Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Transl Med ; 22(1): 769, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39143573

RÉSUMÉ

BACKGROUND: Although immune checkpoint inhibitors (ICIs) have revolutionized the landscape of cancer treatment, only a minority of colorectal cancer (CRC) patients respond to them. Enhancing tumor immunogenicity by increasing major histocompatibility complex I (MHC-I) surface expression is a promising strategy to boost the antitumor efficacy of ICIs. METHODS: Dual luciferase reporter assays were performed to find drug candidates that can increase MHC-I expression. The effect of nilotinib on MHC-I expression was verified by dual luciferase reporter assays, qRT-PCR, flow cytometry and western blotting. The biological functions of nilotinib were evaluated through a series of in vitro and in vivo experiments. Using RNA-seq analysis, immunofluorescence assays, western blotting, flow cytometry, rescue experiments and microarray chip assays, the underlying molecular mechanisms were investigated. RESULTS: Nilotinib induces MHC-I expression in CRC cells, enhances CD8+ T-cell cytotoxicity and subsequently enhances the antitumor effects of anti-PDL1 in both microsatellite instability and microsatellite stable models. Mechanistically, nilotinib promotes MHC-I mRNA expression via the cGAS-STING-NF-κB pathway and reduces MHC-I degradation by suppressing PCSK9 expression in CRC cells. PCSK9 may serve as a potential therapeutic target for CRC, with nilotinib potentially targeting PCSK9 to exert anti-CRC effects. CONCLUSION: This study reveals a previously unknown role of nilotinib in antitumor immunity by inducing MHC-I expression in CRC cells. Our findings suggest that combining nilotinib with anti-PDL1 therapy may be an effective strategy for the treatment of CRC.


Sujet(s)
Tumeurs colorectales , Pyrimidines , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/immunologie , Tumeurs colorectales/métabolisme , Humains , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Animaux , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antigène CD274/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/génétique , Souris , Instabilité des microsatellites/effets des médicaments et des substances chimiques , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques
2.
Biomed Pharmacother ; 173: 116427, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38484558

RÉSUMÉ

Uncertainty exists regarding the mechanisms by which hypoxia-inducible factors (HIFs) control CD8+T-cell migration into tumor microenvironments. Here, we found that HIF-1α knockdown or overexpression resulted in increased or decreased CXCL9, -10, and -11 expression in vitro, respectively. Gene Set Variation Analysis revealed that elevated HIF-1α levels correlated with a poor prognosis, severe pathological stage, and an absence of CD8+ T cells in the tumor microenvironment in colorectal cancer (CRC) patients. HIF-1α was inversely associated with pathways beneficial to anti-tumor immunotherapy and cytokine/chemokine function. In vivo, inhibiting HIF-1α or its upstream regulator BIRC2 significantly suppressed tumor growth and promoted CD8+ T-cell infiltration. CXCR3 neutralizing antibodies reversed these effects, implicating the involvement of CXCL9, -10, and -11/CXCR3 axis. The presence of HIF-1α weakened the upregulation of CXCL9, -10, and -11 by bleomycin and doxorubicin. Combining HIF-1α inhibition with bleomycin promoted CD8+ T-cell infiltration and tumor suppression in vivo. Moreover, doxorubicin could upregulate CXCL9, -10 and -11 by suppressing HIF-1α. Our findings highlight the potential of HIF-1α inhibition to improve CRC microenvironments and increase chemotherapy sensitivity.


Sujet(s)
Tumeurs colorectales , Résistance aux médicaments antinéoplasiques , Sous-unité alpha du facteur-1 induit par l'hypoxie , Humains , Bléomycine , Lymphocytes T CD8+ , Lignée cellulaire tumorale , Chimiokine CXCL9/génétique , Chimiokine CXCL9/métabolisme , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/immunologie , Tumeurs colorectales/métabolisme , Cytokines , Doxorubicine/pharmacologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Microenvironnement tumoral
3.
Biol Pharm Bull ; 45(9): 1238-1245, 2022.
Article de Anglais | MEDLINE | ID: mdl-36047191

RÉSUMÉ

Despite advances in colorectal cancer (CRC) treatment, most advanced CRC patients who experience disease progression after chemotherapy, targeted therapy, and immunotherapy face a situation in which there is no available medicine. Thus, new therapeutic drugs for CRC are urgently needed. Studies have shown that cholesteryl ester transfer protein (CETP) has a vital role in tumor development and is a possible target for CRC therapy. We found that Evacetrapib, a CETP inhibitor, suppressed CRC cell growth by inhibiting the Wnt/ß-catenin signaling pathway and activating the c-Jun NH2-terminal kinase (JNK) signaling pathway in CRC. Therefore, Evacetrapib displays an anti-cancer effect and is a possible option for treating CRC.


Sujet(s)
Tumeurs colorectales , Voie de signalisation Wnt , Benzodiazépines , Lignée cellulaire tumorale , Prolifération cellulaire , Tumeurs colorectales/anatomopathologie , Régulation de l'expression des gènes tumoraux , Humains , Système de signalisation des MAP kinases , bêta-Caténine/métabolisme
4.
J Immunother Cancer ; 7(1): 210, 2019 08 07.
Article de Anglais | MEDLINE | ID: mdl-31391111

RÉSUMÉ

BACKGROUND: Indoleamine 2,3-dioxygenase 1 (IDO1) is a critical regulator of T cell function, contributing to immune tolerance. Upregulation of IDO1 has been found in many cancer types; however, the regulatory mechanisms and clinical significance of IDO1 in colon cancer are still unclear. Here, we investigated the role of dysregulated microRNA (miRNA) targeting IDO1 in the colon cancer microenvironment. METHODS: We elucidated IDO1 function by performing cell-based assays and establishing transplanted tumor models in BALB/c mice and BALB/c nude mice. We evaluated IDO1 protein expression by immunohistochemistry (IHC) in a tissue microarray (TMA) and analyzed IDO1 mRNA expression with The Cancer Genome Atlas (TCGA). We screened miRNAs targeting IDO1 by using a dual luciferase reporter assay. We tested the function of microRNA-448 (miR-448) by using western blotting (WB) and fluorescence-activated cell sorting (FACS). RESULTS: We demonstrated that stable IDO1 overexpression enhanced xenograft tumor growth in BALB/c mice but not in BALB/c nude mice. We also revealed the involvement of posttranscriptional regulation of IDO1 in colon cancer by observing IDO1 protein levels and mRNA levels. Furthermore, ectopic expression of miRNA mimics suggested that miR-448 could significantly downregulate IDO1 protein expression. Notably, we proved that miR-448 suppressed the apoptosis of CD8+ T cells by suppressing IDO1 enzyme function. CONCLUSION: Our findings indicated that IDO1 suppressed the CD8+ T cell response in colon cancer. miR-448, as a tumor-suppressive miRNA, enhanced the CD8+ T cell response by inhibiting IDO1 expression. The results provide a theoretical basis for the development of new immunotherapy for the treatment of colon cancer.


Sujet(s)
Lymphocytes T CD8+/immunologie , Tumeurs du côlon/immunologie , Indoleamine-pyrrole 2,3,-dioxygenase/immunologie , microARN/immunologie , Animaux , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/physiologie , Tumeurs du côlon/génétique , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Cellules HCT116 , Cellules HT29 , Hétérogreffes , Humains , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs , Indoleamine-pyrrole 2,3,-dioxygenase/génétique , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Mâle , Souris , Souris de lignée BALB C , Souris nude , microARN/génétique , microARN/métabolisme , ARN messager/biosynthèse , ARN messager/génétique , Transfection
5.
Sci Rep ; 6: 27583, 2016 07 01.
Article de Anglais | MEDLINE | ID: mdl-27363391

RÉSUMÉ

This study investigated the function of a chloride channel blocker, DIDS. Both in vitro and in vivo studies found that DIDS significantly inhibits lipopolysaccharide (LPS)-induced release of proin flammatory cytokines. Here, we show that DIDS inhibits LPS-induced inflammation, as shown by downregulation of inflammatory cytokines via inhibition of the TLR4/NF-κB pathway. Furthermore, we show that ClC-3siRNA transfection reduces LPS-induced pro-inflammation in Raw264.7 cells, indicating that ClC-3 is involved in the inhibitory effect of DIDS during LPS-induced cytokines release. In vivo, DIDS reduced LPS-induced mortality, decreased LPS-induced organic damage, and down-regulated LPS-induced expression of inflammatory cytokines. In sum, we demonstrate that ClC-3 is a pro-inflammatory factor and that inhibition of ClC-3 inhibits inflammatory induction both in vitro and in vivo, suggesting that ClC-3 is a potential anti-inflammatory target.


Sujet(s)
Acide 4,4'-diisothiocyano-stilbène-2,2'-disulfonique/pharmacologie , Anti-inflammatoires non stéroïdiens/pharmacologie , Canaux chlorure/antagonistes et inhibiteurs , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Péritonite/traitement médicamenteux , Récepteur de type Toll-4/antagonistes et inhibiteurs , Animaux , Canaux chlorure/génétique , Canaux chlorure/immunologie , Femelle , Régulation de l'expression des gènes , Interleukine-1 bêta/antagonistes et inhibiteurs , Interleukine-1 bêta/génétique , Interleukine-1 bêta/immunologie , Lipopolysaccharides , Mâle , Souris , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/immunologie , Péritonite/induit chimiquement , Péritonite/génétique , Péritonite/anatomopathologie , Cellules RAW 264.7 , Petit ARN interférent/génétique , Petit ARN interférent/immunologie , Transduction du signal , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/immunologie
6.
Immunopharmacol Immunotoxicol ; 37(5): 413-20, 2015.
Article de Anglais | MEDLINE | ID: mdl-26369367

RÉSUMÉ

CONTEXT: Endotoxins including lipopolysaccharide (LPS) could cause endotoxemia which often results in excessive inflammation, organ dysfunction, sepsis, disseminated intravascular coagulation (DIC) or even death. Previously, a novel fibrinogenase (FII) showed protective effects on LPS-induced DIC via activating protein C and suppressing inflammatory cytokines. OBJECTIVE: To evaluate whether FII has protective effect on LPS-induced endotoxemia in mice and learn about the role of NF-κB pathway in TNF-α producing process. METHODS: BALB/C mice were intraperitoneally injected (i.p.) with (a) 30 mg/kg LPS, (b) LPS + 0.3 mg/kg FII, (c) LPS + 1.0 mg/kg FII, (d) LPS + 3.0 mg/kg FII or (e) saline. Both survival rate and organ function were tested, including alanine aminotransferase (ALT), blood urine nitrogen (BUN) and tissue section, and TNF-α was examined by ELISA. RAW 264.7 macrophage was administered with (a) LPS, (b) LPS + FII, (c) FII alone or (d) saline, and TNF-α and phosphorylation (P)-NF-κB (P65) were determined by Western blot. RESULTS: The administration of LPS led to 65% mortality rate, a rise of serum TNF-α, BUN and ALT levels, and both liver and renal tissue damage were observed. While FII treatment significantly increased the survival rate of LPS-induced endotoxemia mice model, histopathology and protein analysis results also revealed that FII remarkably protected liver and renal from LPS damage as well as decreasing TNF-α level. In vitro, FII significantly decreased LPS-induced TNF-α production and the expression of P-NF-κB (P65). CONCLUSIONS: Our findings suggested that FII had protective effect on LPS-induced endotoxemia and organ injuries by suppressing the activation of NF-κB which decreased TNF-α level.


Sujet(s)
Venins de crotalidé/composition chimique , Endotoxémie/induit chimiquement , Endotoxémie/prévention et contrôle , Lipopolysaccharides/toxicité , Serine endopeptidases/pharmacologie , Viperidae , Animaux , Endotoxémie/immunologie , Souris , Souris de lignée BALB C
7.
J Biochem Mol Toxicol ; 29(10): 465-471, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-25960139

RÉSUMÉ

In this study, we purified and characterized the procoagulant protein FV-2 from Daboia russelli siamensis (Myanmar) venom using ion-exchange chromatography on CM-Sephadex C-50 and gel filtration on SuperdexTM G-75 column. The activation of factor X and prothrombin was determined, respectively, by specific chromogenic substrates. The fibrinogen-clotting activity, thermal stability, and pH stability were also determined. The N-treminal sequence was determined by the National Center of Biomedical Analysis of China. In the end, FV-2 was achieved with a molecular weight of 13,608.0 Da. It could activate factor X, but did not affect prothrombin or fibrinogen. The suitable pH was 6.5-7.5, and the suitable temperature ranged from 25 to 60°C. The N-terminal sequence was Asn-Phe-Phe-Gln-Phe-Ala-Glu-Met-Ile-Val-Lys-Met-Thr-Gly-Lys. Taken together, our studies suggest that FV-2 is a factor X-activating enzyme, which can activate factor X to factor Xa, but it has no effect on prothrombin and fibrinogen.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE