Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Aging (Albany NY) ; 16(11): 9649-9679, 2024 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-38843392

RESUMO

Oesophageal squamous cell carcinoma (ESCC) is one of the most lethal cancers worldwide. Due to the important role of mitochondrial metabolism in cancer progression, a clinical prognostic model based on mitochondrial metabolism and clinical features was constructed in this study to predict the prognosis of ESCC. Firstly, the mitochondrial metabolism scores (MMs) were calculated based on 152 mitochondrial metabolism-related genes (MMRGs) by single sample gene set enrichment analysis (ssGSEA). Subsequently, univariate Cox regression and LASSO algorithm were used to identify prognosis-associated MMRG and risk-stratify patients. Functional enrichment, interaction network and immune-related analyses were performed to explore the features differences in patients at different risks. Finally, a prognostic nomogram incorporating clinical factors was constructed to assess the prognosis of ESCC. Our results found there were differences in clinical features between the MMs-high group and the MMs-low group in the TCGA-ESCC dataset (P<0.05). Afterwards, we identified 6 MMRGs (COX10, ACADVL, IDH3B, AKR1A1, LIAS, and NDUFB8) signature that could accurately distinguish high-risk and low-risk ESCC patients. A predictive nomogram that combined the 6 MMRGs with sex and N stage to predict the prognosis of ESCC was constructed, and the areas under the receiver operating characteristic (ROC) curve at 1, 2 and 3 years were 0.948, 0.927 and 0.848, respectively. Finally, we found that COX10, one of 6 MMRGs, could inhibit the malignant progression of ESCC in vitro. In summary, we constructed a clinical prognosis model based on 6 MMRGs and clinical features which can accurately predict the prognosis of ESCC patients.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Mitocôndrias , Nomogramas , Humanos , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/mortalidade , Carcinoma de Células Escamosas do Esôfago/patologia , Carcinoma de Células Escamosas do Esôfago/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/metabolismo , Prognóstico , Mitocôndrias/genética , Mitocôndrias/metabolismo , Masculino , Feminino , Regulação Neoplásica da Expressão Gênica , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Pessoa de Meia-Idade , Transcriptoma , Perfilação da Expressão Gênica
2.
Cancer Lett ; 582: 216588, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38097132

RESUMO

Lung adenocarcinoma, the most common histological subtype of non-small cell lung cancer, exhibits heterogeneity that enables adaptability, limits therapeutic success, and remains incompletely understood. Our team uncovers that lncRNA related to chemotherapy resistance in lung adenocarcinoma (lncCRLA) is preferentially expressed in lung adenocarcinoma cells with the mesenchymal phenotype. lncCRLA can not enhance chemotherapy resistance in lung adenocarcinoma due to its binding to RIPK1 in exosomes, which is released into intercellular media and transferred by exosomes from mesenchymal-like to epithelial-like cells. However, plasmatic lncCRLA corresponding to tissue lncCRLA functions as a preferred biomarker to reflect the response to chemotherapy and disease progression of lung adenocarcinoma. Through single-cell sequencing, RNA-Mutect technique and spatial transcriptomics, a handful of hybrid EMT cells with elevated lncCRLA are characterized as the origin of lung adenocarcinoma, which are indiscriminated from hybrid EMT cells by the in-depth sequencing. Plasmatic lncCRLA is properly predictive for the preinvasive lesion of lung adenocarcinoma that would evolve to invasive lesion. That notion is confirmed by a brand-new transgenic mouse model in which EMT is tracked by Cre and Dre system. Dasatinib is potential to hinder the spontaneous progression from preinvasive to invasive lesion of lung adenocarcinoma. Together, plasmatic lncCRLA is defined as a brand-new circulating biomarker to predict the occurrence and evolvement of lung adenocarcinoma, a light for early detection of lung adenocarcinoma.


Assuntos
Adenocarcinoma , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Camundongos , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Biomarcadores , Pulmão/patologia , Transição Epitelial-Mesenquimal/genética , Linhagem Celular Tumoral
3.
Mutat Res ; 827: 111832, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37467675

RESUMO

Esophageal squamous cell carcinoma (ESCC) is a malignancy of the alimentary tract resulting in death worldwide. The role and underlying mechanism of hsa-miR-1269a in the progression of ESCC remain unclear. In this study, hsa-miR-1269a was screened by differential expression analysis in TCGA, and its target gene FAM46C was predicted. qRT-PCR was conducted to assay the expression of hsa-miR-1269a and FAM46C in ESCC cells. The results showed that hsa-miR-1269a was upregulated in ESCC tissues and cell lines. Hsa-miR-1269a overexpression stimulated the proliferation, migration, and invasion capacities of ESCC cells, and FAM46C overexpression inhibited these phenotypes. Dual-luciferase assay verified that hsa-miR-1269a could target FAM46C. Next, qRT-PCR and western blot demonstrated that hsa-miR-1269a overexpression downregulated FAM46C. Rescue experiments revealed that hsa-miR-1269a accelerated the malignant progression of ESCC through FAM46C down-regulation. These results indicate that the interaction between hsa-miR-1269a and FAM46C plays a regulatory role in driving the malignant progression of ESCC cells, thereby providing a novel molecular mechanism for understanding ESCC.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , MicroRNAs , Humanos , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , MicroRNAs/genética , Regulação para Cima , Linhagem Celular Tumoral , Proliferação de Células/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica
4.
Oncol Res ; 28(9): 857-872, 2022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-33985619

RESUMO

EMT confers increased metastatic potential and the resistance to chemotherapies to cancer cells. However, the precise mechanisms of EMT-related chemotherapy resistance remain unclear. c-Src-mediated caspase 8 phosphorylation essential for EMT in lung adenocarcinoma cell lines preferentially occurs in cells with the mesenchymal phenotype, resulting in chemoresistance to cisplatin plus paclitaxel in patients with resectable lung adenocarcinoma and a significantly worse 5-year PFS. Cisplatin killed lung adenocarcinoma cells regardless of caspase 8. Paclitaxel-triggered necroptosis in lung adenocarcinoma cells was dependent on the phosphorylation or deficiency of caspase 8, during which FADD interacted with RIPK1 to activate the RIPK1/RIPK3/MLKL signaling axis. Accompanied with c-Src-mediated caspase 8 phosphorylation to trigger EMT, a novel lncRNA named lncCRLA was markedly upregulated and inhibited RIPK1-induced necroptosis by impairing RIPK1RIPK3 interaction via binding to the intermediate domain of RIPK1. Dasatinib mitigated c-Src-mediated phosphorylation of caspase 8-induced EMT and enhanced necroptosis in mesenchymal-like lung adenocarcinoma cells treated with paclitaxel, while c-FLIP knockdown predominantly sensitized the mesenchymal-like lung adenocarcinoma cells to paclitaxel+dasatinib. c-Srccaspase 8 interaction initiates EMT and chemoresistance via caspase 8 phosphorylation and lncCRLA expression, to which the dasatinib/paclitaxel liposome+siFLIP regimen was lethal.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Apoptose , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Fosforilação , Transdução de Sinais
5.
Toxicol Appl Pharmacol ; 433: 115793, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34758370

RESUMO

Transmembrane emp24 protein transport domain containing 3 (TMED3) is a newly identified cancer-related protein in several malignancies. However, its role in carcinogenesis is still controversial. The project was performed to explore the possible function of TMED3 in the carcinogenesis of non-small cell lung cancer (NSCLC). TMED3 were abundantly expressed in NSCLC tissue, and high TMED3 levels predicted reduced survival in NSCLC patients. NSCLC cells with TMED3 silencing proliferated and invaded more slowly, and were more sensitive to the chemotherapy drug cisplatin than control NSCLC cells. TMED3 silencing reduced the activity of Wnt/ß-catenin pathway, associated with the repression of AKT. Restraint of AKT blocked TMED3-overexpression-evoked enhancing effects on Wnt/ß-catenin pathway. Moreover, down-regulating Wnt/ß-catenin activity reversed TMED3-overexpression-evoked enhancing effects on the proliferation and invasion of NSCLC cells. Additionally, inhibition of TMED3 also displayed antitumor effects in vivo in nude mice. Taken together, our data demonstrate that TMED3 exerts a protumor function in NSCLC by enhancing Wnt/ß-catenin signaling by modulating AKT. Our findings demonstrate that TMED3 inhibition displayed outstanding antitumor effects in vitro and in vivo, and may be a candidate target for future exploiting targeted therapies for NSCLC management.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Via de Sinalização Wnt , Células A549 , Animais , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Movimento Celular , Proliferação de Células , Cisplatino/farmacologia , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Carga Tumoral , Proteínas de Transporte Vesicular/genética
6.
Int J Mol Med ; 47(1): 374-386, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33236138

RESUMO

Sphingosine kinase1 (SphK1) is an oncogenic enzyme that regulates tumor cell apoptosis, proliferation and survival. SphK1 has been reported to promote the development of non­small cell lung cancer (NSCLC), although the underlying mechanism remains to be determined. The aim of the present study was to examine the expression and function of SphK1 in NSCLC and to explore the underlying molecular mechanism. The results of the present study demonstrated that SphK1 expression was upregulated in NSCLC tissues and cell lines. Overexpression of SphK1 increased the proliferation and migration of NSCLC cells. Additionally, overexpression of SphK1 induced expression of antiapoptotic and migration­associated genes, such as Bcl­2, matrix metallopeptidase 2 and cyclin D1. Of note, signal transducer and activator of transcription 3 (STAT3) was also activated in the SphK1­overexpressing cells. By treatment with a STAT3 inhibitor, it was demonstrated that the SphK1­induced changes in expression of target genes, as well as the increase in proliferation and migration of NSCLC cells were mediated by STAT3. In conclusion, the effects of SphK1 overexpression on the development of NSCLC were demonstrated to be mediated by the activation of STAT3. These results suggested that inhibition of the SphK1­STAT3 axis may be a potential strategy for the treatment of NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Fator de Transcrição STAT3/metabolismo , Células A549 , Adulto , Idoso , Carcinoma Pulmonar de Células não Pequenas/patologia , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade
7.
Acta Biochim Biophys Sin (Shanghai) ; 52(10): 1071-1080, 2020 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-33085761

RESUMO

Circular RNAs (circRNAs) have good stability and long half-life in blood and other body fluid, and possess regulatory effects on various biological processes as miRNA/RNA-binding protein sponges, or by competing endogenous RNA, indicating their great potential as biomarkers or targets of cancer therapy. In this study, we mainly explored the role and mechanism of circular RNA SMARCA5 (circsSMARCA5) in non-small cell lung cancer (NSCLC). Quantitative RT-PCR was applied to measure the expression levels of genes, and then, the relationships among circsSMARCA5, microRNA-670-5p (miR-670-5p), and RBM24 were further analyzed. Animal and cell experiments were performed to explore the functions of circsSMARCA5 in NSCLC cells. The results showed that circsSMARCA5 was expressed at low level in NSCLC tissues and cells, while miR-670-5p had high level in NSCLC tissues. Dual luciferase reporter assay verified that miR-670-5p was the target of circsSMARCA5, and RBM24 has the binding site of miR-670-5p. Further analysis showed that circsSMARCA5 could negatively regulate miR-670-5p and had positive relationship with RBM24. Moreover, circsSMARCA5 obviously inhibited tumor growth in vivo, reduced cell proliferation and increased cell apoptosis in vitro, while miR-670-5p mimic or RBM24 knockdown could reverse these effects. Thus, circsSMARCA5 may serve as an NSCLC suppressor by regulating the miR-670-5p/RBM24 axis, and it may have the potential to be a biomarker or therapeutic target for NSCLC.


