Login / Signup

β-glucan-dependent shuttling of conidia from neutrophils to macrophages occurs during fungal infection establishment.

Vahid PazhakhFelix EllettBen A CrokerJoanne A O'DonnellLuke PaseKeith E SchulzeR Stefan GreulichAakash GuptaConstantino Carlos Reyes-AldasoroAlex AndrianopoulosGraham J Lieschke
Published in: PLoS biology (2019)
The initial host response to fungal pathogen invasion is critical to infection establishment and outcome. However, the diversity of leukocyte-pathogen interactions is only recently being appreciated. We describe a new form of interleukocyte conidial exchange called "shuttling." In Talaromyces marneffei and Aspergillus fumigatus zebrafish in vivo infections, live imaging demonstrated conidia initially phagocytosed by neutrophils were transferred to macrophages. Shuttling is unidirectional, not a chance event, and involves alterations of phagocyte mobility, intercellular tethering, and phagosome transfer. Shuttling kinetics were fungal-species-specific, implicating a fungal determinant. β-glucan serves as a fungal-derived signal sufficient for shuttling. Murine phagocytes also shuttled in vitro. The impact of shuttling for microbiological outcomes of in vivo infections is difficult to specifically assess experimentally, but for these two pathogens, shuttling augments initial conidial redistribution away from fungicidal neutrophils into the favorable macrophage intracellular niche. Shuttling is a frequent host-pathogen interaction contributing to fungal infection establishment patterns.
Keyphrases
  • cell wall
  • candida albicans
  • high resolution
  • adipose tissue
  • antimicrobial resistance
  • multidrug resistant
  • reactive oxygen species