Abstract

The invasive ability of the blood-borne fungal pathogen Cryptococcus neoformans can be enhanced through interactions with host plasma components, such as plasminogen. Previously we showed by in vitro studies that plasminogen coats the surface of C. neoformans and is converted to the active serine protease, plasmin, by host plasminogen activators. Viable, but not formaldehyde- or sodium azide-killed, cryptococcal strains undergo brain microvascular endothelial cell-dependent plasminogen-to-plasmin activation, which results in enhanced, plasmin-dependent cryptococcal invasion of primary bovine brain microvascular endothelial cells and fungal ability to degrade plasmin substrates. In the present work, brain microvascular endothelial cells cultured with viable, but not killed, cryptococcal strains led to significant increases in both urokinase mRNA transcription and cell-associated urokinase protein expression. Soluble urokinase was also detected in conditioned medium from brain microvascular endothelial cells cultured with viable, but not killed, C. neoformans. Exposure of plasminogen pre-coated viable C. neoformans to conditioned medium from strain-matched brain microvascular endothelial cell-fungal co-cultures resulted in plasminogen-to-plasmin activation and plasmin-dependent cryptococcal invasion. siRNA-mediated silencing of urokinase gene expression or the use of specific inhibitors of urokinase activity abrogated both plasminogen-to-plasmin activation on C. neoformans and cryptococcal-brain microvascular endothelial cell invasion. Our results suggest that pathogen exploitation of the host urokinase-plasmin(ogen) system may contribute to C. neoformans virulence during invasive cryptococcosis.

Highlights

  • Cryptococcus neoformans is an encapsulated, facultative intracellular pathogen that is globally distributed and recognized as a leading cause of fungal meningitis in immunocompromised people [1,2]

  • Plasminogen pre-coated strains of C. neoformans exhibit plasmindependent invasive activity in the absence of exogenous PA when cultured with brain microvascular endothelial cells (BMEC) [21], implying that the plasminogen becomes activated under these conditions

  • Since BMEC secrete proteins that normally regulate the plasminogen-to-plasmin activation process, we tested whether this regulation occurs when plasminogen is on fungal surfaces

Read more

Summary

Introduction

Cryptococcus neoformans is an encapsulated, facultative intracellular pathogen that is globally distributed and recognized as a leading cause of fungal meningitis in immunocompromised people [1,2]. In the absence of protective T cell immunity, C. neoformans can readily disseminate from the lungs into the CNS by a hematogenous route, where, if left untreated, leads to patient death. Blood-borne C. neoformans can directly invade the blood-brain barrier (BBB) via paracellular passage between brain microvascular endothelial cells (BMEC), or by a transcellular mechanism that is dependent on fungal cell internalization. Specific interactions between C. neoformans and BMEC result in apical-to-basal transcytosis of the fungal pathogen [9,10,11,12,13,14]. Cryptococcal-BBB invasion by a paracellular route occurs in connection with either damage to the brain microvasculature [17,18,19,20,21] or focused proteolytic degradation of inter-endothelial junctions [21]

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