Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cancer Sci ; 110(3): 962-972, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30657231

RESUMEN

Radiotherapy has been widely used for the clinical management of esophageal squamous cell carcinoma. However, radioresistance remains a serious concern that prevents the efficacy of esophageal squamous cell carcinoma (ESCC) radiotherapy. REV7, the structural subunit of eukaryotic DNA polymerase ζ, has multiple functions in bypassing DNA damage and modulating mitotic arrest in human cell lines. However, the expression and molecular function of REV7 in ESCC progression remains unclear. In this study, we first examined the expression of REV7 in clinical ESCC samples, and we found higher expression of REV7 in ESCC tissues compared to matched adjacent or normal tissues. Knockdown of REV7 resulted in decreased colony formation and increased apoptosis in irradiated Eca-109 and TE-1 cells coupled with decreased tumor weight in a xenograft nude mouse model postirradiation. Conversely, overexpression of REV7 resulted in radioresistance in vitro and in vivo. Moreover, silencing of REV7 induced increased reactive oxygen species levels postirradiation. Proteomic analysis of REV7-interacting proteins revealed that REV7 interacted with peroxiredoxin 2 (PRDX2), a well-known antioxidant protein. Existence of REV7-PRDX2 complex and its augmentation postirradiation were further validated by immunoprecipitation and immunofluorescence assays. REV7 knockdown significantly disrupted the presence of nuclear PRDX2 postirradiation, which resulted in oxidative stress. REV7-PRDX2 complex also assembled onto DNA double-strand breaks, whereas REV7 knockdown evidently increased double-strand breaks that were unmerged by PRDX2. Taken together, the present study sheds light on REV7-modulated radiosensitivity through interacting with PRDX2, which provides a novel target for ESCC radiotherapy.


Asunto(s)
Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Proteínas Mad2/genética , Peroxirredoxinas/genética , Tolerancia a Radiación/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estrés Oxidativo/genética , Proteómica/métodos , Especies Reactivas de Oxígeno/metabolismo
2.
Pak J Med Sci ; 30(5): 982-5, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25225511

RESUMEN

OBJECTIVE: To study the relationship between TLR4 and NF-κB p65 protein expressions in tumor tissues after radiotherapy and clinical radiosensitivity of patients with esophageal squamous cell carcinoma. METHODS: A total of 93 patients with esophageal squamous cell carcinoma first treated in our hospital by radiotherapy and surgeries from November 2010 to December 2013 were selected. They were then divided into a severe reaction group, a moderate reaction group and a mild reaction group according to the postoperative pathological examination results of tumor tissues. The expressions of TLR4 and NF-κB p65 in the tumor samples were detected by Western blotting. RESULTS: Compared with the severe reaction group, the expression levels of TLR4 and NF-κB p65 in the moderate reaction group significantly increased (P<0.05). Similarly, the expression levels of the mild reaction group were significantly higher than those of the moderate reaction group (P<0.05). CONCLUSION: Reducing the expression levels of TLR4 and NF-κB p65 proteins may increase the radiosensitivity of patients with esophageal cancer.

3.
Cancer Sci ; 104(11): 1544-51, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23919806

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the deadliest malignancies worldwide. Ying Yang 1 (YY1), a ubiquitously expressed GLI-Krüppel zinc finger transcription factor, plays a regulatory role in a variety of fundamental biological processes, such as embryonic development, growth, apoptosis, differentiation and oncogenic transformation. The purpose of this study was to investigate the expression of YY1 in normal and cancerous esophageal tissues and its function in ESCC development. We found that the expression of YY1 mRNA was significantly increased in the tumor tissues, compared with the para-tissues or normal esophageal tissues. The increased expression of YY1 in tumor samples was further confirmed by immunohistochemistry. Furthermore, the overexpression of YY1 conferred radioresistance to the ESCC TE-1 cells and resulted in markedly reduced cell proliferation. Accordingly, the small interfering RNA-mediated silencing of YY1 expression in TE-1 cells resulted in increased proliferation by enhancing the binding of P21 to Cyclin D1 and CDK4, a protein complex known to mediate cell cycle progression. Moreover, besides P21, heme oxygenase-1 (HO-1) was identified as a YY1 downstream effector, as YY1 stimulated HO-1 expression in esophageal cancer cells. YY1 mediated biological function through transcription of HO-1. Forced expression of HO-1 could moderately suppress proliferation of TE-1 cells. The expression of YY1 significantly correlated with that of HO-1 in ESCC tissues. Taken together, we demonstrated overexpression of YY1 in esophageal carcinoma and identified HO-1 as its target.


Asunto(s)
Carcinogénesis/ultraestructura , Carcinoma de Células Escamosas/enzimología , Neoplasias Esofágicas/enzimología , Hemo-Oxigenasa 1/metabolismo , Factor de Transcripción YY1/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinoma de Células Escamosas/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de la radiación , Neoplasias Esofágicas/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Hemo-Oxigenasa 1/genética , Humanos , Tolerancia a Radiación , Regulación hacia Arriba , Factor de Transcripción YY1/genética
4.
Cell Death Discov ; 8(1): 270, 2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35614065

RESUMEN

Radiotherapy is a main modality of esophageal squamous cell carcinoma (ESCC) treatment, while radioresistance largely limits the effect of this therapy. Evidence exists reporting that SOX17 may sensitize ESCC cells to irradiation, but the downstream mechanism remains poorly understood. Therefore, we attempt to explore the molecular basis of SOX17 effect on radioresistance in ESCC. The SOX17 expression was measured in ESCC tissues and cells, followed by evaluation of its relationship with patient survival. The fractionated irradiation-induced irradiation-resistant cell line KYSE150R was subjected to gain- and loss-of function studies to explore the effect of SOX17 and downstream effectors MALAT1, miR-199a, and HIF1α on the malignant phenotypes of ESCC. The interaction among these factors was explained using ChIP, dual luciferase reporter, RNA pull-down and RIP assays. Further, the in vivo effect of SOX17 on ESCC irradiation tolerance was assessed in nude mice. SOX17 was underexpressed in ESCC tissues and cells, which was negatively correlated with the prognosis of patients with ESCC. Besides, SOX17 inhibited irradiation tolerance of ESCC cells by suppressing MALAT1 transcription. Notably, MALAT1 acted as miR-199a sponge and thereby enhanced HIF1α expression. Moreover, SOX17 reduced the irradiation tolerance of ESCC cells by reducing HIF1α expression via the MALAT1-miR-199a axis, and attenuated tumor formation in nude mice. Our results indicate that SOX17 can impede the radioresistance of ESCC cells through the MALAT1-miR-199a-HIF1α axis, in support of further research for ESCC radiotherapy.

5.
Gene ; 715: 144015, 2019 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-31357025

RESUMEN

Tripartite Motif Containing 13 (TRIM13), a member of TRIM proteins, is deleted in multiple tumor types, especially in B-cell chronic lymphocytic leukemia and multiple myeloma. The present study explored the expression and potential role of TRIM13 in non-small-cell lung carcinoma (NSCLC). We found that TRIM13 mRNA and protein expression was reduced in NSCLC tissues and cell lines in comparison to paired non-cancerous tissues and a human normal bronchial epithelial cell line, respectively. Overexpression of TRIM13 in NCI-H1975 and SPC-A-1 cells hampered cell proliferation. Additionally, TRIM13 overexpression increased the levels of cleaved caspase-3. TRIM13-induced NSCLC cell apoptosis was attenuated by a caspase-3 inhibitor Ac-DEVD-CHO, suggesting that TRIM13 induced cell apoptosis partially through a caspase-3-dependent pathway. Moreover, it has been reported that TRIM13 can regulate nuclear factor kappaB (NF-κB) activity. Our data showed that TRIM13 overexpression inactivated NF-κB as indicated by the increased cytosolic NF-κB and decreased nuclear NF-κB. Exposure to an NF-κB inhibitor PDTC significantly blocked the impact of TRIM13 knockdown on cell proliferation and apoptosis, indicating the functions of TRIM13 in NSCLC cells were mediated by the NF-κB pathway. Finally, we demonstrated that TRIM13 overexpression suppressed tumor growth and induced cell apoptosis in vivo by using a xenograft mouse model. Collectively, our results indicate that TRIM13 behaves as a tumor suppressor in NSCLC through regulating NF-κB pathway. Our findings may offer a promising therapeutic target for NSCLC.


Asunto(s)
Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas de Unión al ADN/genética , Femenino , Xenoinjertos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , FN-kappa B/genética , Trasplante de Neoplasias , Transducción de Señal , Proteínas Supresoras de Tumor/genética
6.
Am J Transl Res ; 11(7): 4077-4088, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396319

RESUMEN

Lung cancer is one of the most common human cancers and is the leading cause of cancer-related mortality. Previous studies have suggested that IL-22 might promote the survival of human lung cancer cells. However, the source of IL-22 and the regulatory mechanism of lung cancer cell proliferation remain unclear. In this study, we found that the expression of IL-22 was upregulated in non-small-cell lung cancer (NSCLC) tumor specimens, as revealed by RT-qPCR analysis. Furthermore, IL-22 was profoundly elevated in cell cultures of primary cancer-associated fibroblasts (CAFs) compared to the levels in cell cultures of normal fibroblasts. Moreover, treatment with the supernatant of CAF cell cultures significantly increased the proliferation, migration and invasion of A549 and H1650 cells but reduced apoptosis via the activation of PI3K-Akt-mTOR signaling, and the application of an anti-IL-22 antibody can partially block the effects induced by the CAF cell culture supernatant. Finally, we also identified a panel of critical genes with differential expression between A549 cells treated with and without IL-22. In summary, our results demonstrate a novel regulatory function of IL-22 secreted by CAFs in NSCLC and provide a potential therapeutic target for treating lung cancer.

7.
Oncol Rep ; 39(3): 993-1002, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29328411

RESUMEN

Esophageal cancer (EC) is the eighth most common highly aggressive cancer worldwide. The purpose of this study was to investigate the effect of the DNA methyltransferase inhibitor RG108 on the radiosensitivity of EC cells. MTT and clonogenic assays were performed to assess the effect of RG108 on the proliferation and radiosensitivity of Eca­109 and TE­1 human EC cells. The cell cycle progression and alterations in apoptosis were analyzed by flow cytometry. For the in vivo analysis, the Eca­109 cells were inoculated into nude mice to establish tumors. Tissues from xenografts were obtained to detect changes to microvessels and tumor growth by immunohistochemistry (IHC). RNA-seq was used to identify differentially expressed genes. We found that RG108 increased the radiosensitivity of EC cells. Apoptosis and G2/M-phase arrest were induced by X-ray irradiation and were significantly enhanced by RG108. In addition, growth of tumor xenografts from the Eca­109 cells was significantly inhibited by irradiation in combination with RG108. The RNA-seq analysis revealed that, compared with radiation alone, X-ray irradiation in combination with RG108 altered the expression of 121 genes in multiple pathways, including the TGF-ß signaling pathway and the Epstein-Barr virus infection pathway. In conclusion, RG108 induced radiosensitivity in EC cells both in vitro and in vivo.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Neoplasias Esofágicas/radioterapia , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ftalimidas/farmacología , Tolerancia a Radiación/efectos de los fármacos , Triptófano/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Biomarcadores de Tumor , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Neoplasias Esofágicas/patología , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Triptófano/farmacología , Células Tumorales Cultivadas , Rayos X , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Oncol Lett ; 14(5): 5681-5687, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29113195

RESUMEN

Lung cancer was demonstrated to be the most lethal type of malignant tumor amongst humans in the global cancer statistics of 2012. As one of the primary treatments, radiotherapy has been reported to induce remission in, and even cure, patients with lung cancer. However, the side effects of radiotherapy may prove lethal in certain patients. In past decades, the transforming growth factor ß1 (TGFB1) signaling pathway has been revealed to serve multiple functions in the control of lung cancer progression and the radiotherapy response. In mammals, this signaling pathway is initiated through activation of the TGFB1 receptor complex, which signals via cytoplasmic SMAD proteins or other downstream signaling pathways. Multiple studies have demonstrated that TGFB1 serves important functions in lung cancer radiotherapy. The present study summarized and reviewed recent progress in elucidating the function of the TGFB1 signaling pathway in predicting radiation pneumonitis, as well as current strategies for targeting the TGFB1 signaling pathway in lung cancer radiotherapy, which may provide potential targets for lung cancer therapy.

