Your browser doesn't support javascript.
loading
A Nucleotide Analog Prevents Colitis-Associated Cancer via Beta-Catenin Independently of Inflammation and Autophagy.
Sheng, Yong Hua; Giri, Rabina; Davies, Julie; Schreiber, Veronika; Alabbas, Saleh; Movva, Ramya; He, Yaowu; Wu, Andy; Hooper, John; McWhinney, Brett; Oancea, Iulia; Kijanka, Gregor; Hasnain, Sumaira; Lucke, Andrew J; Fairlie, David P; McGuckin, Michael A; Florin, Timothy H; Begun, Jakob.
Afiliação
  • Sheng YH; Inflammatory Bowel Diseases Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland; Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongab
  • Giri R; Inflammatory Bowel Diseases Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland.
  • Davies J; Inflammatory Bowel Diseases Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland.
  • Schreiber V; Inflammatory Bowel Diseases Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland.
  • Alabbas S; Inflammatory Bowel Diseases Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland.
  • Movva R; Inflammatory Bowel Diseases Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland.
  • He Y; Cancer Biology Group, Mater Research Institute-University of Queensland, Woolloongabba, Queensland.
  • Wu A; Bones and Immunology Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland.
  • Hooper J; Cancer Biology Group, Mater Research Institute-University of Queensland, Woolloongabba, Queensland.
  • McWhinney B; Queensland Pathology Services, Herston.
  • Oancea I; School of Clinical Medicine, Faculty of Medicine, University of Queensland.
  • Kijanka G; Immune Profiling & Cancer Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland.
  • Hasnain S; Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland.
  • Lucke AJ; ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
  • Fairlie DP; ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
  • McGuckin MA; Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria.
  • Florin TH; Inflammatory Bowel Diseases Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland. Electronic address: t.florin@uq.edu.au.
  • Begun J; Inflammatory Bowel Diseases Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland; Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongab
Cell Mol Gastroenterol Hepatol ; 11(1): 33-53, 2021.
Article em En | MEDLINE | ID: mdl-32497793
ABSTRACT
BACKGROUND &

AIMS:

Chronic bowel inflammation increases the risk of colon cancer; colitis-associated cancer (CAC). Thiopurine treatments are associated with a reduction in dysplasia and CAC in inflammatory bowel disease (IBD). Abnormal Wnt/ß-catenin signalling is characteristic of >90% of colorectal cancers. Immunosuppression by thiopurines is via Rac1 GTPase, which also affects Wnt/ß-catenin signalling. Autophagy is implicated in colonic tumors, and topical delivery of the thiopurine thioguanine (TG) is known to alleviate colitis and augment autophagy. This study investigated the effects of TG in a murine model of CAC and potential mechanisms.

METHODS:

Colonic dysplasia was induced by exposure to azoxymethane (AOM) and dextran sodium sulfate (DSS) in wild-type (WT) mice and mice harboring intestinal epithelial cell-specific deletion of autophagy related 7 gene (Atg7ΔIEC). TG or vehicle was administered intrarectally, and the effect on tumor burden and ß-catenin activity was assessed. The mechanisms of action of TG were investigated in vitro and in vivo.

RESULTS:

TG ameliorated DSS colitis in wild-type but not Atg7ΔIEC mice, demonstrating that anti-inflammatory effects of locally delivered TG are autophagy-dependent. However, TG inhibited CAC in both wild-type and Atg7ΔIEC mice. This was associated with decreased ß-catenin activation/nuclear translocation demonstrating that TG's inhibition of tumorigenesis occurred independently of anti-inflammatory and pro-autophagic actions. These results were confirmed in cell lines, and the dependency on Rac1 GTPase was demonstrated by siRNA knockdown and overexpression of constitutively active Rac1.

CONCLUSIONS:

Our findings provide evidence for a new mechanism that could be exploited to improve CAC chemoprophylactic approaches.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Tioguanina / Colite / Via de Sinalização Wnt / Neoplasias Associadas a Colite Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Tioguanina / Colite / Via de Sinalização Wnt / Neoplasias Associadas a Colite Idioma: En Ano de publicação: 2021 Tipo de documento: Article