Your browser doesn't support javascript.
loading
Candida albicans translocation through the intestinal epithelial barrier is promoted by fungal zinc acquisition and limited by NFκB-mediated barrier protection.
Sprague, Jakob L; Schille, Tim B; Allert, Stefanie; Trümper, Verena; Lier, Adrian; Großmann, Peter; Priest, Emily L; Tsavou, Antzela; Panagiotou, Gianni; Naglik, Julian R; Wilson, Duncan; Schäuble, Sascha; Kasper, Lydia; Hube, Bernhard.
Afiliación
  • Sprague JL; Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany.
  • Schille TB; Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany.
  • Allert S; Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany.
  • Trümper V; Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany.
  • Lier A; Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany.
  • Großmann P; Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany.
  • Priest EL; Department of Microbiome Dynamics, Hans-Knöll-Institute, Jena, Germany.
  • Tsavou A; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom.
  • Panagiotou G; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom.
  • Naglik JR; Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany.
  • Wilson D; Department of Microbiome Dynamics, Hans-Knöll-Institute, Jena, Germany.
  • Schäuble S; Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany.
  • Kasper L; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom.
  • Hube B; Medical Research Council, Centre for Medical Mycology at the University of Exeter, Exeter, United Kingdom.
PLoS Pathog ; 20(3): e1012031, 2024 Mar.
Article en En | MEDLINE | ID: mdl-38427950
ABSTRACT
The opportunistic fungal pathogen Candida albicans thrives on human mucosal surfaces as a harmless commensal, but frequently causes infections under certain predisposing conditions. Translocation across the intestinal barrier into the bloodstream by intestine-colonizing C. albicans cells serves as the main source of disseminated candidiasis. However, the host and microbial mechanisms behind this process remain unclear. In this study we identified fungal and host factors specifically involved in infection of intestinal epithelial cells (IECs) using dual-RNA sequencing. Our data suggest that host-cell damage mediated by the peptide toxin candidalysin-encoding gene ECE1 facilitates fungal zinc acquisition. This in turn is crucial for the full virulence potential of C. albicans during infection. IECs in turn exhibit a filamentation- and damage-specific response to C. albicans infection, including NFκB, MAPK, and TNF signaling. NFκB activation by IECs limits candidalysin-mediated host-cell damage and mediates maintenance of the intestinal barrier and cell-cell junctions to further restrict fungal translocation. This is the first study to show that candidalysin-mediated damage is necessary for C. albicans nutrient acquisition during infection and to explain how IECs counteract damage and limit fungal translocation via NFκB-mediated maintenance of the intestinal barrier.
Asunto(s)

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Candida albicans / Candidiasis Límite: Humans Idioma: En Revista: PLoS Pathog Año: 2024 Tipo del documento: Article País de afiliación: Alemania

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Candida albicans / Candidiasis Límite: Humans Idioma: En Revista: PLoS Pathog Año: 2024 Tipo del documento: Article País de afiliación: Alemania