Assuntos
Adenosina Trifosfatases/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Proteínas Cromossômicas não Histona/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , RNA Circular/genética , Proteínas de Ligação a RNA/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteínas Cromossômicas não Histona/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Circular/metabolismo , Proteínas de Ligação a RNA/genética , Ensaio Tumoral de Célula-Tronco
8.
Pathol Res Pract ; 215(11): 152617, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31563286

RESUMO

BACKGROUND: Recent studies have demonstrated that aberrant expression or activation of kinases results in oncogenesis of a wide range of cancers including GBM. Inhibition of kinases expression induces a reduction of therapy resistance. In this study, we investigate the underlying mechanism by which glioblastoma (GBM) cells acquire resistance to Temozolomide (TMZ) through Aurora kinase B (AURKB) thus to identify novel therapeutic targets and prognostic biomarkers for GBM. METHODS: AURKB was identified as a key candidate kinase-encoding gene in chemoresistance regulation by using kinome-wide bioinformatic analysis. Afterwards, the potential biological functions of AURKB in oncogenesis and chemoresistance were investigated by lentivirus-dependent silencing of AURKB combined with qRT-PCR, western blot and in vivo intra-cranial xenograft mice models. Additionally, immunohistochemistry (IHC) assays were performed to explore the clinical significance of AURKB in glioma patients. Lastly, Chou-Talalay method was used to confirm the synergistic effect of TMZ combined with AURKB inhibitor. RESULTS: AURKB was among the most significantly up-regulated kinase-coding genes in TMZ resistant GBM cells according to database GSE68029, moreover, an increased expression of AURKB was closely associated with poor prognosis in glioma and GBM patients. AURKB knock-down resensitized U87 resistant cells to TMZ both in vitro and in vivo. Additionally, the combination of TMZ and Hesperadin, a specific AURKB inhibitor, significantly suppressed the proliferation of TMZ resistant GBM cells thus dramatically prolonged the survival of xenograft mice viaa synergistic effect with TMZ. CONCLUSION: Elevated AURKB expression was strongly correlated to TMZ resistant acquisition and poor prognosis, furthermore, targeting AURKB would be a potential therapeutic target for GBM patients.


Assuntos
Aurora Quinase B/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/patologia , Indóis/farmacologia , Sulfonamidas/farmacologia , Temozolomida/farmacologia , Animais , Antineoplásicos/farmacologia , Aurora Quinase B/efeitos dos fármacos , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Sinergismo Farmacológico , Glioblastoma/enzimologia , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
BMC Cancer ; 19(1): 670, 2019 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-31286874

RESUMO

BACKGROUND: In epithelial cells, tyrosine kinases induce tyrosine phosphorylation and ubiquitination of the E-cadherin complex, which is responsible for the epithelial-mesenchymal transition (EMT). However, the precise mechanisms remain unclear. METHODS: Protein antibody microarray analysis and E3 ligase profiling were performed to detect the unique E3 ligase underlying E-cadherin downregulation in lung adenocarcinoma tissues. Gene knockdown was performed using viral shRNA. Immunoblotting, immunofluorescence, immunoprecipitation, and xenograft models in vivo were integratively applied to explore RNF43-induced EMT in lung adenocarcinoma cell lines. RESULTS: Protein antibody microarray analysis and E3 ligase profiling revealed that the RING finger protein 43 (RNF43) was linked to E-cadherin downregulation within the context of c-Src activation in lung adenocarcinoma tissues. In addition, the c-Src-Caspase-8 interaction markedly increased c-Src activity. Activated c-Src phosphorylated E-cadherin at the tyrosine 797 site to initiate RNF43-mediated E-cadherin ubiquitination at lysine 816 and subsequent degradation, thus allowing the nuclear translocation of ß-catenin and upregulation of Vimentin and RNF43 expression in lung adenocarcinoma cells. Decreased E-cadherin expression and increased Vimentin expression induced the EMT phenotype and promoted tumor metastasis. The Frizzled 8 (Frz8)-RNF43-induced ubiquitination of phosphorylated E-cadherin was blocked by a monoclonal antibody against the cysteine-rich domain (CRD) of Frz8 but not by antibodies against the protease domain (PA) of RNF43. CONCLUSIONS: Our data suggest that RNF43 participates in the regulation of EMT in the metastasis of lung adenocarcinoma through the ubiquitination and degradation of phosphorylated E-cadherin by activated c-Src.


Assuntos
Adenocarcinoma de Pulmão/secundário , Antígenos CD/metabolismo , Proteína Tirosina Quinase CSK/metabolismo , Caderinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/patologia , Proteínas Oncogênicas/metabolismo , Células A549 , Adenocarcinoma de Pulmão/metabolismo , Animais , Anticorpos Monoclonais , Caspase 8/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/imunologia , Seguimentos , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Oncogênicas/imunologia , Fosforilação , Proteólise , Receptores de Superfície Celular/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Vimentina/metabolismo , beta Catenina/metabolismo
10.
J Cell Biochem ; 120(8): 13717-13725, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30938888

RESUMO

Esophageal squamous cell carcinoma (ESCC) is one of the most common types of esophageal cancer, which is the sixth leading cause of cancer death globally. Homeobox D10 (HOXD10) is a member of the homeobox (HOX) gene family and has been reported to act as a tumor suppressor. However, the potential role of HOXD10 in ESCC has not been reported. Thus, the aim of this study was to examine the expression and function of HOXD10 in ESCC. The expressions of HOXD10 in human ESCC tissues and cell lines were detected by quantitative reverse transcription polymerase chain reaction and Western blot. The HOXD10 overexpressing cell lines were established, then CCK-8 and Transwell assays were performed to examine cell proliferation, migration, and invasion, respectively. The expression of EMT-related proteins and signaling pathway-related proteins were detected by Western blot. Our results showed that HOXD10 is lowly expressed in ESCC tissues as well as in ESCC cell lines. Ectopic overexpression of HOXD10 inhibited cell proliferation, migration, and invasion of ESCC cells (P < 0.05). HOXD10 overexpression repressed the epithelial-mesenchymal transition (EMT) process in ESCC cells. Besides, HOXD10 overexpression suppressed the activation of PI3K/AKT/mTOR signaling pathway. PI3K/Akt agonist, insulin-like growth factor-1, reversed the inhibitory effects of HOXD10 on cell proliferation and migration in ESCC cells. Additional in vivo study proved that ectopic expression of HOXD10 caused an obvious inhibitory effect on the tumor growth. These findings indicated that overexpression of HOXD10 suppressed the proliferation, migration, and invasion via regulating the PI3K/AKT/mTOR signaling pathway in ESCC cells. Thus, targeting HOXD10 may be considered as a therapeutic strategy for ESCC treatment.


Assuntos
Movimento Celular , Proliferação de Células , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular Tumoral , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Proteínas de Homeodomínio/genética , Humanos , Transdução de Sinais/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética
11.
Anticancer Agents Med Chem ; 18(3): 388-393, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29110625

RESUMO

BACKGROUND: CXC Chemokine Receptor 4 (CXCR4) and NFE-related factor 2 (Nrf2) have been observed implicated with cell malignant behavior of human cancers. AIMS: In this study, we detected their expression in gastric carcinoma (GC) tissue specimens and related the result with clinicopathological data and patient survival. METHODS: 120 GC and compared normal tissue specimens were processed to analyse the expression of CXCR4 and Nrf2. We found that the expression of CXCR4 and Nrf2 was dramatically increased in GC tissues when compared to the distant non-cancer tissues (P<0.05). CXCR4 overexpression was associated with the depth of invasion (P= 0.006), Histological grade (P=0.018), TNMstage (P= 0.021), lymph node metastasis (P < 0.001) and distant metastasis (P=0.026), whereas overexpression of Nrf2 protein was significantly associated with tumor size (P=0.045), Histological grade (P=0.026), TNMstage (P= 0.020), lymph node metastases (P < 0.001) and distant metastasis (P=0.008). Furthermore, we observed a significant co-expression of CXCR4 and Nrf2 expression in GC specimens. RESULTS: In the survival part, we found that GC patients with CXCR4+ and Nrf2+ had worse outcomes. The significant prognostic indicators are age, tumor size, histological grade, TNMstage, CXCR4, Nrf2, and coexpression of CXCR4 and Nrf2 in GC patients. Multivariate analysis showed that TNMstage and CXCR4+/Nrf2+ expression were risk factors. Above all we come to the conclusion that the expression of CXCR4 might partly be regulated by the level of Nrf2 and both positive expressions suggest poor prognosis of GC patients.


Assuntos
Fator 2 Relacionado a NF-E2/genética , Receptores CXCR4/genética , Neoplasias Gástricas/genética , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Fator 2 Relacionado a NF-E2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores CXCR4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/metabolismo , Análise de Sobrevida
12.
Biomed Pharmacother ; 91: 402-407, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28472755

RESUMO

Rab3D is a member of the ras-related GTP-binding protein Rab family and was found up-regulated in several types of cancer. However, little is known about the role of Rab3D in carcinogenesis and progression of esophageal squamous cell carcinoma (ESCC). Thus, in this study, we investigated the expression patterns and functional roles of Rab3D in human ESCC. We demonstrated that Rab3D was highly expressed in human ESCC cell lines. In addition, knockdown of Rab3D significantly inhibited the proliferation of ESCC cells and reduced the tumorigenesis in vivo. Moreover, knockdown of Rab3D significantly suppressed ESCC cell migration/invasion and accordingly alerted EMT related markers, which including up-regulated E-cadherin and down-regulated N-cadherin in ESCC cells. Finally, knockdown of Rab3D inhibited the levels of p-PI3K and p-Akt in ECA-109 cells. In conclusion, our data demonstrated that Rab3D functions as an oncogene in ESCC and knockdown of Rab3D suppressed ESCC cell proliferation and invasion, potentially through the PI3K/Akt signaling pathway. Overall, these findings suggest that targeting the Rab3D may be a potential therapeutic target for treatment of ESCC.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/patologia , Inativação Gênica , Proteínas rab3 de Ligação ao GTP/genética , Animais , Carcinogênese/genética , Carcinogênese/patologia , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos Nus , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas rab3 de Ligação ao GTP/metabolismo
13.
Oncol Res ; 25(6): 887-895, 2017 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-28244855

RESUMO

DEAD (Asp-Glu-Ala-Asp) box protein 5 (DDX5), a prototypical member of the DEAD/H-box protein family, has been involved in several human malignancies. However, the expression and biological role of DDX5 in esophageal cancer (EC) remain largely unknown. In this study, we examined the role of DDX5 in regulating EC cell proliferation and tumorigenesis and explored its possible molecular mechanism. We found that DDX5 was overexpressed in human EC cell lines. In addition, knockdown of DDX5 significantly inhibited the proliferation of EC cells in vitro and the growth of EC xenografts in vivo. Knockdown of DDX5 also suppressed the migration/invasion and epithelial-to-mesenchymal transition (EMT) phenotype in EC cells. Furthermore, we observed that knockdown of DDX5 inhibited the expression of ß-catenin, c-Myc, and cyclin D1 in EC cells. In conclusion, our findings provide the first evidence that siRNA-DDX5 inhibited the proliferation and invasion of EC cells through suppressing the Wnt/ß-catenin signaling pathway. Therefore, DDX5 may be a novel potential therapeutic target for the prevention and treatment of EC.


Assuntos
RNA Helicases DEAD-box/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células/genética , RNA Helicases DEAD-box/metabolismo , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos Endogâmicos BALB C , Via de Sinalização Wnt/genética , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/genética , beta Catenina/metabolismo
14.
Oncol Res ; 25(2): 249-257, 2017 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-27629392

RESUMO

Ubiquitin-specific protease 14 (USP14), one of three proteasome-associated deubiquitinating enzymes (DUBs), plays an essential role in the development of human carcinoma. However, to the best of our knowledge, the role of USP14 in esophageal squamous cell carcinoma (ESCC) is unknown. In the current study, we investigated the expression and role of USP14 in ESCC. Our results showed that the level of USP14 was significantly increased in ESCC tissues and cell lines. Downregulation of USP14 significantly inhibited ESCC cell proliferation and ESCC tumor growth in nude mice. Downregulation of USP14 also suppressed the migration/invasion in ESCC cells. Mechanically, downregulation of USP14 decreased the protein expression levels of ß-catenin, cyclin D1, and c-Myc in ESCC cells. In conclusion, our study shows that USP14 plays an important role in the progression and metastasis of ESCC. Therefore, these data suggest that USP14 may be a potentially useful therapeutic strategy for the treatment of ESCC.


Assuntos
Carcinogênese/metabolismo , Carcinoma de Células Escamosas/metabolismo , Proliferação de Células/fisiologia , Neoplasias Esofágicas/metabolismo , Técnicas de Silenciamento de Genes , Ubiquitina Tiolesterase/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Feminino , Técnicas de Silenciamento de Genes/métodos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ubiquitina Tiolesterase/deficiência , Ubiquitina Tiolesterase/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
15.
Oncol Res ; 24(1): 9-15, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27178817

RESUMO

The ribosomal protein (RP)-p53 pathway has been shown to play a key role in apoptosis and senescence of cancer cells. miR-1908 is a newly found miRNA that was reported to have prognostic potential in melanoma. However, its role and mechanism in the progression of non-small cell lung cancer (NSCLC) are largely unknown. In this study, we found that expression of miR-1908 was significantly downregulated in human NSCLC cell lines, including SK-MES-1, A549, and NCI-H460. Then the role of miR-1908 in NSCLC cell proliferation was explored. The miR-1908 mimic was transfected into NSCLC cell lines, and their proliferation was detected. MTT and Cell Titer-Blue H analyses showed that the cell proliferation was notably reduced by the miR-1908 mimic transfection. Moreover, we found the RP-p53 pathway was activated by miR-1908 mimic. Moreover, the miR-1908 inhibitor transfection had a completely opposite effect on the NSCLC cell proliferation than that of miR-1908 mimic. To explore the underlying mechanism of that, TargetScan bioinformatics server and 3'-UTR luciferase reporter assay were applied to identify the targets of miR-1908. Our results showed that AKT1 substrate 1 (AKT1S1), a newly proven suppressor of the RP-p53 pathway, was a target of miR-1908, suggesting a probable mechanism for miR-191 suppressing NSCLC cell proliferation. Our findings provide a novel molecular target for the regulation of NSCLC cell proliferation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células/genética , Hipóxia/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteína Supressora de Tumor p53/genética , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose/genética , Linhagem Celular Tumoral , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Transfecção/métodos
16.
Mol Med Rep ; 12(5): 7367-73, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26461141

RESUMO

The incidence of non­small cell lung cancer (NSCLC) has significantly increased in China, while the prognosis of affected patients is poor. The pathogenesis of NSCLC is thought to be regulated by microRNAs (miRs). The present study used a miR array in order to determine the expression of miR­361­5p, which was significantly lower in NSCLC tissues compared with that in adjacent tissues, indicating a crucial role of miR­361­5p during the progression of NSCLC. Furthermore, the effects of transfection-induced upregulation of miR­361­5p on the NSCLC cell line H23 were assessed. Overexpression of miR­361­5p inhibited the proliferation and colony formation ability of H23 cells. In addition, apoptosis of H23 cells was induced by upregulation of miR­361­5p. Furthermore, signal transducer and activator of transcription 6 (STAT6) was confirmed as a direct target of miR­361­5p by a dual­luciferase reporter assay. Moreover, inhibition of STAT6 by small interfering RNA or miR­361­5p also decreased the expression of B-cell lymphoma extra large (Bcl-xL). In vivo, miR­361­5p significantly reduced tumor growth in a nude mouse xenograft model, and suppressed STAT6 and Bcl-xL expression. In conclusion, the present study indicated that miR­361­5p may represent a novel molecular tool for therapeutic and diagnostic strategies in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Pulmão/patologia , MicroRNAs/genética , Fator de Transcrição STAT6/genética , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/patologia , Camundongos Nus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...