9.
Oncol Lett ; 13(2): 704-714, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28356949

RESUMEN

The aim of the present study was to investigate the association between single-nucleotide polymorphisms (SNPs) in X-ray repair cross-complementing 1-399 (XRCC1-399) or excision repair cross-complementation group 1-118 (ERCC1-118) and the short-term efficacy of radiochemotherapy, tumor metastasis and relapse, as well as the survival time in patients with esophageal squamous cell carcinoma (ESCC). TaqMan probe-based quantitative polymerase chain reaction (qPCR) was conducted to examine the levels of XRCC1-399 and ERCC1-118 SNPs in the peripheral blood of 50 patients with pathologically confirmed ESCC. In addition, the associations between different genotypes and short-term therapeutic efficacy [the complete remission (CR) rate], tumor metastasis and relapse, as well as the survival time following concurrent radiochemotherapy, were determined. A total of 50 ESCC patients who received concurrent radiochemotherapy were enrolled. It was found that the short-term therapeutic efficacy (CR rate) was higher in the group of patients carrying the homozygous mutation of XRCC1-399 (A/A genotype) than in the group of patients without the XRCC1-399 mutation (G/G genotype). In addition, the CR rate was significantly increased in patients carrying one or two ERCC1-118 C alleles (C/C or C/T genotype) compared with patients lacking the C allele (T/T genotype). The differences were statistically significant (A/A vs. G/G, P=0.014; TT vs. C/T+C/C, P=0.040). During the follow-up period, the group of patients carrying the homozygous mutation of XRCC1-399 (A/A genotype) exhibited a markedly reduced risk of metastasis and relapse compared with the group of patients carrying non-mutated XRCC1-399 (G/G genotype; P=0.031). By contrast, ERCC1-118 SNP was not associated with the risk of metastasis and recurrence (P>0.05). The combined results of univariate and multivariate Cox regression analysis showed that the SNP in ERCC1-118 was closely associated with survival time. The mean survival time was significantly prolonged in patients carrying 1 or 2 C alleles (C/C or C/T genotype) compared with patients lacking the C allele (T/T genotype) [T/T vs. C/C, HR=12.96, 95% confidence interval (CI)=3.08-54.61, P<0.001; TT vs. C/T+C/C, HR=11.71, 95% CI=3.06-44.83, P<0.001]. However, XRCC1-399SNP had no effect on survival time (P>0.05). XRCCl-399 SNP was associated with the short-term therapeutic efficacy (the CR rate) and tumor metastasis/relapse in ESCC patients who received the docetaxel plus cisplatin (TP) regimen-based concurrent radiochemotherapy. By contrast, ERCC1-118 SNP was significantly associated with the short-term therapeutic efficacy (the CR rate) and survival time in ESCC patients who received TP regimen-based concurrent radiochemotherapy.

10.
J Cancer ; 8(4): 646-656, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28367244

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the deadliest malignancies worldwide. Radiotherapy plays a critical role in the curative management of inoperable ESCC patients. However, radioresistance restricts the efficacy of radiotherapy for ESCC patients. The molecules involved in radioresistance remain largely unknown, and new approaches to sensitize cells to irradiation are in demand. Technical advances in analysis of mRNA and methylation have enabled the exploration of the etiology of diseases and have the potential to broaden our understanding of the molecular pathways of ESCC radioresistance. In this study, we constructed radioresistant TE-1 and Eca-109 cell lines (TE-1/R and Eca-109/R, respectively). The radioresistant cells showed an increased migration ability but reduced apoptosis and cisplatin sensitivity compared with their parent cells. mRNA and methylation profiling by microarray revealed 1192 preferentially expressed mRNAs and 8841 aberrantly methylated regions between TE-1/R and TE-1 cells. By integrating the mRNA and methylation profiles, we related the decreased expression of transcription factor Sall2 with a corresponding increase in its methylation in TE-1/R cells, indicating its involvement in radioresistance. Upregulation of Sall2 decreased the growth and migration advantage of radioresistant ESCC cells. Taken together, our present findings illustrate the mRNA and DNA methylation changes during the radioresistance of ESCC and the important role of Sall2 in esophageal cancer malignancy.

11.
J Exp Clin Cancer Res ; 35: 7, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26754670

RESUMEN

BACKGROUND: Lung cancer has long been the most dangerous malignant tumor among males in both well developed and poorly developed countries. Radiotherapy plays a critical role in the curative management of inoperable non-small cell lung cancer (NSCLC) and is also used as a post-surgical treatment in lung cancer patients. Radioresistance is an important factor that limits the efficacy of radiotherapy for NSCLC patients. Increasing evidence suggests that microRNAs (miRNAs) possess diverse cellular regulatory roles in radiation responses. METHODS: In this study, we used miRNA microarray technology to identify serum miRNAs that were differentially expressed before and after radiotherapy in lung cancer patients. We further examined the biological function of miR-208a on cell viability, apoptotic death and cell cycle distribution in human lung cancer cells and explored the probable mechanism. RESULTS: Nine miRNAs, including miR-29b-3p, miR-200a-3p, and miR-126-3p were significantly down-regulated, whereas miR-208a was the only miRNA that was up-regulated in the serum of the patients after radiation treatment (P < 0.05). The expression of miR-208a could be induced by X-ray irradiation in lung cancer cells. Forced expression of miR-208a promoted cell proliferation and induced radioresistance via targeting p21 with a corresponding activation of the AKT/mTOR pathway in lung cancer cells, whereas down-regulation of miR-208a resulted in the opposite effects. In addition, down-regulation of miR-208a increased the percentage of cells undergoing apoptosis and inhibited the G1 phase arrest in NSCLC cells. Moreover, miR-208a from the serum exosome fraction of lung cancer patients could shuttle to A549 cells in a time-dependent manner, which was likely to contribute to the subsequent biological effects. CONCLUSIONS: The present study provides evidence that miR-208a can affect the proliferation and radiosensitivity of human lung cancer cells by targeting p21 and can be transported by exosomes. Thus, miR-208a may serve as a potential therapeutic target for lung cancer patients.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Neoplasias Pulmonares/radioterapia , MicroARNs/genética , Tolerancia a Radiación , Regulación hacia Arriba , Línea Celular Tumoral , Proliferación Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/genética , MicroARNs/sangre , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/efectos de la radiación
12.
Int J Biol Sci ; 12(6): 677-87, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27194945

RESUMEN

Ca(2+)-binding protein of 45 kDa (Cab45), a CREC family member, is reported to be associated with Ca(2+)-dependent secretory pathways and involved in multiple diseases including cancers. Cab45-G, a Cab45 isoform protein, plays an important role in protein sorting and secretion at Golgi complex. However, its role in cancer cell migration remains elusive. In this study, we demonstrate that Cab45-G exhibited an increased expression in cell lines with higher metastatic potential and promoted cell migration in multiple types of cancer cells. Overexpression of Cab45-G resulted in an altered expression of the molecular mediators of epithelial-mesenchymal transition (EMT), which is a critical step in the tumor metastasis. Quantitative real-time PCR showed that overexpression of Cab45-G increased the expression of matrix metalloproteinase-2 and -7 (MMP-2 and MMP-7). Conversely, knock-down of Cab45-G reduced the expression of the above MMPs. Moreover, forced expression of Cab45-G upregulated the level of phosphorylated ERK and modulated the secretion of extracellular proteins fibronectin and fibulin. Furthermore, in human cervical and esophageal cancer tissues, the expression of Cab45-G was found to be significantly correlated with that of MMP-2, further supporting the importance of Cab45-G on regulating cancer metastasis. Taken together, these results suggest that Cab45-G could regulate cancer cell migration through various molecular mechanisms, which may serve as a therapeutic target for the treatment of cancers.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Western Blotting , Proteínas de Unión al Calcio/genética , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Ensayo de Inmunoadsorción Enzimática , Fibronectinas/genética , Fibronectinas/metabolismo , Células HeLa , Humanos , Inmunohistoquímica , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
Oncol Lett ; 12(6): 4671-4676, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28105174

