Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres











Base de données
Gamme d'année
3.
Cell Death Dis ; 9(2): 130, 2018 01 26.
Article de Anglais | MEDLINE | ID: mdl-29374140

RÉSUMÉ

The δ opioid receptor (DOR) is involved in the regulation of malignant transformation and tumor progression of hepatocellular carcinoma (HCC). However, regulation of the DOR in HCC remains poorly defined. We found that miR-874 was identified as a negative regulator of the DOR, which is a direct and functional target of miR-874 via its 3' untranslated region (UTR). Moreover, miR-874 was downregulated in HCC and its expression was inversely correlated with DOR expression. Downregulation of miR-874 was also associated with larger tumor size, more vascular invasion, a poor TNM stage, poor tumor differentiation, and inferior patient outcomes. Functionally, overexpression of miR-874 in the HCC cell line SK-hep-1 inhibited cell growth, migration, in vitro invasion, and in vivo tumorigenicity. Furthermore, miR-874 overexpression suppressed the DOR, resulting in a downregulated epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase (ERK) phosphorylation. The EGFR activator-epidermal growth factor (EGF)-can rescue the proliferation and migration suppression induced by miR-874 overexpression, and the rescue effects of the EGF were blocked by an ERK inhibitor. Our study results suggest that miRNA-874 is a negative regulator of the DOR that can suppress tumor proliferation and metastasis in HCC by targeting the DOR/EGFR/ERK pathway, which may be a potential target for HCC treatment.


Sujet(s)
Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Récepteurs ErbB/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Système de signalisation des MAP kinases , microARN/génétique , Récepteur delta/métabolisme , Régions 3' non traduites/génétique , Animaux , Séquence nucléotidique , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation négative/génétique , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Souris de lignée BALB C , Souris nude , microARN/métabolisme , Adulte d'âge moyen , Invasion tumorale , Métastase tumorale , Analyse de survie
4.
Onco Targets Ther ; 9: 6987-6998, 2016.
Article de Anglais | MEDLINE | ID: mdl-27920552

RÉSUMÉ

Recent studies have shown that deubiquitination plays a key role in tumor progression, metastasis, resistance to chemotherapy drugs, and prognosis. In this study, we investigated the effects of the deubiquitinating enzyme USP22 on the expression of the drug-resistance genes BMI1 and EZH2 in hepatocellular carcinoma (HCC) and on prognosis. Downregulation of USP22 expression with interference ribonucleic acid in resistant HCC cell lines with high USP22 expression resulted in decreased BMI1 expression, but had no effect on EZH2 expression. USP22, BMI1, and EZH2 were highly expressed in HCC tissue, and the expression levels were positively correlated with tumor grade and clinical stage. Correlation analysis showed that USP22 expression was positively correlated with that of BMI1. Kaplan-Meier analysis showed that high levels of USP22 and BMI1 expression were associated with poor overall survival and relapse-free survival in all of the cases and in patients treated with transcatheter arterial chemoembolization. These results suggested that high levels of USP22 expression played an important role in drug resistance to chemotherapeutic drugs in HCC patients by upregulating the expression of BMI1; therefore, coexpression of USP22 and BMI1 may become a new predictor for HCC prognosis and may help guide clinical treatment.

5.
Cancer Res ; 76(22): 6520-6532, 2016 11 15.
Article de Anglais | MEDLINE | ID: mdl-27651311

RÉSUMÉ

The Jumonji domain-containing chromatin remodeling factor JMJD3 has important roles in development and cancer. Here, we report a pivotal role for JMJD3 in sustaining the phenotype of aggressive hepatocellular carcinomas. Expression levels of JMJD3 in clinical specimens of hepatocellular carcinoma correlated inversely with patient survival. In hepatocellular carcinoma cells, we found that enforcing its overexpression induced epithelial-mesenchymal transition (EMT), invasive migration, stem cell-like traits, and metastatic properties. Conversely, silencing JMJD3 in hepatocellular carcinoma cells overexpressing it inhibited these aggressive phenotypes. Mechanistically, JMJD3 modulated H3K27me3 in the SLUG gene promoter, a histone mark associated with active SLUG transcription. SLUG silencing blocked JMJD3-induced EMT, stemness, and metastasis. Furthermore, SLUG expression in hepatocellular carcinoma clinical specimens correlated positively with JMJD3 expression. Our results establish JMJD3 as a critical driver of hepatocellular carcinoma stem cell-like and metastatic behaviors, with implications for prognosis and treatment. Cancer Res; 76(22); 6520-32. ©2016 AACR.


Sujet(s)
Carcinome hépatocellulaire/génétique , Régulation de l'expression des gènes tumoraux/génétique , Jumonji Domain-Containing Histone Demethylases/métabolisme , Tumeurs du foie/génétique , Animaux , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire , Humains , Tumeurs du foie/anatomopathologie , Souris , Invasion tumorale , Régulation positive
6.
Mol Cancer ; 15(1): 57, 2016 09 06.
Article de Anglais | MEDLINE | ID: mdl-27600149

RÉSUMÉ

BACKGROUND: Liver cancer is one of the main causes of cancer-related death in human. HOXA7 has been proved to be related with several cancers. METHOD: The expression levels of HOXA7 were examined by Western blot, qRT-PCR or ICH. MTT was used to detect the proliferative rate of liver cancer cells. The invasive abilities were examined by matrigel and transwell assay. The metastatic abilities of liver cancer cells were revealed in BALB/c nude mice. RESULTS: In this report, we revealed that HOXA7 promoted metastasis of HCC patients. First, increased levels of HOXA7 were examined in liver cancer especially in metastatic liver cancer. Moreover, higher expression level of HOXA7 was associated with poorer prognosis of liver cancer patients. Overexpression of HOXA7 significantly enhanced proliferation, migration, invasion in vitro and tumor growth and metastasis in vivo meanwhile silencing HOXA7 significantly inhibited the aboves abilities of liver cancer cells. In this research, we identified that HOXA7 performed its oncogenic characteristics through activating Snail. CONCLUSION: Collectively, we identify the critical role and possible mechanism of HOXA7 in metastasis of liver cancer which suggest that HOXA7 may be a potential therapeutic target of liver cancer patients.


Sujet(s)
Carcinome hépatocellulaire/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Tumeurs du foie/métabolisme , Facteurs de transcription de la famille Snail/métabolisme , Régulation positive , Animaux , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du foie/génétique , Souris , Souris de lignée BALB C , Invasion tumorale , Métastase tumorale , Transplantation tumorale , Pronostic
7.
Oncol Rep ; 36(5): 2579-2586, 2016 Nov.
Article de Anglais | MEDLINE | ID: mdl-27665747

RÉSUMÉ

δ­opioid receptor (DOR) belongs to the family of G protein­coupled receptors (GPCRs). Numerous studies have shown that DOR is widely distributed in human peripheral tissues and is closely related to the development and progression of certain malignant tumours. However, there is controversy in the literature regarding whether DOR has an impact on the development and progression of human breast cancer. The present study comprehensively elaborates on the biological functions of DOR by determining the distribution of DOR expression in breast cancer tissues and cells and by further verifying the effects of DOR on breast cancer progression. DOR was found to be highly expressed in human breast cancer tissues and cells. In addition, the high expression level of DOR positively correlated with tumour grade and clinical stage and negatively correlated with breast cancer metastasis and prognosis. Upregulating and activating DOR promoted the proliferation of human breast cancer cells in a concentration­dependent manner within a specific concentration range, whereas downregulating or inhibiting DOR activation significantly suppressed cell proliferation. The majority of tumour cells were arrested in G1 phase, and some cells exhibited apoptosis. DOR upregulation and activation induced protein kinase C (PKC) activation, resulting in increased phosphorylation levels of extracellular signal­regulated kinases (ERKs). After inhibition of the PKC/ERK signalling pathway, the effects of DOR on breast cancer were significantly attenuated in vivo and in vitro. In summary, DOR is highly expressed in breast cancer and is closely related to its progression. These results suggest that DOR may serve as a potential biomarker for the early diagnosis of breast cancer and may be a viable molecular target for therapeutic intervention.


Sujet(s)
Tumeurs du sein/génétique , Prolifération cellulaire/génétique , Protéine kinase C/génétique , Récepteur delta/biosynthèse , Activation de la transcription/génétique , Adulte , Sujet âgé , Animaux , Apoptose/génétique , Tumeurs du sein/anatomopathologie , Points de contrôle du cycle cellulaire/génétique , Évolution de la maladie , Femelle , Humains , Système de signalisation des MAP kinases/génétique , Cellules MCF-7 , Souris , Adulte d'âge moyen , Phosphorylation , Protéine kinase C/biosynthèse , Récepteur delta/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Drug Des Devel Ther ; 10: 841-50, 2016.
Article de Anglais | MEDLINE | ID: mdl-26966354

RÉSUMÉ

Characterized by its acute onset, critical condition, poor prognosis, and high mortality rate, severe acute pancreatitis (SAP) can cause multiple organ failure at its early stage, particularly acute lung injury (ALI). The pathogenesis of ALI is diffuse alveolar damage, including an increase in pulmonary microvascular permeability, a decrease in compliance, and invasion of many inflammatory cells. Corticosteroids are the main treatment method for ALI; however, the associated high toxicity and side effects induce pain in patients. Recent studies show that the effective components in many traditional Chinese medicines can effectively inhibit inflammation with few side effects, which can decrease the complications caused by steroid consumption. Based on these observations, the main objective of the current study is to investigate the effect of alpinetin, which is a flavonoid extracted from Alpinia katsumadai Hayata, on treating lung injury induced by SAP and to explore the mechanism underlying the alpinetin-mediated decrease in the extent of ALI. In this study, we have shown through in vitro experiments that a therapeutic dose of alpinetin can promote human pulmonary microvascular endothelial cell proliferation. We have also shown via in vitro and in vivo experiments that alpinetin upregulates aquaporin-1 and, thereby, inhibits tumor necrosis factor-α expression as well as reduces the degree of lung injury. Overall, our study shows that alpinetin alleviates SAP-induced ALI. The likely molecular mechanism includes upregulated aquaporin expression, which inhibits tumor necrosis factor-α and, thus, alleviates SAP-induced ALI.


Sujet(s)
Lésion pulmonaire aigüe/complications , Lésion pulmonaire aigüe/traitement médicamenteux , Aquaporine-1/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Flavanones/pharmacologie , Pancréatite/complications , Pancréatite/traitement médicamenteux , Régulation positive/effets des médicaments et des substances chimiques , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Alpinia/composition chimique , Animaux , Aquaporine-1/déficit , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/isolement et purification , Médicaments issus de plantes chinoises/usage thérapeutique , Cellules endothéliales/effets des médicaments et des substances chimiques , Flavanones/composition chimique , Flavanones/isolement et purification , Flavanones/usage thérapeutique , Humains , Mâle , Médecine traditionnelle chinoise , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Rats , Rat Sprague-Dawley , Relation structure-activité , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Facteur de nécrose tumorale alpha/biosynthèse
9.
Oncotarget ; 6(32): 32586-601, 2015 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-26452129

RÉSUMÉ

Increasing evidence reveals that aberrant expression of microRNA contributes to the development and progression of colon cancer, but the roles of microRNA-506 (miR-506) in colon cancer remain elusive. Here, we demonstrated that miR-506 was down-regulated in colon cancer tissue and cells and that miR-506 expression was inversely correlated with EZH2 expression, tumor size, lymph node invasion, TNM stage and metastasis. A high level of miR-506 identified patients with a favorable prognosis. In vitro and in vivo experiments confirmed that miR-506 inhibits the proliferation and metastasis of colon cancer, and a luciferase reporter assay confirmed that EZH2 is a direct and functional target of miR-506 via the 3'UTR of EZH2. The restoration of EZH2 expression partially reversed the proliferation and invasion of miR-506-overexpressing colon cancer cells. Moreover, we confirmed that the miR-506-EZH2 axis inhibits proliferation and metastasis by activating/suppressing specific downstream tumor-associated genes and the Wnt/ß-catenin signaling pathway. Taking together, our study sheds light on the role of miR-506 as a suppressor for tumor growth and metastasis and raises the intriguing possibility that miR-506 may serve as a new potential marker for monitoring and treating colon cancer.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Mouvement cellulaire , Tumeurs du côlon/enzymologie , microARN/métabolisme , Complexe répresseur Polycomb-2/métabolisme , Adulte , Sujet âgé , Animaux , Marqueurs biologiques tumoraux/génétique , Prolifération cellulaire , Tumeurs du côlon/génétique , Tumeurs du côlon/mortalité , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/thérapie , Protéine-2 homologue de l'activateur de Zeste , Femelle , Régulation de l'expression des gènes codant pour des enzymes , Régulation de l'expression des gènes tumoraux , Cellules HCT116 , Cellules HEK293 , Cellules HT29 , Humains , Estimation de Kaplan-Meier , Métastase lymphatique , Mâle , Souris de lignée BALB C , Souris nude , microARN/génétique , Adulte d'âge moyen , Métastase tumorale , Stadification tumorale , Complexe répresseur Polycomb-2/génétique , Interférence par ARN , Facteurs temps , Transfection , Résultat thérapeutique , Voie de signalisation Wnt
10.
Oncotarget ; 6(14): 12723-39, 2015 May 20.
Article de Anglais | MEDLINE | ID: mdl-25909289

RÉSUMÉ

JARID1B is a member of the family of JmjC domain-containing proteins that removes methyl residues from methylated lysine 4 on histone H3 lysine 4 (H3K4). JARID1B has been proposed as an oncogene in many types of tumors; however, its role and underlying mechanisms in hepatocellular carcinoma (HCC) remain unknown. Here we show that JARID1B is elevated in HCC and its expression level is positively correlated with metastasis. In addition Kaplan-Meier survival analysis showed that high expression of JARID1B was associated with decreased overall survival of HCC patients. Overexpression of JARID1B in HCC cells increased proliferation, epithelial-mesenchymal transition, migration and invasion in vitro, and enhanced tumorigenic and metastatic capacities in vivo. In contrast, silencing JARID1B in aggressive and invasive HCC cells inhibited these processes. Mechanistically, we found JARID1B exerts its function through modulation of H3K4me3 at the PTEN gene promoter, which was associated with inactive PTEN transcription. PTEN overexpression blocked JARID1B-driven proliferation, EMT, and metastasis. Our results, for the first time, portray a pivotal role of JARID1B in stimulating metastatic behaviors of HCC cells. Targeting JARID1B may thus be a useful strategy to impede HCC cell invasion and metastasis.


