Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Oncol ; 2022: 5744999, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36245984

RESUMEN

Objective: Hepatocellular carcinoma (HCC) shows a growing incidence over the past few years, and clinical efforts are made to search for more effective novel diagnosis and therapy regimen for it to improve its outcome. This study probed into the association of miR-3651 with the PI3K/AKT/mTOR pathway to offer a more detailed reference to the follow-up exploration of novel diagnosis and therapy methods of HCC. Methods: Totally, 83 patients with HCC treated in our hospital between Apr. 2017 and Aug. 2018, 100 patients with simple liver cirrhosis (LC), and 94 normal persons over the same time span were enrolled, and serum miR-3651 in them was quantified to understand the predictive and prognostic significance of miR-3651 for HCC. In addition, with purchased human HCC cell strains (HepG2), the impacts of miR-3651 on the invasion as well as proliferation of HepG2 were determined using the MTT and Transwell assays, and the PI3K/AKT/mTOR pathway and autophagy-associated proteins in HepG2 were quantified via WT. Results: Serum miR-3651 was found to be higher in HCC patients than in LC patients and normal persons, and it presented a sensitivity and specificity of 57.14% and 94.00%, respectively, in forecasting the occurrence of HCC in LC patients. The decrease of miR-3651 in HCC patients after therapy was strongly bound up with patients' prognosis, and its increase implied an increased risk of death. In in vitro assays, HepG2 presented higher miR-3651 expression than HL-7702, and upregulated miR-3651 intensified the invasion and proliferation of HepG2, while silencing miR-3651 gave rise to opposite results. Additionally, the PI3K/Akt/mTOR pathway in HepG2 presented an obvious activation state, and its activation was further intensified after increase of miR-3651, while its activation was suppressed after silence of miR-3651. Moreover, HepG2 presented notably downregulated autophagy-associated proteins, and the increase of miR-3651 further suppressed the autophagy process, but with the intervention of BEZ235, the impacts of miR-3651 were completely reversed. Conclusion: miR-3651 intensifies the growth and invasion of HCC cells through activating the PI3K/AKT/mTOR signalling pathway, which is probably a breakthrough in the future diagnosis and therapy of HCC.

2.
Exp Cell Res ; 386(2): 111739, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31759055

RESUMEN

Chemo-resistance to conventional therapy is a major barrier requiring further investigation in hepatocellular carcinoma (HCC). Cancer stem like cells (CSCs) contribute to the tumorigenicity, progression, and chemo-resistance of malignancies. Studies have implicated the anti-cancer effects of arsenic trioxide (ATO) and have explored the underlying mechanisms. However, whether ATO might reverse chemo-resistance by inhibiting the CSC like properties remains under investigation. Here, we explored the potential of ATO in chemotherapy in constructed multiple drug resistant (MDR) liver cancer cells. ATO re-sensitized the MDR Bel-7402 cells (BelMDR) cells to chemotherapeutic drugs, an effect mediated by the inhibition of NF-κB pathway and CSCs properties. For the molecular mechanisms, via inducing the DNA de-methylation, ATO activated the microRNA-148a (miR-148a), leading to the repression of NF-κB pathway by targeting the 3'-UTR of p65. In summary, epigenetic regulation of miR-148a by ATO is an important mechanism in drug resistance that decreases the expression of NF-κB and hence represses CSC like phenotype. These findings may suggest a novel mechanism for HCC treatment.


Asunto(s)
Antineoplásicos/farmacología , Trióxido de Arsénico/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Factor de Transcripción ReIA/genética , Regiones no Traducidas 3' , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Epigénesis Genética , Fluorouracilo/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , MicroARNs/metabolismo , Células Madre Neoplásicas , Oxaliplatino/farmacología , Transducción de Señal , Factor de Transcripción ReIA/metabolismo
3.
Cell Death Discov ; 5: 120, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31341646

RESUMEN

Sorafenib resistance is one of the main obstacles to the treatment of advanced/recurrent hepatocellular carcinoma (HCC). Here, sorafenib-resistant HCC cells and xenografts in nude mice were used as experimental models. A cohort of patients with advanced recurrent HCC who were receiving sorafenib therapy was used to assess the clinical significance of this therapy. Our data showed that 14-3-3η maintained sorafenib resistance in HCC. An analysis of the underlying molecular mechanisms revealed that 14-3-3η stabilizes hypoxia-inducible factor 1α (HIF-1α) through the inhibition of ubiquitin-dependent proteasome protein degradation, which leads to the maintenance of cancer stem cell (CSC) properties. We further found that microRNA-16 (miR-16) is a competent miRNA that reverses sorafenib resistance by targeting the 3'-UTR of 14-3-3η and thereby inhibits 14-3-3η/HIF-1α/CSC properties. In HCC patients, significant negative correlations were found between the expression of miR-16 and 14-3-3η, HIF-1α, or CSC properties. Further analysis showed that low miR-16 expression but high 14-3-3η expression can prognosticate sorafenib resistance and poor survival. Collectively, our present study indicated that miR-16/14-3-3η is involved in sorafenib resistance in HCC and that these two factors could be potential therapeutic targets and biomarkers for predicting the response to sorafenib treatment.

4.
J Cell Biochem ; 120(7): 11190-11195, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30775800

RESUMEN

lncRNAs are responsible for a variety of diseases, including gastric cancer (GC). Many recent studies have reported that lncRNAs can serve as crucial regulators of various genes. Nevertheless, the biological function of lncRNA damage induced noncoding (DINO) remained poorly investigated in GC. Therefore, in our present study, the detailed role of DINO was investigated. It was manifested that DINO was significantly downregulated in GC tissues. Then, DINO was modulated by infecting LV-DINO or by LV-shRNA in BGC-823 and MGC-803 cells. Moreover, it was displayed that GC cell proliferation was suppressed by DINO overexpression, whereas silencing DINO increased cell proliferation significantly. For another, it was indicated that DINO dramatically induced apoptotic ratios of BGC-823 and MGC-803 cells, whereas the decrease of DINO depressed GC cell apoptosis. Apart from these, GC cell cycle progression was greatly blocked by LV-DINO. Furthermore, Western blot results displayed that upregulation of DINO elevated p21 expression and Bax expression. Oppositely, inhibition of DINO greatly suppressed p21 and Bax protein expression level. Taken these, DINO might exert a tumor inhibitory role in the progression of GC through modulating p21 and Bax.

5.
Sci Rep ; 6: 36796, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27824145

RESUMEN

Gastric carcinoma (GC) is the second leading cause of cancer-related mortality worldwide. The efficacy of standard chemotherapy for GC, such as cisplatin (CDDP), is dissatisfactory partly due to the toxic/side-effects. Sulforaphane (SFN), which exhibits effective anti-cancer functions, is a phytochemical converted from cruciferous plants. Our present study aimed to identify whether SFN could enhance the anti-cancer effects of low-dose CDDP and to determine the underlying mechanisms. Herein, co-exposure of SFN and CDDP significantly inhibited the viabilities of gastric cancer cells. For the molecular mechanisms, CDDP alone increased the cancer stem cell (CSC)-like properties in gastric cancer cells via activating the interleukin-6 (IL-6)/IL-6 receptor (IL-6R)/signal transducer and activator of transcription 3 (STAT3) signaling. However, SFN could activate the microRNA-124 (miR-124), which directly targets the 3'-untranslated regions (UTR) of the IL-6R and STAT3. Moreover, knockdown of miR-124 eliminated the effects of SFN on CSC-like properties in GC cells, and in turn enhanced the anti-cancer effects of low-dose CDDP. These findings not only suggested a mechanism whereby SFN enhanced the anti-cancer functions of CDDP, but also helped to regard SFN as a potential chemotherapeutic factor in gastric cancer.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Isotiocianatos/farmacología , MicroARNs/metabolismo , Receptores de Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Regiones no Traducidas 3' , Línea Celular Tumoral , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Ligandos , Células Madre Neoplásicas/metabolismo , Interferencia de ARN , Transducción de Señal , Neoplasias Gástricas/tratamiento farmacológico , Sulfóxidos
6.
Oncotarget ; 7(48): 79805-79813, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27806334

RESUMEN

Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality worldwide. Angiogenesis is the major hallmark in NSCLC. So, further elucidation of molecular mechanisms underlying the angiogenesis of NSCLC is urgently needed. Here, we found that microRNA-206 (miR-206) decreased the angiogenic ability in NSCLC via inhibiting the 14-3-3ζ/STAT3/HIF-1α/VEGF pathway. Briefly, 14-3-3ζ bond with phosphorylated-STAT3, and in turn, elevated the expression of HIF-1α. Then, by enhancing the recruitment of HIF-1α to VEGF promoter, 14-3-3ζ increased the angiogenesis. However, miR-206 decreased the angiogenesis by targeting 14-3-3ζ, and inhibiting the STAT3/HIF-1α/VEGF pathway. In NSCLC cell xenograft model, either overexpression of miR-206 or inhibition of 14-3-3ζ inhibited the STAT3/HIF-1α/VEGF pathway and decreased the tumor growth and angiogenesis. Furthermore, there was a negative correlation between miR-206 and 14-3-3ζ in NSCLC specimens. NSCLC patients with low expressions of miR-206 but high expressions of 14-3-3ζ had the worst survival. Collectively, our findings provided the underlying mechanisms of miR-206/14-3-3ζ in tumor growth and angiogenesis, and implicated miR-206 and 14-3-3ζ as potential therapeutic targets for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , MicroARNs/fisiología , Neovascularización Patológica/genética , Proteínas 14-3-3/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/irrigación sanguínea , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
J Hepatol ; 65(5): 953-962, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27210426

RESUMEN

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. The continued search for novel therapeutic strategies for HCC is urgently required. In this study, we aimed to investigate the functions and clinical significance of 14-3-3η protein in HCC. METHODS: Expressions of genes and proteins were determined by quantitative reverse transcription polymerase chain reaction, Western blot, and immunohistochemistry. Their functions were assessed by endothelial cell recruitment, tube formation, wound healing, flow cytometry, immunostaining, immunoprecipitation, and xenograft assay. A tissue microarray followed by univariate and multivariate analyses was performed to indicate the clinical significance. RESULTS: In HCC specimens, overexpression of 14-3-3η was observed not only in tumors but also in intratumoral vessels. In HCC and vascular endothelial cells, 14-3-3η stimulated proliferation and angiogenesis, but attenuated the functions of sorafenib. Briefly, 14-3-3η facilitated the phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2). Then, by binding to the phosphorylated-ERK1/2 (p-ERK1/2), formed a functional positive feed-back loop. A xenograft model showed that, blockage of either 14-3-3η or ERK1/2 inhibited the tumor growth. Finally, tissue microarray analyses showed that overexpression of 14-3-3η, either in tumors or intratumoral vessels, contributed to the poor survival. CONCLUSIONS: The 14-3-3η-ERK1/2 feedback loop played a characteristic growth-promoting role in HCC, not only in tumors but also in intratumoral vessels. Further, 14-3-3η could be a potential therapeutic target for HCC and a biomarker for predicting sorafenib treatment response. LAY SUMMARY: Here we found that, 14-3-3η protein exhibited a characteristic growth-promoting effect in both tumor and intratumoral vessels of hepatocellular carcinoma by interacting with ERK1/2 signaling.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Inductores de la Angiogénesis , Humanos , Inmunohistoquímica , Neovascularización Patológica
8.
Toxicol Sci ; 143(1): 156-64, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25304214

RESUMEN

The elevation of cancer stem cells (CSCs)-like properties is involved in the initiation and progression of various human cancers. Current standard practices for treatment of cancers are less than satisfactory because of CSCs-mediated recurrence. For this reason, targeting the CSCs or the cancer cells with CSCs-like properties has become the new approach for the cancer treatments. In addition to treating leukemia, arsenic trioxide (As2O3) also suppresses other solid tumors. However, the roles of As2O3 in the regulation of CSCs-like properties remain largely uninvestigated. Here by using sphere formation assay, luciferase reporter assay, and some other molecular biology approaches, we found that As2O3 attenuated the CSCs-like properties in human hepatocellular carcinoma (HCC). Briefly, in HCC cells and mice xenograft models, As2O3 improved the expression of miR-491 by DNA-demethylation. MiR-491, which targeted the SMAD3-3'-UTR, decreased the expressions of SMAD3, and inhibited the CSCs-like properties in HCC cells. Knockdown of either miR-491 or SMAD3 attenuated the As2O3-induced inhibition of endogenous transforming growth factor beta signal and the CSCs-like properties. Further, in HCC patients, miR-491 is inversely correlated with the expressions of SMAD3, CD133, and the metastasis/recurrence outcome. By understanding a novel mechanism whereby As2O3 inhibits the CSCs-like properties in HCC, our study would help in the design of future strategies of developing As2O3 as a potential HCC chemopreventive agent when used alone or in combination with other current drugs.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Óxidos/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Regiones no Traducidas 3' , Antígeno AC133 , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Trióxido de Arsénico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Péptidos/genética , Péptidos/metabolismo , Interferencia de ARN , Proteína smad3/genética , Proteína smad3/metabolismo , Factores de Tiempo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Toxicol Lett ; 231(1): 55-61, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25196641

RESUMEN

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related mortality worldwide. Current standard practices for treatment of HCC are less than satisfactory because of metastasis and recurrence, which are primarily attributed to the angiogenesis. So, the anti-angiogenesis treatment has become the new approach for HCC therapy. In addition to treating leukemia, arsenic trioxide (As2O3) also suppresses other solid tumors, including HCC. However, the roles of As2O3 in the angiogenesis potential of HCC cells remain unclear. In our present study, As2O3 attenuated the angiogenic ability by the microRNA-491 (miR-491)-mediated inhibition of TGF-ß/SMAD3/NF-κB signal pathway in MHCC97H and MHCC97L cells. Briefly, in these cells, As2O3 improved the expression of miR-491 via DNA-demethylation; miR-491, which targeted the SMAD3-3'-UTR, decreased the expression/function of SMAD3, leading to the inactivation of NF-κB/IL-6/STAT-3 signaling; knockdown of miR-491 abolished the As2O3-induced inhibitions of the TGF-ß/SMAD3/NF-κB pathway, the VEGF secretion, and the angiogenesis. By understanding a novel mechanism whereby As2O3 inhibits the angiogenic potential in HCC cells, our study would help in the design of future strategies of developing As2O3 as a potential chemopreventive agent when used alone or in combination with other current anticancer drugs.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Arsenicales/farmacología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , FN-kappa B/metabolismo , Neovascularización Patológica , Óxidos/farmacología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Regiones no Traducidas 3' , Trióxido de Arsénico , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , MicroARNs/genética , FN-kappa B/genética , Neovascularización Fisiológica/efectos de los fármacos , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Proteína smad3/genética , Factores de Tiempo , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA