Your browser doesn't support javascript.
loading
Extracellular vesicles from gastric epithelial GES-1 cells infected with Helicobacter pylori promote changes in recipient cells associated with malignancy.
González, María Fernanda; Burgos-Ravanal, Renato; Shao, Baohai; Heinecke, Jay; Valenzuela-Valderrama, Manuel; Corvalán, Alejandro H; Quest, Andrew F G.
Afiliación
  • González MF; Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
  • Burgos-Ravanal R; Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, Chile.
  • Shao B; Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
  • Heinecke J; Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, Chile.
  • Valenzuela-Valderrama M; Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, United States.
  • Corvalán AH; Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, United States.
  • Quest AFG; Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, Chile.
Front Oncol ; 12: 962920, 2022.
Article en En | MEDLINE | ID: mdl-36313672
ABSTRACT
Chronic Helicobacter pylori (H. pylori) infection is considered the main risk factor for the development of gastric cancer. Pathophysiological changes in the gastric mucosa initiated by this bacterium can persist even after pharmacological eradication and are likely attributable also to changes induced in non-infected cells as a consequence of intercellular communication via extracellular vesicles (EVs). To better understand what such changes might entail, we isolated EVs from immortalized normal gastric GES-1 cells infected (EVHp+) or not with H. pylori (EVHp-) by ultracentrifugation and characterized them. Infection of GES-1 cells with H. pylori significantly increased the release of EVs and slightly decreased the EV mean size. Incubation with EVHp+ for 24 h decreased the viability of GES-1 cells, but increased the levels of IL-23 in GES-1 cells, as well as the migration of GES-1 and gastric cancer AGS cells. Furthermore, incubation of GES-1 and AGS cells with EVHp+, but not with EVHp-, promoted cell invasion and trans-endothelial migration in vitro. Moreover, stimulation of endothelial EA.hy926 cells for 16 h with EVHp+ promoted the formation of linked networks. Finally, analysis by mass spectrometry identified proteins uniquely present and others enriched in EVHp+ compared to EVHp-, several of which are known targets of hypoxia induced factor-1α (HIF-1α) that may promote the acquisition of traits important for the genesis/progression of gastric pre-neoplastic changes associated with H. pylori infection. In conclusion, the harmful effects of H. pylori infection associated with the development of gastric malignancies may spread via EVs to non-infected areas in the early and later stages of gastric carcinogenesis.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Prognostic_studies / Risk_factors_studies Idioma: En Revista: Front Oncol Año: 2022 Tipo del documento: Article País de afiliación: Chile

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Prognostic_studies / Risk_factors_studies Idioma: En Revista: Front Oncol Año: 2022 Tipo del documento: Article País de afiliación: Chile