Abstract

Candida albicans is among the most prevalent opportunistic human fungal pathogens. The ability to mask the immunogenic polysaccharide β (1,3)-glucan from immune detection via a layer of mannosylated proteins is a key virulence factor of C. albicans We previously reported that hyperactivation of the Cek1 mitogen-activated protein (MAP) kinase pathway promotes β (1,3)-glucan exposure. In this communication, we report a novel upstream regulator of Cek1 activation and characterize the impact of Cek1 activity on fungal virulence. Lrg1 encodes a GTPase-activating protein (GAP) that has been suggested to inhibit the GTPase Rho1. We found that disruption of LRG1 causes Cek1 hyperactivation and β (1,3)-glucan unmasking. However, when GTPase activation was measured for a panel of GTPases, the lrg1ΔΔ mutant exhibited increased activation of Cdc42 and Ras1 but not Rho1 or Rac1. Unmasking and Cek1 activation in the lrg1ΔΔ mutant can be blocked by inhibition of the Ste11 MAP kinase kinase kinase (MAPKKK), indicating that the lrg1ΔΔ mutant acts through the canonical Cek1 MAP kinase cascade. In order to determine how Cek1 hyperactivation specifically impacts virulence, a doxycycline-repressible hyperactive STE11ΔN467 allele was expressed in C. albicans In the absence of doxycycline, this allele overexpressed STE11ΔN467 , which induced production of proinflammatory tumor necrosis factor alpha (TNF-α) from murine macrophages. This in vitro phenotype correlates with decreased colonization and virulence in a mouse model of systemic infection. The mechanism by which Ste11ΔN467 causes unmasking was explored with RNA sequencing (RNA-Seq) analysis. Overexpression of Ste11ΔN467 caused upregulation of the Cph1 transcription factor and of a group of cell wall-modifying proteins which are predicted to impact cell wall architecture.IMPORTANCECandida albicans is an important source of systemic infections in humans. The ability to mask the immunogenic cell wall polymer β (1,3)-glucan from host immune surveillance contributes to fungal virulence. We previously reported that the hyperactivation of the Cek1 MAP kinase cascade promotes cell wall unmasking, thus increasing strain immunogenicity. In this study, we identified a novel regulator of the Cek1 pathway called Lrg1. Lrg1 is a predicted GTPase-activating protein (GAP) that represses Cek1 activity by downregulating the GTPase Cdc42 and its downstream MAPKKK, Ste11. Upregulation of Cek1 activity diminished fungal virulence in the mouse model of infection, and this correlates with increased cytokine responses from macrophages. We also analyzed the transcriptional profile determined during β (1,3)-glucan exposure driven by Cek1 hyperactivation. Our report provides a model where Cek1 hyperactivation causes β (1,3)-glucan exposure by upregulation of cell wall proteins and leads to more robust immune detection in vivo, promoting more effective clearance.

Highlights

  • Candida albicans is among the most prevalent opportunistic human fungal pathogens

  • We previously showed that hyperactive Rho[1] (Rho1Q67L) exposed ␤ (1,3)glucan in the cell wall but appeared to act through the Cek[1] mitogenactivated protein kinase (MAPK) rather than Mkc[1] (17)

  • Since Lrg[1] has been described as the Rho[1] GTPaseactivating protein (GAP) in C. albicans, we hypothesized that disruption of LRG1 would cause ␤ (1,3)-glucan exposure

Read more

Summary

Introduction

Candida albicans is among the most prevalent opportunistic human fungal pathogens. The ability to mask the immunogenic polysaccharide ␤ (1,3)glucan from immune detection via a layer of mannosylated proteins is a key virulence factor of C. albicans. Our report provides a model where Cek[1] hyperactivation causes ␤ (1,3)-glucan exposure by upregulation of cell wall proteins and leads to more robust immune detection in vivo, promoting more effective clearance. Treatment with the cell wall inhibitor caspofungin, or damage by neutrophils can expose C. albicans ␤ (1,3)-glucan (9–11) This exposure facilitates recognition by host immune cells through receptors such as Dectin-1 and launches immune responses more efficiently and rapidly, including induction of proinflammatory cytokines such as tumor necrosis factor alpha (TNF-␣), which promotes fungal clearance (12). In C. albicans, Lrg[1] is proposed to act as the Rho[1] GAP based on the evidence that the lrg1⌬⌬ mutant and hyperactivated RHO1Q67L mutant each induce hyphal formation, and a mutation of the PKC1 gene, which acts downstream of Rho[1], blocks lrg1ΔΔ hyperfilamentation. These data suggest that Lrg[1] could negatively regulate Rho[1] activity (24, 25), but the GAP activity of Lrg[1] for Rho[1] has not been measured in C. albicans (24)

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call