Your browser doesn't support javascript.
loading
Antithetical NFATc1-Sox2 and p53-miR200 signaling networks govern pancreatic cancer cell plasticity.
Singh, Shiv K; Chen, Nai-Ming; Hessmann, Elisabeth; Siveke, Jens; Lahmann, Marlen; Singh, Garima; Voelker, Nadine; Vogt, Sophia; Esposito, Irene; Schmidt, Ansgar; Brendel, Cornelia; Stiewe, Thorsten; Gaedcke, Jochen; Mernberger, Marco; Crawford, Howard C; Bamlet, William R; Zhang, Jin-San; Li, Xiao-Kun; Smyrk, Thomas C; Billadeau, Daniel D; Hebrok, Matthias; Neesse, Albrecht; Koenig, Alexander; Ellenrieder, Volker.
Afiliação
  • Singh SK; Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg, Germany.
  • Chen NM; Department of Gastroenterology II, University Medical Center Goettingen, Goettingen, Germany.
  • Hessmann E; Department of Gastroenterology II, University Medical Center Goettingen, Goettingen, Germany.
  • Siveke J; II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität, Munich, Germany.
  • Lahmann M; Institute for Molecular Tumor Biology, Philipps University, Marburg, Germany.
  • Singh G; Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg, Germany.
  • Voelker N; Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg, Germany.
  • Vogt S; Signaling and Transcription Laboratory, Department of Gastroenterology, Philipps University, Marburg, Germany.
  • Esposito I; Institute of Pathology, Helmholtz Zentrum, Munich, Germany.
  • Schmidt A; Institute of Pathology, Philipps University, Marburg, Germany.
  • Brendel C; Department of Hematology and Oncology, Philipps University, Marburg, Germany.
  • Stiewe T; Institute for Molecular Tumor Biology, Philipps University, Marburg, Germany.
  • Gaedcke J; Department of Surgery, University Medical Center Goettingen, Goettingen, Germany.
  • Mernberger M; Institute for Molecular Tumor Biology, Philipps University, Marburg, Germany.
  • Crawford HC; Department of Cancer Biology, Mayo Clinic Florida, Jacksonville, FL, USA.
  • Bamlet WR; Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, MN, USA.
  • Zhang JS; Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA School of Pharmaceutical Sciences and Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Li XK; School of Pharmaceutical Sciences and Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Smyrk TC; Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA.
  • Billadeau DD; Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA.
  • Hebrok M; Diabetes Center, USCF, San Francisco, CA, USA.
  • Neesse A; Department of Gastroenterology II, University Medical Center Goettingen, Goettingen, Germany.
  • Koenig A; Department of Gastroenterology II, University Medical Center Goettingen, Goettingen, Germany Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA.
  • Ellenrieder V; Department of Gastroenterology II, University Medical Center Goettingen, Goettingen, Germany volker.ellenrieder@med.uni-goettingen.de.
EMBO J ; 34(4): 517-30, 2015 Feb 12.
Article em En | MEDLINE | ID: mdl-25586376
ABSTRACT
In adaptation to oncogenic signals, pancreatic ductal adenocarcinoma (PDAC) cells undergo epithelial-mesenchymal transition (EMT), a process combining tumor cell dedifferentiation with acquisition of stemness features. However, the mechanisms linking oncogene-induced signaling pathways with EMT and stemness remain largely elusive. Here, we uncover the inflammation-induced transcription factor NFATc1 as a central regulator of pancreatic cancer cell plasticity. In particular, we show that NFATc1 drives EMT reprogramming and maintains pancreatic cancer cells in a stem cell-like state through Sox2-dependent transcription of EMT and stemness factors. Intriguingly, NFATc1-Sox2 complex-mediated PDAC dedifferentiation and progression is opposed by antithetical p53-miR200c signaling, and inactivation of the tumor suppressor pathway is essential for tumor dedifferentiation and dissemination both in genetically engineered mouse models (GEMM) and human PDAC. Based on these findings, we propose the existence of a hierarchical signaling network regulating PDAC cell plasticity and suggest that the molecular decision between epithelial cell preservation and conversion into a dedifferentiated cancer stem cell-like phenotype depends on opposing levels of p53 and NFATc1 signaling activities.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Pancreáticas / Proteína Supressora de Tumor p53 / MicroRNAs / Fatores de Transcrição NFATC / Fatores de Transcrição SOXB1 Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: EMBO J Ano de publicação: 2015 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Pancreáticas / Proteína Supressora de Tumor p53 / MicroRNAs / Fatores de Transcrição NFATC / Fatores de Transcrição SOXB1 Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: EMBO J Ano de publicação: 2015 Tipo de documento: Article