RESUMEN

Radiotherapy is widely used in esophageal squamous cell carcinoma (ESCC) treatment. Promoting the radiation sensitivity of cancer cells is required. Recent studies have shown that sunitinib can inhibit the growth of several cancer lines. However, few studies on the radiosensitive effect of sunitinib on ESCC cells under hypoxic conditions have been conducted. In the present study, the radiosensitive effects of sunitinib on human ESCC cells were assessed, and the underlying mechanisms were explored. ESCC cells were exposed to hypoxia and treated with sunitinib at different concentrations prior to irradiation. Sunitinib potently inhibited ESCC cell proliferation in an MTT assay. In a clonogenic survival assay, sunitinib sensitized hypoxic ESCC cells to radiation, with sensitizing enhancement ratios of 1.31-1.59. In addition, sunitinib promoted the apoptosis of ESCC cells, but did not alter their cell cycle distribution. Radiosensitization was accompanied by inhibition of the radiation-induced upregulation of hypoxia-inducible factor (HIF)-1α and vascular endothelial growth factor (VEGF) expression. Thus, sunitinib confers radiosensitivity to esophageal cancer cells, which is associated with the downregulation of HIF-1α and VEGF expression. Sunitinib can be a promising radiosensitizer for esophageal cancer radiotherapy.

14.
Int J Biol Markers ; 30(4): e347-58, 2015 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-26349663

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) have been emerging as valuable prognostic biomarkers of breast cancer. We therefore summarized recent research into miRNAs involved in human breast cancer and, further, completed a meta-analysis to predict the role of specific miRNAs in the survival of breast cancer patients. METHODS: Studies were identified by searching PubMed, Embase and Web of Science. Descriptive characteristics for studies were described, and an additional meta-analysis for specific miRNAs was performed. Pooled hazard ratios (HRs) and their corresponding 95% confidence intervals (CIs) were calculated. RESULTS: A total of 41 articles including 27 types of miRNAs were found regarding prognostic biomarkers for breast cancer survival, of which, micRNA-21 (miR-21) was the most-studied specific miRNA that appeared repeatedly among the selected classifiers. For the studies evaluating miR-21's association with clinical outcomes, the median HR in the studies was 2.32 (interquartile range [IQR] = 1.04-3.40), and the pooled HR suggested that high expression of miR-21 has a negative impact on overall survival (OS; HR = 1.46, 95% CI, 1.25-1.70; p<0.05) and disease/recurrence-free survival in breast cancer (HR = 1.49, 95% CI, 1.17-1.90; p<0.01). We also found that higher expression levels of miR-210 significantly predicted poorer outcome, with median HR in the reported studies of 4.07 (IQR = 1.54-4.43) and a pooled HR of 2.94 (95% CI, 2.08-4.17; p<0.05). CONCLUSIONS: These results indicate that miRNAs show promising associations with prognosis in breast cancer. Moreover, specific miRNAs such as miR-21 and miR-210 can predict poor survival rates in breast cancer patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Expresión Génica , MicroARNs/metabolismo , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Supervivencia sin Enfermedad , Femenino , Humanos , MicroARNs/genética , Pronóstico , Modelos de Riesgos Proporcionales
15.
Oncol Rep ; 33(2): 705-12, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25435090

RESUMEN

Lung cancer is the leading cause of cancer-related mortality worldwide. microRNAs (miRNAs) are small post-transcriptional regulatory non-coding RNAs that function as oncogenes or tumor suppressors in human cancers. Emerging evidence reveals that deregulation of miRNAs contributes to the progression of human lung cancer, which is the leading cause of cancer-related deaths worldwide. In the present study, we found that upregulation of the miR-212/132 cluster significantly suppressed the growth and focus formation of A549 and H1299 cells. Moreover, forced expression of this cluster conferred radiosensitivity and inhibited the migration of lung cancer cells, whereas downregulation of miR-212/132 reversed the above effects. Furthermore, miR-212/132 overexpression induced cell cycle arrest at the G1/S phase transition of the lung cancer cells, and inhibition of miR-132 and miR-212 abrogated this arrest. In addition, miR-212/132 overexpression increased the percentage of cells undergoing apoptosis. Cells transfected with the miR-212/132 cluster exhibited upregulated p21 expression and reduced cyclin D1 expression. Conversely, cells transfected with the miR-212/132 inhibitor showed reduced expression of p21 and upregulated expression of cyclin D1, suggesting that miR-212/132 may mediate proliferation and cell cycle arrest through p21 and cyclin D1. Our study provides insight into the biological function of the miR-212/132 cluster in lung cancer. The present study may provide a potential therapeutic target for the treatment of lung cancer.


Asunto(s)
Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , Apoptosis , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/efectos de la radiación , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/radioterapia
16.
Sci Rep ; 5: 9476, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25820571

RESUMEN

Radioresistance has been an important factor in restricting efficacy of radiotherapy for non-small cell lung cancer (NSCLC) patients and new approaches to inhibit cancer growth and sensitize irradiation were warranted. Despite the important role of ubiquitin/proteasome system (UPS) during cancer progression and treatment, the expression and biological role of ubiquitin (Ub) in human NSCLC has not been characterized. In this study, we found that ubiquitin was significantly overexpressed in 75 NSCLC tissues, compared to their respective benign tissues by immunohistochemistry (P < 0.0001). Knock-down of ubiquitin by mixed shRNAs targeting its coding genes ubiquitin B (UBB) and ubiquitin C (UBC) suppressed the growth and increased the radiosensitivity in NSCLC H1299 cells. Apoptosis and γ H2AX foci induced by X-ray irradiation were enhanced by knock-down of ubiquitin. Western blot and immunostaining showed that knock-down of ubiquitin decreased the expression and translocation of NF-κB to the nucleus by reduced phospho-IκBα after irradiation. Suppression of ubiquitin decreased the proliferation and radioresistance of H1299 transplanted xenografts in nude mice by promoting apoptosis. Taken together, our results demonstrate the critical role of ubiquitin in NSCLC proliferation and radiosensitivity. Targeting ubiquitin may serve as a potentially important and novel approach for NSCLC prevention and therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular/genética , Ubiquitina/biosíntesis , Animales , Apoptosis/efectos de la radiación , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Ratones , FN-kappa B/biosíntesis , FN-kappa B/genética , Tolerancia a Radiación/genética , Ubiquitina/genética , Rayos X , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Oncol Lett ; 10(3): 1495-1500, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26622697

RESUMEN

The present study investigated the effects of small interfering RNAs (siRNAs) specific to the epidermal growth factor receptor (EGFR) gene, on the radiosensitivity of esophageal squamous cell carcinoma cells. EGFR gene siRNAs (EGFR-siRNA) were introduced into esophageal cancer Eca109 cells using Lipofectamine® 2000. The EGFR messenger (m)RNA expression levels, EGFR protein expression and cell growth were assessed using reverse transcription-polymerase chain reaction analysis, western blot analysis and a Cell Counting Kit-8 (CCK-8), respectively. In addition, colony assays were used to determine the inhibitory effects of X-ray radiation on EGFR-silenced cells. EGFR mRNA and protein levels were reduced in the Eca109 cells transfected with EGFR-siRNA. The relative EGFR mRNA expression levels were reduced to 26.74, 9.52 and 4.61% in Eca109 cells transfected with EGFR-siRNA1, 2 and 3, respectively. These mRNA levels were significantly reduced compared with the those of the control group (42.44%; P<0.0001). Transfection with siRNA3 resulted in the greatest reduction in EGFR mRNA expression, with an inhibition rate of 85%. The relative EGFR protein expression levels were reduced to 24.05, 34.91 and 34.14% in Eca109 cells transfected with EGFR-siRNA1, 2 and 3, respectively. These protein levels were significantly reduced compared with those of the control group (78.57%; P<0.0001). Transfection with siRNA1 resulted in the greatest reduction in EGFR protein expression, with an inhibition rate of 72.84%. This reduction in EGFR expression inhibited the proliferation of Eca109 cells, which was identified using the CCK-8 assay. The proliferation inhibition ratio was 28.2%. The cells treated with irradiation in addition to EGFR-siRNA, demonstrated reduced radiobiological parameters (D0, Dq and SF2) compared with those of cells treated with irradiation only, with a sensitization enhancing ratio of 1.5. In conclusion, suppression of EGFR expression may enhance the radiosensitivity of esophageal cancer Eca109 cells and therefore may represent a promising approach for future clinical practice.

18.
Sci Rep ; 5: 14503, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26412785

RESUMEN

The aim of this study was to investigate the effect of recombinant human endostatin (rh-Endo) in combination with radiation therapy (RT) on esophageal squamous cell carcinoma (ESCC) and explore the potential mechanisms. ECA109-bearing nude mice were administered RT and/or rh-Endo treatment. Tumor volume, survival, hypoxia and vascular parameters were recorded during the treatment schedule and follow-up as measures of treatment response. ESCC cell lines (ECA109 and TE13) and human umbilical vein endothelial cells (HUVECs) were developed to investigate the outcomes and toxicities of rh-Endo and RT in vitro. Hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) were also evaluated. In vivo studies of ECA109-bearing xenografts showed that rh-Endo improved the radioresponse, with normalization of tumor vasculature and a reduction in hypoxia. In vitro studies showed that rh-Endo did not radiosensitize ESCC cell lines but did affect endothelial cells with a time- and dose-dependent manner. Studies of the molecular mechanism indicated that the improved radioresponse might be due to crosstalk between cancer cells and endothelial cells involving HIF and VEGF expression. Our data suggest that rh-Endo may be a potential anti-angiogenic agent in ESCC especially when combined with RT. The improved radioresponse arises from normalization of tumor vasculature and a reduction in hypoxia.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Endostatinas/farmacología , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Neovascularización Patológica/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Proteínas Recombinantes/farmacología , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/efectos de la radiación , Línea Celular Tumoral , Modelos Animales de Enfermedad , Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Clin Exp Med ; 8(2): 1589-97, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25932087

RESUMEN

BACKGROUND: Primary gastric small cell carcinomas (GSCCs) are increasingly identified by endoscopy, and account for 15-20% of all gastric neuroendocrine tumors (NETs). GSCCs have the worst prognosis with the highest rate of metastases. PURPOSE: To provide useful information for clinicians and researchers to better manage patients with GSCC, we studied the clinical features of GSCC and explored the corresponding therapies and prognosis. METHODS: A literature search was conducted through PUBMED, EMBASE, CNKI and WanFang Databases using search terms "stomach" or "gastric" and "small cell carcinoma" or "poorly differentiated neuroendocrine carcinoma", for the period 1999 to 2012. And the cases reported were all from China. Relevant articles were identified through manual review. The reference lists of these articles were reviewed to include further appropriate articles. RESULTS: Two hundred and five eligible cases were analyzed. The median age of patients was 62 years, with a male-to-female ratio of 5.4:1. Of the tumors, 53.17% were located in the upper stomach, 25.37% in the mid, 18.54% in the distal stomach, the remaining 2.93% were found in the total stomach. The mean size was 68mm in maximum diameter, with a range of 15-150 mm. Of the one hundred and thirty-five patients, fifty appeared to be pure GSCCs, eighty-five were mixed. The median overall survival time of 195 patients was 18.50 months. The 1-, 2-, and 5-year average survival rates of 142 patients were 66.75%, 37.13%, and 20.15%, respectively. CONCLUSIONS: GSCC is a rare tumor and it is notoriously aggressive with a strong propensity for both regional and distant spread. Therapies including surgical resection, chemotherapy, and local radiotherapy, by itself or in combination with other treatment, have been used to treat GSCCs in China. To identify the most effective treatment modalities for GSCCs, we still need prospective, multicenter, randomized clinical researches.

20.
Onco Targets Ther ; 7: 1129-34, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25018639

RESUMEN

Lung cancer is a devastating disease that is responsible for around 160,000 deaths each year in United States. The discovery that lung cancer, like most other solid tumors, contains a subpopulation of cancer stem cells or cancer stem-like cells (CSCs/CS-LCs) that if eliminated could lead to a cure has brought new hope. However, the exact nature of the putative lung CSCs/CS-LCs is not known and therefore therapies to eliminate this subpopulation have been elusive. A limited knowledge and understanding of cancer stem cell properties and tumor biology may be responsible for the limited clinical success. In this review we discuss the stemness and plasticity properties of lung cancer cells that are critical aspects in terms of developing effective therapies. We suggest that the available experimental evidence obtained from lung cancer cell lines and patients' derived primary cultures does not support a tumor model consistent with the classical CSC model. Instead, all lung cancer cells may be extremely versatile and new models of cancer stem cells may be better working models.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA