Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Med ; 22(1): 42, 2024 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-38281914

RESUMEN

BACKGROUND: Microsatellite instability-high (MSI-H) is a unique genomic status in many cancers. However, its role in the genomic features and immunotherapy in cholangiocarcinoma (CCA) is unclear. This study aimed to systematically investigate the genomic characterization and immunotherapy efficacy of MSI-H patients with CCA. METHODS: We enrolled 887 patients with CCA in this study. Tumor samples were collected for next-generation sequencing. Differences in genomic alterations between the MSI-H and microsatellite stability (MSS) groups were analyzed. We also investigated the survival of PD-1 inhibitor-based immunotherapy between two groups of 139 patients with advanced CCA. RESULTS: Differential genetic alterations between the MSI-H and MSS groups included mutations in ARID1A, ACVR2A, TGFBR2, KMT2D, RNF43, and PBRM1 which were enriched in MSI-H groups. Patients with an MSI-H status have a significantly higher tumor mutation burden (TMB) (median 41.7 vs. 3.1 muts/Mb, P < 0.001) and more positive programmed death ligand 1 (PD-L1) expression (37.5% vs. 11.9%, P < 0.001) than those with an MSS status. Among patients receiving PD-1 inhibitor-based therapy, those with MSI-H had a longer median overall survival (OS, hazard ratio (HR) = 0.17, P = 0.001) and progression-free survival (PFS, HR = 0.14, P < 0.001) than patients with MSS. Integrating MSI-H and PD-L1 expression status (combined positive score ≥ 5) could distinguish the efficacy of immunotherapy. CONCLUSIONS: MSI-H status was associated with a higher TMB value and more positive PD-L1 expression in CCA tumors. Moreover, in patients with advanced CCA who received PD-1 inhibitor-based immunotherapy, MSI-H and positive PD-L1 expression were associated with improved both OS and PFS. TRIAL REGISTRATION: This study was registered on ClinicalTrials.gov on 07/01/2017 (NCT03892577).


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Humanos , Inestabilidad de Microsatélites , Antígeno B7-H1/genética , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Colangiocarcinoma/genética , Colangiocarcinoma/terapia , Mutación , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/terapia , Conductos Biliares Intrahepáticos/metabolismo , Inmunoterapia , Genómica , Biomarcadores de Tumor/genética
2.
Cell Biol Toxicol ; 34(4): 263-277, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29063978

RESUMEN

The Hippo-Mst1 pathway is associated with tumor development and progression. However, little evidence is available for its role in colorectal cancer (CRC) stress response via mitochondrial homeostasis. In this study, we conducted gain-of function assay about Mst1 in CRC via adenovirus transfection. Then, cellular viability and apoptosis were measured via MTT, TUNEL assay, and typan blue staining. Mitochondrial function was detected via JC1 staining, mPTP opening assay, and immunofluorescence of cyt-c. Mitophagy was observed via western blots and immunofluorescence. Cell migration and proliferation were evaluated via Transwell and BrdU assay. Western blots were used to analyze the signaling pathways with JNK inhibitors or p53 siRNA. We found that Mst1 was down-regulated in CRC. Overexpression of Mst1 induced CRC apoptosis and impaired cell proliferation and migration. Functional studies have illustrated that recovery of Mst1 could activate JNK pathway which upregulated the p53 expression. The latter repressed Bnip3 transcription and activity, leading to the mitophagy arrest. The defective mitophagy impaired mitochondrial homeostasis, evoked cellular oxidative stress, and initiated the mitochondrial apoptosis. Meanwhile, bad-structured mitophagy also hindered the cancer proliferation via CyclinD/E. Moreover, Mst1-suppressed mitophagy was associated with CRC migration inhibition via regulation of CXCR4/7 expression. Collectively, our data described the comprehensive role of Mst1 in colorectal cancer stress response involving apoptosis, mobilization, and growth via handling mitophagy by JNK/p53/Bnip3 pathways.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Mitofagia , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Regulación hacia Abajo , Homeostasis , Humanos , Péptidos y Proteínas de Señalización Intracelular , Mitocondrias/metabolismo , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...