Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
FASEB J ; 35(8): e21826, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34320244

RESUMEN

In pancreatic cancer, autocrine insulin-like growth factor-1 (IGF-1) and paracrine insulin stimulate both IGF-1 receptor (IGF1R) and insulin receptor (IR) to increase tumor growth and glycolysis. In pancreatic cancer patients, cancer-induced glycolysis increases hepatic gluconeogenesis, skeletal muscle proteolysis, and fat lipolysis and, thereby, causes cancer cachexia. As a protein coexisting with IGF1R and IR, caveolin-1 (cav-1) may be involved in pancreatic cancer-induced cachexia. We undertook the present study to test this hypothesis. Out of wild-type MiaPaCa2 and AsPC1 human pancreatic cancer cell lines, we created their stable sub-lines whose cav-1 expression was diminished with RNA interference or increased with transgene expression. When these cells were studied in vitro, we found that cav-1 regulated IGF1R/IR expression and activation and also regulated cellular glycolysis. We transplanted the different types of MiaPaCa2 cells in growing athymic mice for 8 weeks, using intact athymic mice as tumor-free controls. We found that cav-1 levels in tumor grafts were correlated with expression levels of the enzymes that regulated hepatic gluconeogenesis, skeletal muscle proteolysis, and fat lipolysis in the respective tissues. When the tumors had original or increased cav-1, their carriers' body weight gain was less than the tumor-free reference. When cav-1 was diminished in tumors, the tumor carriers' body weight gain was not changed significantly, compared to the tumor-free reference. In conclusion, cav-1 in pancreatic cancer cells stimulated IGF1R/IR and glycolysis in the cancer cells and triggered cachectic states in the tumor carrier.


Asunto(s)
Caquexia/etiología , Caveolina 1/metabolismo , Glucólisis/fisiología , Neoplasias Pancreáticas/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animales , Caveolina 1/genética , Línea Celular Tumoral , Eliminación de Gen , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética
2.
J Appl Toxicol ; 42(12): 1937-1947, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35853845

RESUMEN

A hypoxic microenvironment results in significantly elevated hypoxia-inducible factor-1 (HIF-1) level in pancreatic cancer. HIF-1 functions to maintain the survival of cancer cells. The present study was performed to investigate whether inhibition of HIF-1α expression was involved in the in vitro antitumor effect of rhein in pancreatic cancer cells and to explore the underlying mechanism. sh-RNA knockout technique and western blotting were used to investigate the role of HIF-1α in autophagy activation in MiaPaCa-2 and PANC-1 cells. The survival and glycolysis were assessed using MTT assay and colorimetric kits, respectively. Apoptosis was evaluated by detecting the levels of apoptosis-related proteins using western blotting. Among the five pancreatic cancer cell lines, MiaPaCa-2 and PANC-1 cells were more sensitive to hypoxia-induced autophagy. HIF-1α regulated hypoxia-induced autophagy in MiaPaCa-2 and PANC-1 cells. Treatment with rhein inhibited the survival and suppressed glycolysis in MiaPaCa-2 and PANC-1 cells exposed to hypoxia. Bafilomycin A1 enhanced the suppressive effects of rhein on cell survival and glycolysis under hypoxia. Treatment with rhein, but not bafilomycin A1, significantly reduced HIF-1α expression. In conclusion, inhibition of HIF-1α-mediated autophagy enhances the in vitro antitumor activity of rhein in pancreatic cancer cells under hypoxia.


Asunto(s)
Neoplasias Pancreáticas , Humanos , Hipoxia de la Célula , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Autofagia , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
3.
Eur Surg Res ; 61(4-5): 130-135, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33260177