Sujet(s)
Carcinome hépatocellulaire/anatomopathologie , Transition épithélio-mésenchymateuse/génétique , Jumonji Domain-Containing Histone Demethylases/biosynthèse , Tumeurs du foie/anatomopathologie , Invasion tumorale/génétique , Protéines nucléaires/biosynthèse , Protéines de répression/biosynthèse , Adulte , Animaux , Marqueurs biologiques tumoraux/analyse , Technique de Western , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/mortalité , Lignée cellulaire tumorale , Immunoprécipitation de la chromatine , Femelle , Hétérogreffes , Humains , Immunohistochimie , Jumonji Domain-Containing Histone Demethylases/analyse , Estimation de Kaplan-Meier , Tumeurs du foie/génétique , Tumeurs du foie/mortalité , Mâle , Souris , Microscopie confocale , Adulte d'âge moyen , Invasion tumorale/anatomopathologie , Protéines nucléaires/analyse , Phosphohydrolase PTEN/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines de répression/analyse , RT-PCR , Transduction du signal/physiologie , Transcriptome
11.
Transpl Int ; 28(6): 751-60, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25611689

RÉSUMÉ

Liver ischemia-reperfusion injury (IRI) is a major cause of morbidity and mortality after resection surgery, liver transplantation, and hemorrhagic and septic shock. Mir-155 is upregulated by a broad range of inflammatory mediators, and it has been demonstrated to be involved in both innate and adaptive immune responses. However, the role of mir-155 in liver IRI has never been investigated. In this study, mir-155 deficiency protected mice from liver IRI, as shown by lower serum alanine aminotransferase (ALT) levels and Suzuki scores. Mir-155 deficiency results in the development of M2 macrophages, which respond to IR-induced innate immune stimulation by producing a regulatory inflammatory response with higher level of IL-10, but lower levels of TNF-α, IL-6, and IL-12p40. Mir-155 deficiency suppresses IL-17 expression, which contributes to the liver IRI development. In our further in vitro study, the results show that the Th17 differentiation is inhibited by SOCS1 overexpression and the promoted M2 macrophage development induced by mir-155 deficiency is abolished by SOCS1 knockdown. In conclusion, mir-155 deficiency attenuates liver IRI through upregulation of SOCS1, and this was associated with promoted M2 macrophage and inhibited Th17 differentiation.


Sujet(s)
Transplantation hépatique , microARN/génétique , Lésion d'ischémie-reperfusion/immunologie , Immunité acquise , Alanine transaminase/sang , Animaux , Lymphocytes T CD4+/cytologie , Différenciation cellulaire , Séparation cellulaire , Modèles animaux de maladie humaine , Cytométrie en flux , Immunité innée , Inflammation , Interleukine-10/métabolisme , Sous-unité p40 de l'interleukine-12/métabolisme , Interleukine-6/métabolisme , Cellules de Küpffer/cytologie , Foie/métabolisme , Macrophages/métabolisme , Mâle , Souris , Souris de lignée C57BL , microARN/physiologie , Myeloperoxidase/métabolisme , Protéine-1 suppressive de la signalisation des cytokines , Protéines SOCS/génétique , Cellules Th17/cytologie , Facteur de nécrose tumorale alpha/métabolisme , Régulation positive
12.
Anal Bioanal Chem ; 406(7): 1985-98, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24429977

RÉSUMÉ

Extraction of qualitative and quantitative information from large numbers of analytical signals is difficult with drifted baselines, particularly in multivariate analysis. Baseline drift obscures and "fuzzies" signals, and even deteriorates analytical results. In order to obtain accurate and clear results, some effective methods should be proposed and implemented to perform baseline correction before conducting further data analysis. However, most of the classic methods require user intervention or are prone to variability, especially with low signal-to-noise signals. In this study, a novel baseline correction algorithm based on quantile regression and iteratively reweighting strategy is proposed. This does not require user intervention and prior information, such as peak detection. The iteratively reweighting strategy iteratively changes weights of residuals between fitted baseline and original signals. After a series of tests and comparisons with several other popular methods, using various kinds of analytical signals, the proposed method is found to be fast, flexible, robust, and easy to use both in simulated and real datasets.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE