Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
1.
Cell Death Discov ; 7(1): 135, 2021 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-34091600

RÉSUMÉ

Phosphoglycerate kinase (PGK) is involved in glycolytic and various metabolic events. Dysfunction of PGK may induce metabolic reprogramming and the Warburg effect. In this study, we demonstrated that PGK1, but not PGK2, may play a key role in tumorigenesis and is associated with metastasis. We observed an inverse correlation between PGK1 and the survival rate in several clinical cohorts through bioinformatics statistical and immunohistochemical staining analyses. Surprisingly, we found that PGK1 was significantly increased in adenocarcinoma compared with other subtypes. Thus, we established a PGK1-based proteomics dataset by a pull-down assay. We further investigated HIV-1 Tat Specific Factor 1 (HTATSF1), a potential binding partner, through protein-protein interactions. Then, we confirmed that PGK1 indeed bound to HTATSF1 by two-way immunoprecipitation experiments. In addition, we generated several mutant clones of PGK1 through site-directed mutagenesis, including mutagenesis of the N-terminal region, the enzyme catalytic domain, and the C-terminal region. We observed that even though the phosphoglycerate kinase activity had been inhibited, the migration ability induced by PGK1 was maintained. Moreover, our immunofluorescence staining also indicated the translocation of PGK1 from the cytoplasm to the nucleus and its colocalization with HTATSF1. From the results presented in this study, we propose a novel model in which the PGK1 binds to HTATSF1 and exerts functional control of cancer metastasis. In addition, we also showed a nonenzymatic function of PGK1.

2.
Cell Death Dis ; 11(3): 195, 2020 03 18.
Article de Anglais | MEDLINE | ID: mdl-32188842

RÉSUMÉ

Drug resistance remains a serious issue of clinical importance and is a consequence of cancer stemness. In this study, we showed that the level of Aldolase A (ALDOA) expression is significantly associated with the IC50 value of chemotherapy drugs in lung cancer. Our data revealed that ALDOA overexpression resulted in a significant increase of lung tumor spheres. The use of ingenuity pathway analysis (IPA) resulted in the identification of POU5F1 (Oct4) as the leading transcription factor of ALDOA. We observed high expression of ALDOA, Oct4 and stemness markers in collected spheroid cells. DUSP4 and TRAF4 were confirmed as major downstream targets of the ALDOA-Oct4 axis. Knockdown of these molecules significantly decreased the stemness ability of cells. In addition, we investigated whether miR-145 targets the 3'-UTR of Oct4 and is regulated by ALDOA due to the involvement of ALDOA in glycolysis and metabolic reprogramming. Furthermore, we constructed several mutant forms of ALDOA that disrupted its enzymatic activity and showed that they still induced significant in vitro sphere formation and in vivo tumorigenicity. These results demonstrated that ALDOA-mediated spheroid formation is independent of its enzymatic activity. In the clinical component, we also showed that the combination of ALDOA and TRAF4 or DUSP4 is positively correlated with poor overall survival in a xenograft model and cancer patients through immunohistochemical analyses. The results of our study revealed novel functional roles of ALDOA in inducing cancer stemness via the inhibition of miR-145 expression and the activation of Oct4 transcription. These findings offer new therapeutic strategies for modulation of lung cancer stemness to enhance chemotherapeutic responses in lung cancer patients.


Sujet(s)
Fructose bisphosphate aldolase/métabolisme , Tumeurs du poumon/métabolisme , microARN/métabolisme , Cellules souches tumorales/métabolisme , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire/physiologie , Régulation négative , Dual-specificity phosphatases/métabolisme , Fructose bisphosphate aldolase/génétique , Hétérogreffes , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mâle , Souris , Souris de lignée NOD , Souris SCID , microARN/génétique , Mitogen-Activated Protein Kinase Phosphatases/métabolisme , Cellules souches tumorales/anatomopathologie , Facteur de transcription Oct-3/métabolisme , Transduction du signal , Facteur-4 associé aux récepteurs de TNF/métabolisme
3.
Cancer Res ; 79(24): 6113-6125, 2019 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-31653686

RÉSUMÉ

Follistatin-like protein 1 (FSTL1) plays a critical role in lung organogenesis, but is downregulated during lung cancer development and progression. The prognostic significance and functional consequences of FSTL1 downregulation in lung cancer are unclear. Here, reduced levels of FSTL1 were detected in various tumors compared with normal tissues and were associated with poor clinical outcome in patients with non-small cell lung cancer, particularly those with lung adenocarcinoma. FSTL1 expression negatively correlated with the metastatic potential of lung cancer cells. Antibody-based neutralization of extracellular FSTL1 increased cellular migration/invasion while addition of recombinant FSTL1 protein diminished the metastatic capacity of lung cancer cells in vitro and in vivo. Notably, treatment with FSTL1 effectively prevented the metastatic progression of lung cancer cells in an orthotopic animal model. Mechanistically, FSTL1 directly bound to the proform of secreted phosphoprotein 1 (SPP1)/osteopontin, restraining proteolytic activation of SPP1, which led to inactivation of integrin/CD44-associated signaling and rearrangement of the actin cytoskeleton. Combined low expression of FSTL1 and high expression of SPP1 predicted a poorer prognosis for patients with lung cancer. This study highlights the novel interaction between FSTL1 and SPP1 and new opportunities to effectively target SPP1-driven metastatic cancers characterized by FSTL1 downregulation. SIGNIFICANCE: These findings describe the novel interaction between FSTL1 and SPP1 and its role in the metastatic progression of lung adenocarcinoma.


Sujet(s)
Adénocarcinome pulmonaire/anatomopathologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines apparentées à la follistatine/métabolisme , Tumeurs du poumon/anatomopathologie , Ostéopontine/métabolisme , Cellules A549 , Cytosquelette d'actine/anatomopathologie , Adénocarcinome pulmonaire/mortalité , Adénocarcinome pulmonaire/chirurgie , Animaux , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/chirurgie , Régulation négative , Femelle , Protéines apparentées à la follistatine/génétique , Techniques de knock-down de gènes , Humains , Poumon/anatomopathologie , Poumon/chirurgie , Tumeurs du poumon/mortalité , Tumeurs du poumon/chirurgie , Mâle , Souris , Pneumonectomie , Pronostic , Liaison aux protéines , Protéolyse , Petit ARN interférent/métabolisme , Protéines recombinantes/métabolisme , Transduction du signal , Analyse de survie
4.
Sci Rep ; 9(1): 14624, 2019 10 10.
Article de Anglais | MEDLINE | ID: mdl-31601833

RÉSUMÉ

The basic leucine zipper and the W2 domain-containing protein 1 (BZW1) plays a key role in the cell cycle and transcriptionally control the histone H4 gene during G1/S phase. Since cellular proliferation rates are frequently dysregulated in human cancers, we identified the characteristics of BZW1 in cancer cells and analyzed its prognostic value in lung cancer patients. By searching public databases, we found that high BZW1 expression was significantly correlated with poor survival rate in non-small cell lung cancer (NSCLC), especially in lung adenocarcinoma. Similar trends were also shown in an array comprising NSCLC patient tissue. Knockdown of BZW1 inhibited cell metastatic ability, but did not affect the cell proliferation rate of NSCLC cells. From transcriptomics data mining, we found that coordination between BZW1 and EGFR overexpression was correlated with a worse outcome for lung cancer patients. In summary, BZW1 expression serves as an independent prognostic factor of NSCLC, especially in lung adenocarcinoma. Overexpression of BZW1 in lung cancer cells revealed a novel pathway underlying the induction of lung cancer metastasis.


Sujet(s)
Adénocarcinome pulmonaire/anatomopathologie , Marqueurs biologiques tumoraux/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines du cycle cellulaire/métabolisme , Protéines de liaison à l'ADN/métabolisme , Tumeurs du poumon/anatomopathologie , Récidive tumorale locale/anatomopathologie , Cellules A549 , Adénocarcinome pulmonaire/mortalité , Sujet âgé , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/génétique , Carcinome pulmonaire non à petites cellules/mortalité , Protéines du cycle cellulaire/analyse , Protéines du cycle cellulaire/génétique , Protéines de liaison à l'ADN/analyse , Protéines de liaison à l'ADN/génétique , Survie sans rechute , Femelle , Techniques de knock-down de gènes , Humains , Immunohistochimie , Estimation de Kaplan-Meier , Poumon/anatomopathologie , Tumeurs du poumon/mortalité , Mâle , Adulte d'âge moyen , Métastase tumorale/anatomopathologie , Récidive tumorale locale/épidémiologie , Études rétrospectives , Taux de survie , Régulation positive
5.
Cancer Res ; 79(18): 4754-4766, 2019 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-31358528

RÉSUMÉ

Cancer metabolic reprogramming promotes tumorigenesis and metastasis; however, the underlying molecular mechanisms are still being uncovered. In this study, we show that the glycolytic enzyme aldolase A (ALDOA) is a key enzyme involved in lung cancer metabolic reprogramming and metastasis. Overexpression of ALDOA increased migration and invasion of lung cancer cell lines in vitro and formation of metastatic lung cancer foci in vivo. ALDOA promoted metastasis independent of its enzymatic activity. Immunoprecipitation and proteomic analyses revealed γ-actin binds to ALDOA; blocking this interaction using specific peptides decreased metastasis both in vitro and in vivo. Screening of clinically available drugs based on the crystal structure of ALDOA identified raltegravir, an antiretroviral agent that targets HIV integrase, as a pharmacologic inhibitor of ALDOA-γ-actin binding that produced antimetastatic and survival benefits in a xenograft model with no significant toxicity. In summary, ALDOA promotes lung cancer metastasis by interacting with γ-actin. Targeting this interaction provides a new therapeutic strategy to treat lung cancer metastasis. SIGNIFICANCE: This study demonstrates the role of aldolase A and its interaction with γ-actin in the metastasis of non-small lung cancer and that blocking this interaction could be an effective cancer treatment.


Sujet(s)
Adénocarcinome pulmonaire/traitement médicamenteux , Antinéoplasiques/pharmacologie , Fructose bisphosphate aldolase/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs du poumon/traitement médicamenteux , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/pharmacologie , Actines/antagonistes et inhibiteurs , Actines/métabolisme , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/secondaire , Animaux , Apoptose , Carcinome à grandes cellules/traitement médicamenteux , Carcinome à grandes cellules/métabolisme , Carcinome à grandes cellules/secondaire , Prolifération cellulaire , Femelle , Fructose bisphosphate aldolase/métabolisme , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Pronostic , Études rétrospectives , Taux de survie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Biochem Biophys Res Commun ; 501(3): 619-627, 2018 06 27.
Article de Anglais | MEDLINE | ID: mdl-29729273

RÉSUMÉ

Lower grade gliomas (LGGs) have highly diverse clinical phenotypes. The histological grade and type are insufficient to accurately predict the clinical outcomes of patients with LGGs. Therefore, identification of biomarkers that can facilitate the prediction of clinical outcomes in LGGs is urgently needed. Gene expression of LOX has been identified as a biomarker for various cancers. However, the clinical significance of LOX expression in LGGs has not been investigated. In this study, we analyzed the glioma RNA-seq dataset from TCGA (The Cancer Genome atlas) and identified lysyl oxidase (LOX) as a potential biomarker for LGGs. Kaplan-Meier survival analysis revealed that high LOX expression is associated with worse overall survival and recurrence free survival in LGG patients. Besides, high LOX expression is associated with poor response to primary therapy, follow-up treatment, targeted molecular therapy, and radiation therapy. Univariate and multivariate Cox regression analyses further confirmed LOX expression as an independent prognostic factor for LGG patients. Finally, we observed that LOX expression is significantly correlated with EMT (epithelial to mesenchymal transition) and IDH1 status in LGGs. In conclusion, our analyses suggest that LOX expression is a potential biomarker for prognosis and therapeutic response in LGGs.


Sujet(s)
Tumeurs du cerveau/génétique , Régulation de l'expression des gènes tumoraux , Gliome/génétique , Lysyloxidase/génétique , Adulte , Tumeurs du cerveau/diagnostic , Tumeurs du cerveau/anatomopathologie , Transition épithélio-mésenchymateuse , Femelle , Gliome/diagnostic , Gliome/anatomopathologie , Humains , Estimation de Kaplan-Meier , Mâle , Pronostic , Lysyloxidase/analyse
7.
Sci Rep ; 7(1): 9830, 2017 08 29.
Article de Anglais | MEDLINE | ID: mdl-28852126

RÉSUMÉ

Follistatin-related protein 1 (FSTL1) plays a critical role in lung development through regulating BMP4-p-Smad1/5/8-Smad4 pathway. Regarding that many developmental pathways in embryogenesis are dysregulated in cancer, we aim to unravel the role of FSTL1-BMP4-Smad pathway in lung cancer. Our results showed low FSTL1 immunoexpression was significantly correlated with poor prognosis while patients with low BMP4 or low Smad4 immunoexpression showed a trend toward poor prognosis. When stratified by different histological types, low FSTL1, BMP4, and Smad4 expression retained their trends in predicting poor prognosis in lung adenocarcinoma (LUAD) but not in lung squamous cell carcinoma (SCC). Low FSTL1, BMP4, and Smad4 expression were more frequently observed in LUAD patients with smoking history. To determine smoking effect on FSTL1, normal cell BEAS2B and lung cancer cell lines was treated with nicotine and the results showed nicotine increased the proliferation of these cells. Interestingly, FSTL1 attenuated nicotine-induced BEAS2B and lung cancer cell line proliferation. Altogether, low FSTL1, BMP4, and Smad4 expression significantly correlated with poor prognosis in LUAD but not in SCC. Frequent decrease of FSTL1 expression in smokers LUAD further indicates its importance and therapeutic potential for lung cancer patients with specific subtypes. FSTL1 may prevent nicotine-induced lung cancer cell proliferation.


Sujet(s)
Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/mortalité , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/mortalité , Protéines apparentées à la follistatine/métabolisme , Transduction du signal , Protéines Smad/métabolisme , Protéine Smad-4/métabolisme , Adénocarcinome pulmonaire/diagnostic , Adénocarcinome pulmonaire/génétique , Kinase du lymphome anaplasique/génétique , Animaux , Marqueurs biologiques , Carcinome épidermoïde/diagnostic , Carcinome épidermoïde/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Modèles animaux de maladie humaine , Femelle , Protéines apparentées à la follistatine/génétique , Expression des gènes , Humains , Immunohistochimie , Estimation de Kaplan-Meier , Mâle , Souris , Mutation , Stadification tumorale , Fusion oncogène , Pronostic , Protéines proto-oncogènes p21(ras)/génétique , Protéines Smad/génétique , Protéine Smad-4/génétique
9.
PLoS One ; 10(7): e0133411, 2015.
Article de Anglais | MEDLINE | ID: mdl-26230665

RÉSUMÉ

BACKGROUND: In lung cancer, uPA, its receptor (uPAR), and the inhibitors PAI-1 and PAI-2 of the plasminogen activator family interact with MMP-2 and MMP-9 of the MMP family to promote cancer progression. However, it remains undetermined which of these markers plays the most important role and may be the most useful indicator to stratify the patients by risk. METHODS: We determined the individual prognostic value of these 6 markers by analyzing a derivation cohort with 98 non-small cell lung cancer patients by immunohistochemical staining. The correlation between the IHC expression levels of these markers and disease prognosis was investigated, and an immunohistochemical panel for prognostic prediction was subsequently generated through prognostic model analysis. The value of the immunohistochemical panel was then verified by a validation cohort with 91 lung cancer patients. RESULTS: In derivation cohort, PAI-2 is the most powerful prognostic factor (HR = 2.30; P = 0.001), followed by MMP-9 (HR = 2.09; P = 0.019) according to multivariate analysis. When combining PAI-2 and MMP-9, the most unfavorable prognostic group (low PAI-2 and high MMP-9 IHC expression levels) showed a 6.40-fold increased risk of a poor prognosis compared to the most favorable prognostic group (high PAI-2 and low MMP-9 IHC expression levels). PAI-2 and MMP-9 IHC panel could more precisely identify high risk patients in both derivation and validation cohort. CONCLUSIONS: We revealed PAI-2 as the most powerful prognostic marker among PA and MMP protease family even after considering their close relationships with each other. By utilizing a combination of PAI-2 and MMP-9, more precise prognostic information than merely using pathological stage alone can be obtained for lung cancer patients.


Sujet(s)
Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Peptide hydrolases/métabolisme , Inhibiteur-2 d'activateur du plasminogène/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Femelle , Humains , Mâle , Matrix metalloproteinase 9/métabolisme , Adulte d'âge moyen , Analyse multifactorielle , Pronostic
10.
Cancer Res ; 74(21): 6280-90, 2014 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-25205106

RÉSUMÉ

Esophageal cancer is an aggressive human malignancy with increasing incidence in the developed world. VEGF-C makes crucial contributions to esophageal cancer progression that are not well understood. Here, we report the discovery of regulatory relationship in esophageal cancers between the expression of VEGF-C and cortactin (CTTN), a regulator of the cortical actin cytoskeleton. Upregulation of CTTN expression by VEGF-C enhanced the invasive properties of esophageal squamous cell carcinoma in vitro and tumor metastasis in vivo. Mechanistic investigations showed that VEGF-C increased CTTN expression by downregulating Dicer-mediated maturation of miR326, thereby relieving the suppressive effect of miR326 on CTTN expression. Clinically, expression of Dicer and miR326 correlated with poor prognosis in patients with esophageal cancer. Our findings offer insights into how VEGF-C enhances the robust invasive and metastatic properties of esophageal cancer, which has potential implications for the development of new biomarkers or therapies in this setting.


Sujet(s)
Carcinome épidermoïde/génétique , Cortactine/métabolisme , Tumeurs de l'oesophage/génétique , microARN/génétique , Facteur de croissance endothéliale vasculaire de type C/génétique , Marqueurs biologiques tumoraux , Carcinome épidermoïde/anatomopathologie , Lignée cellulaire tumorale , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Cortactine/génétique , Évolution de la maladie , Tumeurs de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage , Régulation de l'expression des gènes tumoraux , Humains , Pronostic , Facteur de croissance endothéliale vasculaire de type C/métabolisme
11.
Am J Respir Crit Care Med ; 190(6): 675-87, 2014 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-25152164

RÉSUMÉ

RATIONALE: Metabolic alterations contribute to cancer development and progression. However, the molecular mechanisms relating metabolism to cancer metastasis remain largely unknown. OBJECTIVES: To identify a key metabolic enzyme that is aberrantly overexpressed in invasive lung cancer cells and to investigate its functional role and prognostic value in lung cancer. METHODS: The differential expression of metabolic enzymes in noninvasive CL1-0 cells and invasive CL1-5 cells was analyzed by a gene expression microarray. The expression of target genes in clinical specimens from patients with lung cancer was examined by immunohistochemistry. Pharmacologic and gene knockdown/overexpression approaches were used to investigate the function of the target gene during invasion and metastasis in vitro and in vivo. The association between the target gene expression and clinicopathologic parameters was further analyzed. Bioinformatic analyses were used to discover the signaling pathways involved in target gene-regulated invasion and migration. MEASUREMENTS AND MAIN RESULTS: Squalene synthase (SQS) was up-regulated in CL1-5 cells and in the tumor regions of the lung cancer specimens. Loss of function or knockdown of SQS significantly inhibited invasion/migration and metastasis in cell and animal models and vice versa. High expression of SQS was significantly associated with poor prognosis among patients with lung cancer. Mechanistically, SQS contributed to a lipid-raft-localized enrichment of tumor necrosis factor receptor 1 in a cholesterol-dependent manner, which resulted in the enhancement of nuclear factor-κB activation leading to matrix metallopeptidase 1 up-regulation. CONCLUSIONS: Up-regulation of SQS promotes metastasis of lung cancer by enhancing tumor necrosis factor-α receptor 1 and nuclear factor-κB activation and matrix metallopeptidase 1 expression. Targeting SQS may have considerable potential as a novel therapeutic strategy to treat metastatic lung cancer.


Sujet(s)
Farnesyl-diphosphate farnesyltransferase/métabolisme , Tumeurs du poumon/enzymologie , Tumeurs du poumon/secondaire , Microdomaines membranaires/métabolisme , Invasion tumorale/physiopathologie , Récepteur au facteur de nécrose tumorale de type I/métabolisme , Animaux , Lignée cellulaire tumorale , Cholestérol/biosynthèse , Modèles animaux de maladie humaine , Farnesyl-diphosphate farnesyltransferase/génétique , Régulation de l'expression des gènes codant pour des enzymes , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du poumon/anatomopathologie , Matrix metalloproteinase 1/métabolisme , Pronostic , Régulation positive
12.
J Clin Invest ; 123(3): 1082-95, 2013 Mar.
Article de Anglais | MEDLINE | ID: mdl-23434592

RÉSUMÉ

Angiopoietin-like protein 1 (ANGPTL1) is a potent regulator of angiogenesis. Growing evidence suggests that ANGPTL family proteins not only target endothelial cells but also affect tumor cell behavior. In a screen of 102 patients with lung cancer, we found that ANGPTL1 expression was inversely correlated with invasion, lymph node metastasis, and poor clinical outcomes. ANGPTL1 suppressed the migratory, invasive, and metastatic capabilities of lung and breast cancer cell lines in vitro and reduced metastasis in mice injected with cancer cell lines overexpressing ANGPTL1. Ectopic expression of ANGPTL1 suppressed the epithelial-to-mesenchymal transition (EMT) by reducing the expression of the zinc-finger protein SLUG. A microRNA screen revealed that ANGPTL1 suppressed SLUG by inducing expression of miR-630 in an integrin α(1)ß(1)/FAK/ERK/SP1 pathway-dependent manner. These results demonstrate that ANGPTL1 represses lung cancer cell motility by abrogating the expression of the EMT mediator SLUG.


Sujet(s)
Adénocarcinome/métabolisme , Angiopoïétines/physiologie , Mouvement cellulaire , Tumeurs du poumon/métabolisme , Facteurs de transcription/métabolisme , Adénocarcinome/mortalité , Adénocarcinome/secondaire , Protéine-1 de type angiopoïétine , Protéines semblables à l'angiopoïétine , Angiopoïétines/métabolisme , Animaux , Séquence nucléotidique , Sites de fixation , Lignée cellulaire tumorale , Régulation négative , Transition épithélio-mésenchymateuse , Femelle , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Humains , Intégrine alpha1 bêta1/métabolisme , Estimation de Kaplan-Meier , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Métastase lymphatique , Système de signalisation des MAP kinases , Souris , Souris SCID , microARN/génétique , microARN/métabolisme , Transplantation tumorale , Pronostic , Transduction du signal , Facteurs de transcription de la famille Snail , Facteurs de transcription/génétique , Transcriptome
13.
Ann Acad Med Singap ; 40(7): 319-24, 2011 Jul.
Article de Anglais | MEDLINE | ID: mdl-21870023

RÉSUMÉ

INTRODUCTION: Stage I non-small cell lung cancer (NSCLC) is potentially curable after completely resection, but early recurrence may infl uence prognosis. This study hypothesises that vascular endothelial growth factor C (VEGF-C) plays a key role in predicting early recurrence and poor survival of patients with stage I NSCLC. MATERIALS AND METHODS: The expression of VEGF-C was immuno-histochemically (IHC) analysed in tumour samples of primary stage I NSCLC and correlated to early recurrence (< 36 months), disease-free survival, and overall survival in all 49 patients. RESULTS: Early recurrence was identifi ed in 16 patients (33%), and the early recurrence rate in strong and weak VEGF-C activity was significantly different (P = 0.016). VEGF-C was also an independent risk factor in predicting early recurrence (HR = 3.98, P = 0.02). Patients with strong VEGF-C staining also had poor 3-year disease-free survival (P = 0.008) and overall survival (P = 0.007). CONCLUSION: Strong VEGF-C IHC staining could be a biomarker for predicting early recurrence and poor prognosis of resected stage I NSCLC, if the results of the present study are confirmed in a larger study. A more aggressive adjuvant therapy should be used in this group of patients.


Sujet(s)
Carcinome pulmonaire non à petites cellules/sang , Carcinome pulmonaire non à petites cellules/mortalité , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques/sang , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/chirurgie , Survie sans rechute , Femelle , Humains , Immunohistochimie , Mâle , Adulte d'âge moyen , Stadification tumorale , Pronostic , Analyse de survie , Taïwan , Facteur de croissance endothéliale vasculaire de type A/sang
14.
J Biol Chem ; 285(41): 31325-36, 2010 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-20675382

RÉSUMÉ

Osteoporosis is one of the most common bone pathologies. A number of novel molecules have been reported to increase bone formation including cysteine-rich protein 61 (CYR61), a ligand of integrin receptor, but mechanisms remain unclear. It is known that bone morphogenetic proteins (BMPs), especially BMP-2, are crucial regulators of osteogenesis. However, the interaction between CYR61 and BMP-2 is unclear. We found that CYR61 significantly increases proliferation and osteoblastic differentiation in MC3T3-E1 osteoblasts and primary cultured osteoblasts. CYR61 enhances mRNA and protein expression of BMP-2 in a time- and dose-dependent manner. Moreover, CYR61-mediated proliferation and osteoblastic differentiation are significantly decreased by knockdown of BMP-2 expression or inhibition of BMP-2 activity. In this study we found integrin α(v)ß(3) is critical for CYR61-mediated BMP-2 expression and osteoblastic differentiation. We also found that integrin-linked kinase, which is downstream of the α(v)ß(3) receptor, is involved in CYR61-induced BMP-2 expression and subsequent osteoblastic differentiation through an ERK-dependent pathway. Taken together, our results show that CYR61 up-regulates BMP-2 mRNA and protein expression, resulting in enhanced cell proliferation and osteoblastic differentiation through activation of the α(v)ß(3) integrin/integrin-linked kinase/ERK signaling pathway.


Sujet(s)
Protéine morphogénétique osseuse de type 2/biosynthèse , Différenciation cellulaire/physiologie , Protéine-61 riche en cystéine/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Régulation de l'expression des gènes codant pour des enzymes/physiologie , Intégrine alphaVbêta3/métabolisme , Système de signalisation des MAP kinases/physiologie , Ostéoblastes/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéine-61 riche en cystéine/pharmacologie , Relation dose-effet des médicaments , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Ostéoblastes/cytologie , Facteurs temps
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...