying mechanisms of macrophage phenotype switching and efferocytosis in the regulation of inflammatory responses during AKI are still largely unclear. The present study elucidated the role of junctional adhesion molecule-like protein (JAML) in the pathogenesis of AKI. We found that JAML was significantly upregulated in kidneys from 2 different murine AKI models including renal ischemia/reperfusion injury (IRI) and cisplatin-induced AKI. By generation of bone marrow chimeric mice, macrophage-specific and tubular cell-specific Jaml conditional knockout mice, we demonstrated JAML promoted AKI mainly via a macrophage-dependent mechanism and found that JAML-mediated macrophage phenotype polarization and efferocytosis is one of the critical signal transduction pathways linking inflammatory responses to AKI. Mechanistically, the effects of JAML on the regulation of macrophages were, at least in part, associated with a macrophage-inducible C-type lectin-dependent mechanism. Collectively, our studies explore for the first time to our knowledge new biological functions of JAML in macrophages and conclude that JAML is an important mediator and biomarker of AKI. Pharmacological targeting of JAML-mediated signaling pathways at multiple levels may provide a novel therapeutic strategy for patients with AKI.


Asunto(s)
Lesión Renal Aguda , Lesión Renal Aguda/patología , Animales , Moléculas de Adhesión Celular , Moléculas de Adhesión de Unión/metabolismo , Riñón/patología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL
4.
Theranostics ; 12(9): 4386-4398, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35673560

RESUMEN

Rationale: Oxaliplatin is a widely used chemotherapy drug for advanced colorectal cancer (CRC) and its resistance is a major challenge for disease treatment. However, the molecular mechanism underlying oxaliplatin resistance remains largely elusive. Methods: An integrative analysis was performed to determine differentially expressed genes involved in oxaliplatin resistance. Loss- and gain-of-function studies were employed to investigate the roles of type Iγ phosphatidylinositol phosphate kinase (PIPKIγ) on oxaliplatin resistance in CRC cells. Exosomes derived from CRC cell lines were assessed for PD-L1 level and the ability to promote oxaliplatin resistance. Quantitative real-time PCR, immunofluorescence, luciferase reporter assay, Western blotting and other techniques were conducted to decipher the molecular mechanism. Results: PIPKIγ was identified as a critical gene related to oxaliplatin resistance in CRC. Genetic manipulation studies revealed that PIPKIγ profoundly facilitated oxaliplatin resistance and affected the expression of DNA damage repair proteins. Mechanistically, PIPKIγ promoted the expression of the immune checkpoint molecule PD-L1 via activation of NF-κB signaling pathway. Genetic silencing of PD-L1 did not affect CRC cell proliferation but significantly sensitized CRC cells to oxaliplatin. Notably, PD-L1 was revealed to be encapsulated in the exosomes, and the addition of exosomal PD-L1 to sh-PD-L1 CRC cells restored oxaliplatin resistance. Pharmacological hijacking PIPKIγ-exosomal PD-L1 axis largely reduced oxaliplatin resistance in CRC cells. In vivo experiments showed that PD-L1 loss significantly blocked oxaliplatin resistance and the addition of PD-L1-enriched exosomes promoted tumor growth and reduced mouse survival time. Conclusion: Our findings reveal a previous unprecedented role of PIPKIγ in oxaliplatin resistance and provide a key mechanism of exosomal PD-L1 in CRC with potential therapeutics.


Asunto(s)
Antígeno B7-H1 , Neoplasias Colorrectales , Animales , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/genética , Ratones , Oxaliplatino/farmacología , Fosfatos/uso terapéutico , Fosfatos de Fosfatidilinositol/uso terapéutico
5.
Int J Biol Sci ; 17(1): 107-118, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390837

RESUMEN

Aerobic glycolysis, also known as the Warburg effect, is emerged as a hallmark of most cancer cells. Increased aerobic glycolysis is closely associated with tumor aggressiveness and predicts a poor prognosis. Pancreatic ductal adenocarcinoma (PDAC) is characterized by prominent genomic aberrations and increased glycolytic phenotype. However, the detailed molecular events implicated in aerobic glycolysis of PDAC are not well understood. In this study, we performed a comprehensive molecular characterization using multidimensional ''omic'' data from The Cancer Genome Atlas (TCGA). Detailed analysis of 89 informative PDAC tumors identified substantial copy number variations (MYC, GATA6, FGFR1, IDO1, and SMAD4) and mutations (KRAS, SMAD4, and RNF43) related to aerobic glycolysis. Moreover, integrated analysis of transcriptional profiles revealed many differentially expressed long non-coding RNAs involved in PDAC aerobic glycolysis. Loss-of-function studies showed that LINC01559 and UNC5B-AS1 knockdown significantly inhibited the glycolytic capacity of PDAC cells as revealed by reduced glucose uptake, lactate production, and extracellular acidification rate. Moreover, genetic silencing of LINC01559 and UNC5B-AS1 suppressed tumor growth and resulted in alterations in several signaling pathways, such as TNF signaling pathway, IL-17 signaling pathway, and transcriptional misregulation in cancer. Notably, high expression of LINC01559 and UNC5B-AS1 predicted poor patient prognosis and correlated with the maximum standard uptakevalue (SUVmax) in PDAC patients who received preoperative 18F-FDG PET/CT. Taken together, our results decipher the glycolysis-associated copy number variations, mutations, and lncRNA landscapes in PDAC. These findings improve our knowledge of the molecular mechanism of PDAC aerobic glycolysis and may have practical implications for precision cancer therapy.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/genética , ARN Largo no Codificante/metabolismo , Efecto Warburg en Oncología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/terapia , Variaciones en el Número de Copia de ADN , Genoma Humano , Humanos , Terapia Molecular Dirigida , Mutación , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia
6.
J Exp Clin Cancer Res ; 38(1): 214, 2019 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-31118109

RESUMEN

BACKGROUND: Gastric cancer is one of the deadliest malignant tumours, with a high incidence in China, and is regulated by aberrantly overexpressed oncogenes. However, existing therapies are insufficient to meet patients' needs; thus, the identification of additional therapeutic targets and exploration of the underlying mechanism are urgently needed. GPAA1 is the subunit of the GPI transamidase that transfers the GPI anchor to proteins within the ER. The functional impacts of increased expression levels of GPAA1 in human cancers are not well understood. METHODS: Data mining was performed to determine the pattern of GPAA1 expression and the reason for its overexpression in tumour and adjacent normal tissues. In vitro and in vivo experiments evaluating proliferation and metastasis were performed using cells with stable deletion or overexpression of GPAA1. A tissue microarray established by the Ren Ji Hospital was utilized to analyse the expression profile of GPAA1 and its correlation with prognosis. Western blotting, an in situ proximity ligation assay, and co-immunoprecipitation (co-IP) were performed to reveal the mechanism of GPAA1 in gastric cancer. RESULTS: GPAA1 was a markedly upregulated oncogene in gastric cancer due to chromosomal amplification. GPAA1 overexpression was confirmed in specimens from the Ren Ji cohort and was associated with ERBB2 expression, predicting unsatisfactory patient outcomes. Aberrantly upregulated GPAA1 dramatically contributed to cancer growth and metastasis in in vitro and in vivo studies. Mechanistically, GPAA1 enhanced the levels of metastasis-associated GPI-anchored proteins to increase tumour metastasis and intensified lipid raft formation, which consequently promoted the interaction between EGFR and ERBB2 as well as downstream pro-proliferative signalling. CONCLUSIONS: GPAA1 facilitates the expression of cancer-related GPI-anchored proteins and supplies a more robust platform-the lipid raft-to promote EGFR-ERBB2 dimerization, which further contributes to tumour growth and metastasis and to cancer progression. GPAA1 could be a promising diagnostic biomarker and therapeutic target for gastric cancer.


Asunto(s)
Proteínas Ligadas a GPI/genética , Glicoproteínas de Membrana/genética , Receptor ErbB-2/genética , Neoplasias Gástricas/genética , Aciltransferasas/genética , Anciano , Animales , Proliferación Celular/genética , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Receptores ErbB/química , Receptores ErbB/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Xenoinjertos , Humanos , Masculino , Glicoproteínas de Membrana/química , Microdominios de Membrana/genética , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Pronóstico , Multimerización de Proteína/genética , Receptor ErbB-2/química , Transducción de Señal/genética , Neoplasias Gástricas/patología , Análisis de Matrices Tisulares
7.
Gut ; 68(11): 1994-2006, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30826748

RESUMEN

BACKGROUND AND AIMS: Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related death worldwide. Neurotransmitter-initiated signalling pathway is profoundly implicated in tumour initiation and progression. Here, we investigated whether dysregulated neurotransmitter receptors play a role during pancreatic tumourigenesis. METHODS: The Cancer Genome Atlas and Gene Expression Omnibus datasets were used to identify differentially expressed neurotransmitter receptors. The expression pattern of gamma-aminobutyric acid type A receptor pi subunit (GABRP) in human and mouse PDAC tissues and cells was studied by immunohistochemistry and western blot analysis. The in vivo implications of GABRP in PDAC were tested by subcutaneous xenograft model and lung metastasis model. Bioinformatics analysis, transwell experiment and orthotopic xenograft model were used to identify the in vitro and in vivo effects of GABRP on macrophages in PDAC. ELISA, co-immunoprecipitation, proximity ligation assay, electrophysiology, promoter luciferase activity and quantitative real-time PCR analyses were used to identify molecular mechanism. RESULTS: GABRP expression was remarkably increased in PDAC tissues and associated with poor prognosis, contributed to tumour growth and metastasis. GABRP was correlated with macrophage infiltration in PDAC and pharmacological deletion of macrophages largely abrogated the oncogenic functions of GABRP in PDAC. Mechanistically, GABRP interacted with KCNN4 to induce Ca2+ entry, which leads to activation of nuclear factor κB signalling and ultimately facilitates macrophage infiltration by inducing CXCL5 and CCL20 expression. CONCLUSIONS: Overexpressed GABRP exhibits an immunomodulatory role in PDAC in a neurotransmitter-independent manner. Targeting GABRP or its interaction partner KCNN4 may be an effective therapeutic strategy for PDAC.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Macrófagos/fisiología , Ratones , Transducción de Señal/fisiología
8.
J Immunol Res ; 2017: 3072745, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29464186

RESUMEN

Exemestane (EXE) is an irreversible steroidal aromatase inhibitor mainly used as an adjuvant endocrine therapy for postmenopausal women suffering from breast cancer. Besides inhibiting aromatase activity, EXE has multiple biological functions, such as antiproliferation, anti-inflammatory, and antioxidant activities which are all involved in hepatic fibrosis. Therefore, we investigated the role of EXE during the progress of hepatic fibrosis. The effect of EXE on liver injury and fibrosis were assessed in two hepatic fibrosis rat models, which were induced by either carbon tetrachloride (CCl4) or bile duct ligation (BDL). The influence of EXE treatment on activation and proliferation of primary rat hepatic stellate cells (HSCs) was observed in vitro. The results showed that EXE attenuated the liver fibrosis by decreasing the collagen deposition and α-SMA expression in vivo and inhibited the activation and proliferation of primary rat HSCs in vitro. Additionally, EXE promoted the secretion of antifibrotic and anti-inflammatory cytokine IL-10 in vivo and in HSC-T6 culture media. In conclusion, our findings reveal a new function of EXE on hepatic fibrosis and prompted its latent application in liver fibrotic-related disease.


Asunto(s)
Androstadienos/uso terapéutico , Antiinflamatorios/uso terapéutico , Inhibidores de la Aromatasa/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Células Estrelladas Hepáticas/fisiología , Hígado/patología , Actinas/metabolismo , Animales , Conductos Biliares/cirugía , Tetracloruro de Carbono/toxicidad , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Interleucina-10/metabolismo , Hígado/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
9.
Sci Rep ; 6: 31071, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27506146

RESUMEN

Gastrointestinal stromal tumor (GIST) is the most major mesenchymal neoplasm of the digestive tract. Up to now, imatinib mesylate has been used as a standard first-line treatment for irresectable and metastasized GIST patients or adjuvant treatment for advanced GIST patients who received surgical resection. However, secondary resistance to imatinib usually happens, resulting in a major obstacle in GIST successful therapy. In this study, we first found that collagen and calcium binding EGF domains 1 (CCBE1) expression gradually elevated along with the risk degree of NIH classification, and poor prognosis emerged in the CCBE1-positive patients. In vitro experiments showed that recombinant CCBE1 protein can enhance angiogenesis and neutralize partial effect of imatinib on the GIST-T1 cells. In conclusion, these data indicated that CCBE1 may be served as a new predictor of prognosis in post-operative GIST patients and may play an important role in stimulating GIST progression.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas de Unión al Calcio/metabolismo , Tumores del Estroma Gastrointestinal/metabolismo , Mesilato de Imatinib/uso terapéutico , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al Calcio/genética , Carcinogénesis , Línea Celular Tumoral , Resistencia a Medicamentos , Femenino , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/mortalidad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica , Pronóstico , Análisis de Supervivencia , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
10.
J Cancer Res Clin Oncol ; 141(2): 269-81, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25216702

RESUMEN

BACKGROUND: Lysyl oxidase-like 4 (LOXL4) has been found up-regulated in a variety of human malignancies, but its clinical significance and functional roles in gastric cancer (GC) remain unknown. METHODS: Lysyl oxidase-like 4 (LOXL4) expression level in tumor tissues and human GC cell lines was evaluated by quantitative real-time polymerase chain reaction, Western blotting and immunohistochemical analyses. Its clinical significance was inferred from the analysis of 379 tissue samples of patients with GC using tissue microarray. The roles of LOXL4 in cell proliferation, migration and invasion in vitro were analyzed by gene over-expression, RNA interference and recombinant protein. Effects of LOXL4 on regulation of focal adhesion kinase/Src kinase (FAK/Src) pathway were examined by Western blotting. RESULTS: Lysyl oxidase-like 4 (LOXL4) was up-regulated in GC tissues relative to paired non-tumor tissues, and this over-expression was significantly associated with tumor size, depth of tumor invasion, lymph node metastasis, tumor-node-metastasis (TNM) stages and poorer overall survival. Over-expression of LOXL4 has promotive effects on GC cell proliferation, migration and invasion in vitro, consistent with this, LOXL4 knockdown has inhibitive effects on GC cell proliferation, migration and invasion. Furthermore, recombinant human LOXL4 protein also promoted GC cell proliferation and migration. Subsequent mechanistic studies showed that LOXL4 could activate FAK/Src pathway to enhance cell-extracellular matrix adhesion. CONCLUSIONS: Taken together, our data reveal that up-regulation of LOXL4 expression is a frequent event in GC progression, contributes to tumor cell proliferation and metastasis, and LOXL4 may be a potential independent prognostic marker and therapeutic target for GC.


Asunto(s)
Adenocarcinoma/secundario , Aminoácido Oxidorreductasas/metabolismo , Movimiento Celular , Proliferación Celular , Quinasa 1 de Adhesión Focal/metabolismo , Neoplasias Gástricas/patología , Familia-src Quinasas/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Aminoácido Oxidorreductasas/genética , Apoptosis , Western Blotting , Adhesión Celular , Femenino , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Proteína-Lisina 6-Oxidasa , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidad , Tasa de Supervivencia , Células Tumorales Cultivadas , Familia-src Quinasas/genética
11.
Int J Clin Exp Pathol ; 7(10): 6447-61, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25400723

RESUMEN

CCN6/Wnt1-inducible signaling protein-3 (CCN6/WISP3) is a cysteine-rich protein that belongs to the CCN (Cyr61, CTGF, Nov) family of matricellular proteins, which are often dysregulated in cancers. However, the functional role and clinical significance of WISP3 in gastric cancer remain unclear. In this study, we found that silencing of WISP3 suppressed gastric cancer cell proliferation, migration and invasion. Cell adhesion to collagens (collagen I and IV), but not to fibronectin, were significantly inhibited by silencing of WISP3. Furthermore, silencing of WISP3 prevented ß-catenin transferring from cell cytoplasm to nuclear, and suppressed canonical Wnt/ß-catenin signaling and its downstream target genes, cyclin D1 and TCF-4. By immunohistochemical analysis of 379 patients, we found that the expression of WISP3 is closely associated with gastric cancer size and tumor invasion, and indicates a poor prognosis in both test cohort (253 patients) and validation cohort (126 patients). Moreover, the expression of WISP3 was positively correlated with the expression of cyclin D1 and TCF-4 in gastric cancer tissues. Taken together, our data suggests that WISP3 might be a promising prognostic factor and WISP3-Wnt/ß-catenin axis may be a new therapeutic target for the intervention of gastric cancer growth and metastasis.


Asunto(s)
Adenocarcinoma/metabolismo , Proteínas CCN de Señalización Intercelular/metabolismo , Movimiento Celular , Proliferación Celular , Técnicas de Silenciamiento del Gen , Neoplasias Gástricas/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular , Adenocarcinoma/genética , Adenocarcinoma/secundario , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proteínas CCN de Señalización Intercelular/genética , Adhesión Celular , Línea Celular Tumoral , Ciclina D1/metabolismo , Regulación hacia Abajo , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico , Interferencia de ARN , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factores de Tiempo , Factor de Transcripción 4 , Factores de Transcripción/metabolismo , Transfección , Carga Tumoral
12.
Int J Clin Exp Pathol ; 7(7): 3827-35, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25120759

RESUMEN

Paraneoplastic Ma1 (PNMA1) is a member of an expanding family of 'brain/testis' proteins involved in an autoimmune disorder defined as paraneoplastic neurological syndrome (PNS). Although it is widely studied in PNS, little is known about the underlying clinical significance and biological function of PNMA1 in tumors. Here, we find that elevated PNMA1 expression is more commonly observed in pancreatic ductal adenocarcinoma (PDAC) cell lines, compared with normal pancreatic cell and tissues from pancreatic ductal adenocarcinoma patient. Besides, higher PNMA1 expression is closely correlated with large tumor size. Suppression of endogenous PNMA1 expression decreases cell viability and promotes cell apoptosis. Subsequent studies reveal that the PI3K/AKT, MAPK/ERK pathway and members of the anti-apoptotic Bcl-2 family may be involved in the pro-survival and anti-apoptotic effect of PNMA1 on PDAC. Taken together, this study provides evidence that PNMA1 is involved in tumor growth of pancreatic carcinoma and PNMA1-related pathways might represent a new treatment strategy.


Asunto(s)
Antígenos/metabolismo , Carcinoma Ductal Pancreático/patología , Proliferación Celular/fisiología , Neoplasias Pancreáticas/patología , Transducción de Señal/fisiología , Anciano , Western Blotting , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares
13.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 41(4): 595-9, 2010 Jul.
Artículo en Chino | MEDLINE | ID: mdl-20848776

RESUMEN

OBJECTIVE: To investigate whether EGFR inhibitor AG1478 combined with celecoxib could enhance the inhibitive effects on the growth of gastric cancer cells. METHODS: Human gastric cancer cell line SGC-7901 was cultured and treated with different concentration of AG1478 and celecoxib, the proliferation of SGC-7901 cells was determined by MTT. The expression of proliferation cell nuclear antigen (PCNA) of SGC-7901 cells was detected by immunocytochemistry. ERK and p-ERK expression were determined by immunoblot. The TdT-mediated dUTP nick end labeling (TUNEL) assay was used to detect apoptosis. RESULTS: When SGC-7901 cells was treated with AG1478 or celecoxib alone, cell growth was only inhibited by high concentration of the two agents (10, 100 micromol/L) significantly, the IC50 of AG1478 and celecoxib were 69.69 micromol/L and 70.98 micromol/L respectively. Compared with AG1478 or celecoxib alone, combination of AG1478 and celecoxib at different doses significantly enhanced the inhibitive effects on cell growth (P < 0.01). Compared with control (56. 55%), the expression of PCNA was decreased in the cells treated with AG1478, celecoxib and the combination of these two agents, PCNA indeices were 26.24%, 38.16%, and 9.08%, respectively (P < 0.01). AG1478 induced cell apoptosis at 10 micromol/L, the rate was 7.88% vs. 3.54% in control (P < 0.01), and the combination of AG1478 with celecoxib showed an enhanced effect, the apoptosis rate was 14.90%. There were no any effects on ERK expression with the treatments of either AG1478, celecoxib alone or toghether, but the phospholation of ERK was decreased by AG1478 (P < 0.01). CONCLUSION: AG1478 combined with celecoxib results in enhanced inhibitive effect on the growth of SGC-7901, which may partly due to the suppression of ERK phospholation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Pirazoles/farmacología , Neoplasias Gástricas/patología , Sulfonamidas/farmacología , Tirfostinos/farmacología , Celecoxib , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Antígeno Nuclear de Célula en Proliferación/análisis , Quinazolinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...