Your browser doesn't support javascript.
loading
Autophagy Inhibition Dysregulates TBK1 Signaling and Promotes Pancreatic Inflammation.
Yang, Shenghong; Imamura, Yu; Jenkins, Russell W; Cañadas, Israel; Kitajima, Shunsuke; Aref, Amir; Brannon, Arthur; Oki, Eiji; Castoreno, Adam; Zhu, Zehua; Thai, Tran; Reibel, Jacob; Qian, Zhirong; Ogino, Shuji; Wong, Kwok K; Baba, Hideo; Kimmelman, Alec C; Pasca Di Magliano, Marina; Barbie, David A.
Afiliação
  • Yang S; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Imamura Y; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan. Department of Gastroenterological Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan. Department of Surgery and Science, Graduate of Medi
  • Jenkins RW; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Cañadas I; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Kitajima S; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Aref A; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Brannon A; Department of Surgery, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan. Cell and Developmental Biology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan.
  • Oki E; Department of Surgery and Science, Graduate of Medical Sciences, Kyushu University, Fukuoka, Japan.
  • Castoreno A; Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Zhu Z; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Thai T; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Reibel J; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Qian Z; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts.
  • Ogino S; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts.
  • Wong KK; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Baba H; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
  • Kimmelman AC; Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Pasca Di Magliano M; Department of Surgery, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan. Cell and Developmental Biology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan.
  • Barbie DA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts. dbarbie@partners.org.
Cancer Immunol Res ; 4(6): 520-30, 2016 06.
Article em En | MEDLINE | ID: mdl-27068336
ABSTRACT
Autophagy promotes tumor progression downstream of oncogenic KRAS, yet also restrains inflammation and dysplasia through mechanisms that remain incompletely characterized. Understanding the basis of this paradox has important implications for the optimal targeting of autophagy in cancer. Using a mouse model of cerulein-induced pancreatitis, we found that loss of autophagy by deletion of Atg5 enhanced activation of the IκB kinase (IKK)-related kinase TBK1 in vivo, associated with increased neutrophil and T-cell infiltration and PD-L1 upregulation. Consistent with this observation, pharmacologic or genetic inhibition of autophagy in pancreatic ductal adenocarcinoma cells, including suppression of the autophagy receptors NDP52 or p62, prolonged TBK1 activation and increased expression of CCL5, IL6, and several other T-cell and neutrophil chemotactic cytokines in vitro Defective autophagy also promoted PD-L1 upregulation, which is particularly pronounced downstream of IFNγ signaling and involves JAK pathway activation. Treatment with the TBK1/IKKε/JAK inhibitor CYT387 (also known as momelotinib) not only inhibits autophagy, but also suppresses this feedback inflammation and reduces PD-L1 expression, limiting KRAS-driven pancreatic dysplasia. These findings could contribute to the dual role of autophagy in oncogenesis and have important consequences for its therapeutic targeting. Cancer Immunol Res; 4(6); 520-30. ©2016 AACR.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Pancreatite / Autofagia / Proteínas Serina-Treonina Quinases Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2016 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Pancreatite / Autofagia / Proteínas Serina-Treonina Quinases Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2016 Tipo de documento: Article