Immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent Cryptococcus neoformans in mice.

C. neoformans is a leading cause of fatal mycosis linked to CNS dissemination. Laccase, encoded by the LAC1 gene, is an important virulence factor implicated in brain dissemination yet little is known about the mechanism(s) accounting for this observation. Here, we investigated whether the presence...

Full description

Bibliographic Details
Main Authors: Yafeng Qiu, Michael J Davis, Jeremy K Dayrit, Zachary Hadd, Daniel L Meister, John J Osterholzer, Peter R Williamson, Michal A Olszewski
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2012-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3478276?pdf=render
id doaj-14f108be0f3f432b852455f2eec1ca8b
record_format Article
spelling doaj-14f108be0f3f432b852455f2eec1ca8b2020-11-24T22:08:10ZengPublic Library of Science (PLoS)PLoS ONE1932-62032012-01-01710e4785310.1371/journal.pone.0047853Immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent Cryptococcus neoformans in mice.Yafeng QiuMichael J DavisJeremy K DayritZachary HaddDaniel L MeisterJohn J OsterholzerPeter R WilliamsonMichal A OlszewskiC. neoformans is a leading cause of fatal mycosis linked to CNS dissemination. Laccase, encoded by the LAC1 gene, is an important virulence factor implicated in brain dissemination yet little is known about the mechanism(s) accounting for this observation. Here, we investigated whether the presence or absence of laccase altered the local immune response in the lungs by comparing infections with the highly virulent strain, H99 (which expresses laccase) and mutant strain of H99 deficient in laccase (lac1Δ) in a mouse model of pulmonary infection. We found that LAC1 gene deletion decreased the pulmonary fungal burden and abolished CNS dissemination at weeks 2 and 3. Furthermore, LAC1 deletion lead to: 1) diminished pulmonary eosinophilia; 2) increased accumulation of CD4+ and CD8+ T cells; 3) increased Th1 and Th17 cytokines yet decreased Th2 cytokines; and 4) lung macrophage shifting of the lung macrophage phenotype from M2- towards M1-type activation. Next, we used adoptively transferred CD4+ T cells isolated from pulmonary lymph nodes of mice infected with either lac1Δ or H99 to evaluate the role of laccase-induced immunomodulation on CNS dissemination. We found that in comparison to PBS treated mice, adoptively transferred CD4+ T cells isolated from lac1Δ-infected mice decreased CNS dissemination, while those isolated from H99-infected mice increased CNS dissemination. Collectively, our findings reveal that immune modulation away from Th1/Th17 responses and towards Th2 responses represents a novel mechanism through which laccase can contribute to cryptococcal virulence. Furthermore, our data support the hypothesis that laccase-induced changes in polarization of CD4+ T cells contribute to CNS dissemination.http://europepmc.org/articles/PMC3478276?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Yafeng Qiu
Michael J Davis
Jeremy K Dayrit
Zachary Hadd
Daniel L Meister
John J Osterholzer
Peter R Williamson
Michal A Olszewski
spellingShingle Yafeng Qiu
Michael J Davis
Jeremy K Dayrit
Zachary Hadd
Daniel L Meister
John J Osterholzer
Peter R Williamson
Michal A Olszewski
Immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent Cryptococcus neoformans in mice.
PLoS ONE
author_facet Yafeng Qiu
Michael J Davis
Jeremy K Dayrit
Zachary Hadd
Daniel L Meister
John J Osterholzer
Peter R Williamson
Michal A Olszewski
author_sort Yafeng Qiu
title Immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent Cryptococcus neoformans in mice.
title_short Immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent Cryptococcus neoformans in mice.
title_full Immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent Cryptococcus neoformans in mice.
title_fullStr Immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent Cryptococcus neoformans in mice.
title_full_unstemmed Immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent Cryptococcus neoformans in mice.
title_sort immune modulation mediated by cryptococcal laccase promotes pulmonary growth and brain dissemination of virulent cryptococcus neoformans in mice.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2012-01-01
description C. neoformans is a leading cause of fatal mycosis linked to CNS dissemination. Laccase, encoded by the LAC1 gene, is an important virulence factor implicated in brain dissemination yet little is known about the mechanism(s) accounting for this observation. Here, we investigated whether the presence or absence of laccase altered the local immune response in the lungs by comparing infections with the highly virulent strain, H99 (which expresses laccase) and mutant strain of H99 deficient in laccase (lac1Δ) in a mouse model of pulmonary infection. We found that LAC1 gene deletion decreased the pulmonary fungal burden and abolished CNS dissemination at weeks 2 and 3. Furthermore, LAC1 deletion lead to: 1) diminished pulmonary eosinophilia; 2) increased accumulation of CD4+ and CD8+ T cells; 3) increased Th1 and Th17 cytokines yet decreased Th2 cytokines; and 4) lung macrophage shifting of the lung macrophage phenotype from M2- towards M1-type activation. Next, we used adoptively transferred CD4+ T cells isolated from pulmonary lymph nodes of mice infected with either lac1Δ or H99 to evaluate the role of laccase-induced immunomodulation on CNS dissemination. We found that in comparison to PBS treated mice, adoptively transferred CD4+ T cells isolated from lac1Δ-infected mice decreased CNS dissemination, while those isolated from H99-infected mice increased CNS dissemination. Collectively, our findings reveal that immune modulation away from Th1/Th17 responses and towards Th2 responses represents a novel mechanism through which laccase can contribute to cryptococcal virulence. Furthermore, our data support the hypothesis that laccase-induced changes in polarization of CD4+ T cells contribute to CNS dissemination.
url http://europepmc.org/articles/PMC3478276?pdf=render
work_keys_str_mv AT yafengqiu immunemodulationmediatedbycryptococcallaccasepromotespulmonarygrowthandbraindisseminationofvirulentcryptococcusneoformansinmice
AT michaeljdavis immunemodulationmediatedbycryptococcallaccasepromotespulmonarygrowthandbraindisseminationofvirulentcryptococcusneoformansinmice
AT jeremykdayrit immunemodulationmediatedbycryptococcallaccasepromotespulmonarygrowthandbraindisseminationofvirulentcryptococcusneoformansinmice
AT zacharyhadd immunemodulationmediatedbycryptococcallaccasepromotespulmonarygrowthandbraindisseminationofvirulentcryptococcusneoformansinmice
AT daniellmeister immunemodulationmediatedbycryptococcallaccasepromotespulmonarygrowthandbraindisseminationofvirulentcryptococcusneoformansinmice
AT johnjosterholzer immunemodulationmediatedbycryptococcallaccasepromotespulmonarygrowthandbraindisseminationofvirulentcryptococcusneoformansinmice
AT peterrwilliamson immunemodulationmediatedbycryptococcallaccasepromotespulmonarygrowthandbraindisseminationofvirulentcryptococcusneoformansinmice
AT michalaolszewski immunemodulationmediatedbycryptococcallaccasepromotespulmonarygrowthandbraindisseminationofvirulentcryptococcusneoformansinmice
_version_ 1725817333307408384