Your browser doesn't support javascript.
loading
Epithelial X-Box Binding Protein 1 Coordinates Tumor Protein p53-Driven DNA Damage Responses and Suppression of Intestinal Carcinogenesis.
Welz, Lina; Kakavand, Nassim; Hang, Xiang; Laue, Georg; Ito, Go; Silva, Miguel Gomes; Plattner, Christina; Mishra, Neha; Tengen, Felicitas; Ogris, Christoph; Jesinghaus, Moritz; Wottawa, Felix; Arnold, Philipp; Kaikkonen, Leena; Stengel, Stefanie; Tran, Florian; Das, Saumya; Kaser, Arthur; Trajanoski, Zlatko; Blumberg, Richard; Roecken, Christoph; Saur, Dieter; Tschurtschenthaler, Markus; Schreiber, Stefan; Rosenstiel, Philip; Aden, Konrad.
Afiliação
  • Welz L; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Kakavand N; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Hang X; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Laue G; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Ito G; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan.
  • Silva MG; Center for Translational Cancer Research (TranslaTUM), Technische Universität München, Munich, Germany.
  • Plattner C; Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria.
  • Mishra N; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Tengen F; Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
  • Ogris C; Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
  • Jesinghaus M; Institute of Pathology, University Hospital Marburg, Marburg, Germany.
  • Wottawa F; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Arnold P; Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany.
  • Kaikkonen L; Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts.
  • Stengel S; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Tran F; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Das S; Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts.
  • Kaser A; Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom.
  • Trajanoski Z; Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria.
  • Blumberg R; Gastroenterology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
  • Roecken C; Department of Pathology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Saur D; Center for Translational Cancer Research (TranslaTUM), Technische Universität München, Munich, Germany.
  • Tschurtschenthaler M; Center for Translational Cancer Research (TranslaTUM), Technische Universität München, Munich, Germany.
  • Schreiber S; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
  • Rosenstiel P; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany. Electronic address: p.rosenstiel@mucosa.de.
  • Aden K; Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany. Electronic address
Gastroenterology ; 162(1): 223-237.e11, 2022 01.
Article em En | MEDLINE | ID: mdl-34599932
ABSTRACT
BACKGROUND &

AIMS:

Throughout life, the intestinal epithelium undergoes constant self-renewal from intestinal stem cells. Together with genotoxic stressors and failing DNA repair, this self-renewal causes susceptibility toward malignant transformation. X-box binding protein 1 (XBP1) is a stress sensor involved in the unfolded protein response (UPR). We hypothesized that XBP1 acts as a signaling hub to regulate epithelial DNA damage responses.

METHODS:

Data from The Cancer Genome Atlas were analyzed for association of XBP1 with colorectal cancer (CRC) survival and molecular interactions between XBP1 and p53 pathway activity. The role of XBP1 in orchestrating p53-driven DNA damage response was tested in vitro in mouse models of chronic intestinal epithelial cell (IEC) DNA damage (Xbp1/H2bfl/fl, Xbp1ΔIEC, H2bΔIEC, H2b/Xbp1ΔIEC) and via orthotopic tumor organoid transplantation. Transcriptome analysis of intestinal organoids was performed to identify molecular targets of Xbp1-mediated DNA damage response.

RESULTS:

In The Cancer Genome Atlas data set of CRC, low XBP1 expression was significantly associated with poor overall survival and reduced p53 pathway activity. In vivo, H2b/Xbp1ΔIEC mice developed spontaneous intestinal carcinomas. Orthotopic tumor organoid transplantation revealed a metastatic potential of H2b/Xbp1ΔIEC-derived tumors. RNA sequencing of intestinal organoids (H2b/Xbp1fl/fl, H2bΔIEC, H2b/Xbp1ΔIEC, and H2b/p53ΔIEC) identified a transcriptional program downstream of p53, in which XBP1 directs DNA-damage-inducible transcript 4-like (Ddit4l) expression. DDIT4L inhibits mechanistic target of rapamycin-mediated phosphorylation of 4E-binding protein 1. Pharmacologic mechanistic target of rapamycin inhibition suppressed epithelial hyperproliferation via 4E-binding protein 1.

CONCLUSIONS:

Our data suggest a crucial role for XBP1 in coordinating epithelial DNA damage responses and stem cell function via a p53-DDIT4L-dependent feedback mechanism.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Dano ao DNA / Adenocarcinoma / Adenoma / Transformação Celular Neoplásica / Células Epiteliais / Proteína 1 de Ligação a X-Box / Mucosa Intestinal / Neoplasias Intestinais Idioma: En Revista: Gastroenterology Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Alemanha

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Dano ao DNA / Adenocarcinoma / Adenoma / Transformação Celular Neoplásica / Células Epiteliais / Proteína 1 de Ligação a X-Box / Mucosa Intestinal / Neoplasias Intestinais Idioma: En Revista: Gastroenterology Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Alemanha