Your browser doesn't support javascript.
loading
Deletion of cystathionine-γ-lyase in bone marrow-derived cells promotes colitis-associated carcinogenesis.
Thanki, Ketan K; Johnson, Paul; Higgins, Edward J; Maskey, Manjit; Phillips, Ches'Nique; Dash, Swetaleena; Almenas, Francisco Arroyo; Govar, Armita Abdollahi; Tian, Bing; Villéger, Romain; Beswick, Ellen; Wang, Rui; Szabo, Csaba; Chao, Celia; Pinchuk, Irina V; Hellmich, Mark R; Módis, Katalin.
Afiliación
  • Thanki KK; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: kkthanki@gmail.com.
  • Johnson P; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: pjohnson08.2012@gmail.com.
  • Higgins EJ; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: Edward.higgins1@gmail.com.
  • Maskey M; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: mamaskey@utmb.edu.
  • Phillips C; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: cphi11@lsuhsc.edu.
  • Dash S; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: swetaleena.dash@outlook.com.
  • Almenas FA; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: farroyo10@yahoo.com.
  • Govar AA; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: armita.lsu@gmail.com.
  • Tian B; Department of Internal Medicine, University of Texas Medical, Galveston, TX, USA. Electronic address: bitian@utmb.edu.
  • Villéger R; Department of Internal Medicine, University of Texas Medical, Galveston, TX, USA. Electronic address: romain.villeger@univ-poitiers.fr.
  • Beswick E; Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA. Electronic address: ellen.beswick@hsc.utah.edu.
  • Wang R; Department of Biology, York University, Toronto, ON, Canada. Electronic address: ruiwang@yorku.ca.
  • Szabo C; Chair of Pharmacology, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland. Electronic address: csaba.szabo@unifr.ch.
  • Chao C; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: cchao986@gmail.com.
  • Pinchuk IV; Department of Internal Medicine, University of Texas Medical, Galveston, TX, USA. Electronic address: ipinchuk@pennstatehealth.psu.edu.
  • Hellmich MR; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: mhellmic@utmb.edu.
  • Módis K; Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA. Electronic address: kamodis@utmb.edu.
Redox Biol ; 55: 102417, 2022 Sep.
Article en En | MEDLINE | ID: mdl-35933902
ABSTRACT
Ulcerative colitis (UC) is characterized by widespread relapsing inflammation of the colonic mucosa. Colitis-associated cancer (CAC) is one of the most serious complications of a prolonged history of UC. Hydrogen sulfide (H2S) has emerged as an important physiological mediator of gastrointestinal homeostasis, limiting mucosal inflammation and promoting tissue healing in response to injury. Inhibition of cystathionine-γ-lyase (CSE)-dependent H2S production in animal models of UC has been shown to exacerbate colitis and delay tissue repair. It is unknown whether CSE plays a role in CAC, or the downregulation of CSE expression and/or activity promotes CAC development. In humans, we observed a significant decrease in CSE expression in colonic biopsies from patients with UC. Using the dextran sodium sulfate (DSS) model of epithelium injury-induced colitis and global CSE KO mouse strain, we demonstrated that CSE is critical in limiting mucosal inflammation and stimulating epithelial cell proliferation in response to injury. In vitro studies showed that CSE activity stimulates epithelial cell proliferation, basal and cytokine-stimulated cell migration, as well as cytokine regulation of transepithelial permeability. In the azoxymethane (AOM)/DSS model of CAC, the loss of CSE expression accelerated both the development and progression of CAC. The increased tumor multiplicity and severity of CAC observed in CSE-KO mice were associated with reduced levels of mucosal IL-10 expression and increased levels of IL-6. Restoring CSE expression in bone marrow (BM) cells of CSE-KO mice through reciprocal BM transplantation raised mucosal IL-10 expression, decreased IL-6 level, and reduced the number of aberrant crypt foci and tumors in AOM/DSS-treated mice. These studies demonstrate that CSE expression in BM cells plays a critical role in suppressing CAC in mice. Furthermore, the data suggest that the inhibitory effects of CSE on the development of CAC are due, in part, to the modulation of mucosal pro-and anti-inflammatory cytokine expression.
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Tipo de estudio: Prognostic_studies / Risk_factors_studies Idioma: En Revista: Redox Biol Año: 2022 Tipo del documento: Article

Texto completo: 1 Bases de datos: MEDLINE Tipo de estudio: Prognostic_studies / Risk_factors_studies Idioma: En Revista: Redox Biol Año: 2022 Tipo del documento: Article