• Medientyp: E-Artikel
  • Titel: Glycolysis is critical for granulocytic myeloid-derived suppressor cell (G-MDSC) activity during Staphylococcus aureus biofilm infection
  • Beteiligte: Horn, Christopher Michael; Heim, Cortney E.; Kielian, Tammy L
  • Erschienen: The American Association of Immunologists, 2022
  • Erschienen in: The Journal of Immunology
  • Sprache: Englisch
  • DOI: 10.4049/jimmunol.208.supp.51.14
  • ISSN: 0022-1767; 1550-6606
  • Schlagwörter: Immunology ; Immunology and Allergy
  • Entstehung:
  • Anmerkungen:
  • Beschreibung: <jats:title>Abstract</jats:title> <jats:p>Staphylococcus aureus (S. aureus) is a leading cause of prosthetic joint infection (PJI). These infections are often intractable due to biofilm formation, which are complex bacterial communities that adhere to biotic and abiotic surfaces. Within a biofilm, there is considerable heterogeneity in bacterial metabolism and gene expression that serves to create an anti-inflammatory milieu at the site of infection. This is due, in part, to the preferential recruitment of granulocytic myeloid-derived suppressor cells (G-MDSCs), a population of pathologically activated immature leukocytes with immunosuppressive features. Our prior work revealed that G-MDSCs are integral for promoting biofilm persistence via the production of interleukin-10 (IL-10) in response to biofilm-derived lactate. However, the mechanism responsible for G-MDSC anti-inflammatory activity in the context of biofilm infection is unknown. Using a mouse model of S. aureus PJI, time-course single-cell RNA sequencing (scRNA-seq) revealed a significant enrichment in glycolytic and hypoxic transcriptional signatures in infiltrating G-MDSCs. G-MDSCs significantly increased their glycolytic activity when co-cultured with S. aureus biofilm in vitro and inhibiting glycolysis during PJI in vivo with 2-DG nanoparticles significantly attenuated G-MDSC suppressive activity concomitant with a significant reduction in bacterial burden. This study demonstrates that glycolysis is required for G-MDSC-mediated immunosuppression and biofilm persistence during S. aureus PJI. Furthermore, our results demonstrate that G-MDSC metabolism can be targeted to improve infection outcomes.</jats:p> <jats:p>Supported by funding from NIH (2P01 AI083211) &amp; UNMC (UNMC Graduate Fellowship)</jats:p>
  • Zugangsstatus: Freier Zugang