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











Base de données
Gamme d'année
1.
Gastroenterology ; 159(5): 1882-1897.e5, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32768595

RÉSUMÉ

BACKGROUND & AIMS: Pancreatic ductal adenocarcinomas (PDACs) are hypovascular, resulting in the up-regulation of hypoxia inducible factor 1 alpha (HIF1A), which promotes the survival of cells under low-oxygen conditions. We studied the roles of HIF1A in the development of pancreatic tumors in mice. METHODS: We performed studies with KrasLSL-G12D/+;Trp53LSL-R172H/+;Pdx1-Cre (KPC) mice, KPC mice with labeled pancreatic epithelial cells (EKPC), and EKPC mice with pancreas-specific depletion of HIF1A. Pancreatic and other tissues were collected and analyzed by histology and immunohistochemistry. Cancer cells were cultured from PDACs from mice and analyzed in cell migration and invasion assays and by immunoblots, real-time polymerase chain reaction, and liquid chromatography-mass spectrometry. We performed studies with the human pancreatic cancer cell lines PATU-8988T, BxPC-3, PANC-1, and MiaPACA-2, which have no or low metastatic activity, and PATU-8988S, AsPC-1, SUIT-2 and Capan-1, which have high metastatic activity. Expression of genes was knocked down in primary cancer cells and pancreatic cancer cell lines by using small hairpin RNAs; cells were injected intravenously into immune-competent and NOD/SCID mice, and lung metastases were quantified. We compared levels of messenger RNAs in pancreatic tumors and normal pancreas in The Cancer Genome Atlas. RESULTS: EKPC mice with pancreas-specific deletion of HIF1A developed more advanced pancreatic neoplasias and PDACs with more invasion and metastasis, and had significantly shorter survival times, than EKPC mice. Pancreatic cancer cells from these tumors had higher invasive and metastatic activity in culture than cells from tumors of EKPC mice. HIF1A-knockout pancreatic cancer cells had increased expression of protein phosphatase 1 regulatory inhibitor subunit 1B (PPP1R1B). There was an inverse correlation between levels of HIF1A and PPP1R1B in human PDAC tumors; higher expression of PPP1R1B correlated with shorter survival times of patients. Metastatic human pancreatic cancer cell lines had increased levels of PPP1R1B and lower levels of HIF1A compared with nonmetastatic cancer cell lines; knockdown of PPP1R1B significantly reduced the ability of pancreatic cancer cells to form lung metastases in mice. PPP1R1B promoted degradation of p53 by stabilizing phosphorylation of MDM2 at Ser166. CONCLUSIONS: HIF1A can act a tumor suppressor by preventing the expression of PPP1R1B and subsequent degradation of the p53 protein in pancreatic cancer cells. Loss of HIF1A from pancreatic cancer cells increases their invasive and metastatic activity.


Sujet(s)
Carcinome du canal pancréatique/métabolisme , Mouvement cellulaire , Phosphoprotéine DARPP-32 régulée par la dopamine et l'AMPc/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Tumeurs du poumon/métabolisme , Tumeurs du pancréas/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/secondaire , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Phosphoprotéine DARPP-32 régulée par la dopamine et l'AMPc/génétique , Transition épithélio-mésenchymateuse , Femelle , Régulation de l'expression des gènes tumoraux , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/déficit , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/secondaire , Mâle , Souris de lignée C57BL , Souris transgéniques , Mutation , Invasion tumorale , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Protéolyse , Protéines proto-oncogènes p21(ras)/génétique , Transduction du signal , Transactivateurs/génétique , Transactivateurs/métabolisme , Hypoxie tumorale , Microenvironnement tumoral , Protéine p53 suppresseur de tumeur/génétique , Régulation positive
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE