Core N-glycan structures are critical for the pathogenicity of Cryptococcus neoformans by modulating host cell death

Cited 17 time in scopus
Metadata Downloads
Title
Core N-glycan structures are critical for the pathogenicity of Cryptococcus neoformans by modulating host cell death
Author(s)
E J Thak; S B Lee; S Xu-Vanpala; D J Lee; S Y Chung; Y S Bahn; Doo-Byoung Oh; M L Shinohara; H A Kang
Bibliographic Citation
Mbio, vol. 11, no. 3, pp. e00711-e00711
Publication Year
2020
Abstract
Cryptococcus neoformans is a human-pathogenic fungal pathogen that causes life-threatening meningoencephalitis in immunocompromised individuals. To investigate the roles of N-glycan core structure in cryptococcal pathogenicity, we constructed mutant strains of C. neoformans with defects in the assembly of lipid-linked N-glycans in the luminal side of the endoplasmic reticulum (ER). Deletion of ALG3 (alg3Δ), which encodes dolichyl-phosphate-mannose (Dol-P-Man)-dependent ?-1,3-mannosyltransferase, resulted in the production of truncated neutral N-glycans carrying five mannose residues as a major species. Despite moderate or nondetect-able defects in virulence-associated phenotypes in vitro, the alg3Δ mutant was aviru-lent in a mouse model of systemic cryptococcosis. Notably, the mutant did not show defects in early stages of host cell interaction during infection, including attachment to lung epithelial cells, opsonic/nonopsonic phagocytosis, and manipulation of phagosome acidification. However, the ability to drive macrophage cell death was greatly decreased in this mutant, without loss of cell wall remodeling capacity. Fur-thermore, deletion of ALG9 and ALG12, encoding Dol-P-Man-dependent ?-1,2-mannosyltransferases and ?-1,6-mannosyltransferases, generating truncated core N-glycans with six and seven mannose residues, respectively, also displayed remark-ably reduced macrophage cell death and in vivo virulence. However, secretion levels of interleukin-1ß (IL-1ß) were not reduced in the bone marrow-derived dendritic cells obtained from Asc-and Gsdmd-deficient mice infected with the alg3Δ mutant strain, excluding the possibility that pyroptosis is a main host cell death pathway dependent on intact core N-glycans. Our results demonstrated N-glycan structures as a critical feature in modulating death of host cells, which is exploited by as a strategy for host cell escape for dissemination of C. neoformans. IMPORTANCE We previously reported that the outer mannose chains of N-glycans are dispensable for the virulence of C. neoformans, which is in stark contrast to findings for the other human-pathogenic yeast, Candida albicans. Here, we present evi-dence that an intact core N-glycan structure is required for C. neoformans pathoge-nicity by systematically analyzing alg3?, alg9?, and alg12? strains that have defects in lipid-linked N-glycan assembly and in in vivo virulence. The alg null mutants pro-ducing truncated core N-glycans were defective in inducing host cell death after phagocytosis, which is triggered as a mechanism of pulmonary escape and dissemination of C. neoformans, thus becoming inactive in causing fatal infection. The results clearly demonstrated the critical features of the N-glycan structure in mediating the interaction with host cells during fungal infection. The delineation of the roles of protein glycosylation in fungal pathogenesis not only provides insight into the glycan-based fungal infection mechanism but also will aid in the development of novel antifungal agents.
Keyword
ALGCryptococcus neoformansFungal pathogenesisN-linked protein glycosylation
ISSN
2150-7511
Publisher
Amer Soc Microb
DOI
http://dx.doi.org/10.1128/mBio.00711-20
Type
Article
Appears in Collections:
Aging Convergence Research Center > 1. Journal Articles
Files in This Item:
  • There are no files associated with this item.


Items in OpenAccess@KRIBB are protected by copyright, with all rights reserved, unless otherwise indicated.