RESUMEN

The gut hormone cholecystokinin (CCK) is primarily secreted from I-cells in the duodenum and proximal jejunum. CCK secretion is stimulated by food digests and inhibited by proteases from pancreatic juice. CCK regulates digestion and appetite, stimulates pancreatic growth, and participates in pancreatic carcinogenesis. The molecular mechanisms of CCK-induced effects are not fully understood. When the mechanisms are studied in animals, the surgical model of pancreatobiliary diversion (PBD) is frequently used. After animals have had PBD, their CCK secretion is no longer inhibited by pancreas-derived proteases, so circulating CCK is increased. PBD is established in rats and hamsters, but not in mice. In this study, we modified PBD procedures and established the model in the mouse. In an experiment, we performed PBD and sham operation (SO) in two groups of mice (20 mice per group). Twenty days after operation, 75% of the PBD mice and all SO mice survived. When plasma CCK was determined by radioimmunoassay, the PBD group had higher levels than the SO group (p < 0.001). To assess pancreatic growth, we determined pancreatic weight and pancreatic contents of protein and DNA. We also stained pancreatic sections by immunohistochemistry to show the proliferating cells that either expressed the proliferating cell nuclear antigen or were labeled with 5-bromo-2'-deoxyuridine. As a result, the pancreases of the PBD mice were heavier (p < 0.001) and had more protein (p < 0.001), DNA (p < 0.01), and proliferating cells (p < 0.01) than those of the SO counterparts. Thus, pancreatic growth was increased as a result of PBD-induced hypercholecystokininemia. The plasma and pancreatic data demonstrated that the PBD model was a success. This model may be used in CCK-related research. For instance, pancreatic cancer is frequently studied in transgenic mice. PBD may be combined with the cancer model to study the role of CCK in the molecular biology of pancreatic cancer.


Asunto(s)
Desviación Biliopancreática/métodos , Colecistoquinina/fisiología , Animales , Colecistoquinina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Tamaño de los Órganos , Páncreas/patología
4.
J Cell Biochem ; 119(6): 4783-4791, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29331036

RESUMEN

Many studies have shown that randomized clinical trial with long-term follow-up found no improvement in stage II esophageal carcinoma (EC) patients receiving preoperative neoadjuvant chemoradiotherapy or chemotherapy treatment, this limitation underscored the urgent need for novel and reliable biomarkers for prognosis and prediction in stage II EC. miR-21 is frequently over-expressed while programmed cell death 4 (PDCD4) is often down-regulated in solid tumors. This study aimed to investigate the clinicopathological and prognostic significance of miR-21 and PDCD4 expression and to elucidate any correlation between miR-21 and PDCD4 expression in stage II EC patients. The expression level of miR-21 was up-regulated while the PDCD4 protein was down-regulated in stage II EC tissues compared with the adjacent non-cancerous tissues. Analyses of the clinicopathological parameters indicated that miR-21 expression was associated with differentiation grade, T stage, and N stage. PDCD4 protein expression was associated with T stage, N stage, and tumor size. The univariate linear regression analysis suggested a significant negative correlation between miR-21 and PDCD4 expression. The Kaplan-Meier curve showed that high miR-21 expression or low PDCD4 expression predicted poor progression-free survival (PFS) and overall survival (OS) of patients with stag II EC. In conclusion, both up-regulated miR-21 and down-regulated PDCD4 expression were associated with the aggressive progression and poor prognosis of stage II EC. miR-21 and PDCD4 might be potential biomarkers of tumor progression and indicators of prognosis of stag II EC.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Neoplasias Esofágicas , Regulación Neoplásica de la Expresión Génica , MicroARNs/biosíntesis , Proteínas de Neoplasias/biosíntesis , ARN Neoplásico/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Anciano , Supervivencia sin Enfermedad , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/cirugía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Tasa de Supervivencia
5.
Exp Ther Med ; 15(1): 1113-1118, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29434701

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive and lethal malignancies worldwide. At present, the underlying mechanisms of ESCC development and progression are poorly understood. Previous studies have demonstrated that homeodomain-interacting protein kinase-2 (HIPK2) serves an important role in cancer biology, particularly in proliferation and metastasis. However, the role of HIPK2 in ESCC cells remains unclear. In the current study, the expression of HIPK2 in ESCC specimens, adjacent non-cancerous tissues and cell lines was assessed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The effects of HIPK2 on cell metastasis, epithelial-mesenchymal transition (EMT) and proliferation were studied using a Transwell assay, RT-qPCR and a Cell Counting Kit-8 assay, respectively. The results indicated that HIPK2 expression was downregulated in ESCC specimens and cell lines, and HIPK2 expression was associated with tumor invasion and lymph node metastasis. Functional studies demonstrated that HIPK2 overexpression inhibited cell metastasis and EMT. Furthermore, HIPK2 overexpression suppressed cell viability during cisplatin treatment. These results suggest that HIPK2 serves an important role in regulating metastasis and the chemosensitivity of ESCC cells, implicating the potential application of HIPK2 in ESCC therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA