The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression

Detalhes bibliográficos
Autor(a) principal: Gresnigt, Mark S.
Data de Publicação: 2017
Outros Autores: Jaeger, Martin, Malireddi, R. K. Subbarao, Rasid, Orhan, Jouvion, Gregory, Fitting, Catherine, Melchers, Willem J. G., Kanneganti, Thirumala-Devi, Carvalho, Agostinho, Ibrahim-Granet, Oumaima, van de Veerdonk, Frank L.
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/1822/50601
Resumo: One of the major life-threatening infections for which severely immunocompromised patients are at risk is invasive aspergillosis (IA). Despite the current treatment options, the increasing antifungal resistance and poor outcome highlight the need for novel therapeutic strategies to improve outcome of patients with IA. In the current study, we investigated whether and how the intracellular pattern recognition receptor NOD1 is involved in host defense against Aspergillus fumigatus. When exploring the role of NOD1 in an experimental mouse model, we found that Nod1(-/-) mice were protected against IA and demonstrated reduced fungal outgrowth in the lungs. We found that macrophages derived from bone marrow of Nod1(-/-) mice were more efficiently inducing reactive oxygen species and cytokines in response to Aspergillus. Most strikingly, these cells were highly potent in killing A. fumigatus compared with wild-type cells. In line, human macrophages in which NOD1 was silenced demonstrated augmented Aspergillus killing and NOD1 stimulation decreased fungal killing. The differentially altered killing capacity of NOD1 silencing versus NOD1 activation was associated with alterations in dectin-1 expression, with activation of NOD1 reducing dectin-1 expression. Furthermore, we were able to demonstrate that Nod1(-/-) mice have elevated dectin-1 expression in the lung and bone marrow, and silencing of NOD1 gene expression in human macrophages increases dectin-1 expression. The enhanced dectin-1 expression may be the mechanism of enhanced fungal killing of Nod1(-/-) cells and human cells in which NOD1 was silenced, since blockade of dectin-1 reversed the augmented killing in these cells. Collectively, our data demonstrate that NOD1 receptor plays an inhibitory role in the host defense against Aspergillus. This provides a rationale to develop novel immunotherapeutic strategies for treatment of aspergillosis that target the NOD1 receptor, to enhance the efficiency of host immune cells to clear the infection by increasing fungal kil
id RCAP_7c77f1c2e12e851bfde31453adc6d6a7
oai_identifier_str oai:repositorium.sdum.uminho.pt:1822/50601
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 ExpressionNOD1Aspergillus fumigatusNucleotide-binding oligomerization domaindectin-1Fungal killingCiências Médicas::Medicina BásicaScience & TechnologyOne of the major life-threatening infections for which severely immunocompromised patients are at risk is invasive aspergillosis (IA). Despite the current treatment options, the increasing antifungal resistance and poor outcome highlight the need for novel therapeutic strategies to improve outcome of patients with IA. In the current study, we investigated whether and how the intracellular pattern recognition receptor NOD1 is involved in host defense against Aspergillus fumigatus. When exploring the role of NOD1 in an experimental mouse model, we found that Nod1(-/-) mice were protected against IA and demonstrated reduced fungal outgrowth in the lungs. We found that macrophages derived from bone marrow of Nod1(-/-) mice were more efficiently inducing reactive oxygen species and cytokines in response to Aspergillus. Most strikingly, these cells were highly potent in killing A. fumigatus compared with wild-type cells. In line, human macrophages in which NOD1 was silenced demonstrated augmented Aspergillus killing and NOD1 stimulation decreased fungal killing. The differentially altered killing capacity of NOD1 silencing versus NOD1 activation was associated with alterations in dectin-1 expression, with activation of NOD1 reducing dectin-1 expression. Furthermore, we were able to demonstrate that Nod1(-/-) mice have elevated dectin-1 expression in the lung and bone marrow, and silencing of NOD1 gene expression in human macrophages increases dectin-1 expression. The enhanced dectin-1 expression may be the mechanism of enhanced fungal killing of Nod1(-/-) cells and human cells in which NOD1 was silenced, since blockade of dectin-1 reversed the augmented killing in these cells. Collectively, our data demonstrate that NOD1 receptor plays an inhibitory role in the host defense against Aspergillus. This provides a rationale to develop novel immunotherapeutic strategies for treatment of aspergillosis that target the NOD1 receptor, to enhance the efficiency of host immune cells to clear the infection by increasing fungal kilMG was supported by the Erasmus lifelong learning program. FV was supported by the E-rare project EURO-CMC. TK was supported by the National Institutes of Health [grant numbers AI101935, AI124346, AR056296, and CA163507]. AC was supported by the Northern Portugal Regional Operational Programme (NORTE 2020), under the Portugal 2020 Partnership Agreement, through the European Regional Development Fund (FEDER) (NORTE-01-0145FEDER-000013), and the Fundação para a Ciência e Tecnologia (FCT) (IF/00735/2014).info:eu-repo/semantics/publishedVersionFrontiers MediaUniversidade do MinhoGresnigt, Mark S.Jaeger, MartinMalireddi, R. K. SubbaraoRasid, OrhanJouvion, GregoryFitting, CatherineMelchers, Willem J. G.Kanneganti, Thirumala-DeviCarvalho, AgostinhoIbrahim-Granet, Oumaimavan de Veerdonk, Frank L.2017-12-132017-12-13T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://hdl.handle.net/1822/50601engGresnigt, M. S., Jaeger, M., Malireddi, R. S., Rasid, O., et. al. (2017). The absence of nOD1 enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 expression. Frontiers in immunology, 81664-322410.3389/fimmu.2017.01777https://www.frontiersin.org/articles/10.3389/fimmu.2017.01777/fullinfo:eu-repo/semantics/openAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2023-07-21T12:19:26Zoai:repositorium.sdum.uminho.pt:1822/50601Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-19T19:12:22.226287Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãofalse
dc.title.none.fl_str_mv The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
title The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
spellingShingle The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
Gresnigt, Mark S.
NOD1
Aspergillus fumigatus
Nucleotide-binding oligomerization domain
dectin-1
Fungal killing
Ciências Médicas::Medicina Básica
Science & Technology
title_short The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
title_full The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
title_fullStr The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
title_full_unstemmed The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
title_sort The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
author Gresnigt, Mark S.
author_facet Gresnigt, Mark S.
Jaeger, Martin
Malireddi, R. K. Subbarao
Rasid, Orhan
Jouvion, Gregory
Fitting, Catherine
Melchers, Willem J. G.
Kanneganti, Thirumala-Devi
Carvalho, Agostinho
Ibrahim-Granet, Oumaima
van de Veerdonk, Frank L.
author_role author
author2 Jaeger, Martin
Malireddi, R. K. Subbarao
Rasid, Orhan
Jouvion, Gregory
Fitting, Catherine
Melchers, Willem J. G.
Kanneganti, Thirumala-Devi
Carvalho, Agostinho
Ibrahim-Granet, Oumaima
van de Veerdonk, Frank L.
author2_role author
author
author
author
author
author
author
author
author
author
dc.contributor.none.fl_str_mv Universidade do Minho
dc.contributor.author.fl_str_mv Gresnigt, Mark S.
Jaeger, Martin
Malireddi, R. K. Subbarao
Rasid, Orhan
Jouvion, Gregory
Fitting, Catherine
Melchers, Willem J. G.
Kanneganti, Thirumala-Devi
Carvalho, Agostinho
Ibrahim-Granet, Oumaima
van de Veerdonk, Frank L.
dc.subject.por.fl_str_mv NOD1
Aspergillus fumigatus
Nucleotide-binding oligomerization domain
dectin-1
Fungal killing
Ciências Médicas::Medicina Básica
Science & Technology
topic NOD1
Aspergillus fumigatus
Nucleotide-binding oligomerization domain
dectin-1
Fungal killing
Ciências Médicas::Medicina Básica
Science & Technology
description One of the major life-threatening infections for which severely immunocompromised patients are at risk is invasive aspergillosis (IA). Despite the current treatment options, the increasing antifungal resistance and poor outcome highlight the need for novel therapeutic strategies to improve outcome of patients with IA. In the current study, we investigated whether and how the intracellular pattern recognition receptor NOD1 is involved in host defense against Aspergillus fumigatus. When exploring the role of NOD1 in an experimental mouse model, we found that Nod1(-/-) mice were protected against IA and demonstrated reduced fungal outgrowth in the lungs. We found that macrophages derived from bone marrow of Nod1(-/-) mice were more efficiently inducing reactive oxygen species and cytokines in response to Aspergillus. Most strikingly, these cells were highly potent in killing A. fumigatus compared with wild-type cells. In line, human macrophages in which NOD1 was silenced demonstrated augmented Aspergillus killing and NOD1 stimulation decreased fungal killing. The differentially altered killing capacity of NOD1 silencing versus NOD1 activation was associated with alterations in dectin-1 expression, with activation of NOD1 reducing dectin-1 expression. Furthermore, we were able to demonstrate that Nod1(-/-) mice have elevated dectin-1 expression in the lung and bone marrow, and silencing of NOD1 gene expression in human macrophages increases dectin-1 expression. The enhanced dectin-1 expression may be the mechanism of enhanced fungal killing of Nod1(-/-) cells and human cells in which NOD1 was silenced, since blockade of dectin-1 reversed the augmented killing in these cells. Collectively, our data demonstrate that NOD1 receptor plays an inhibitory role in the host defense against Aspergillus. This provides a rationale to develop novel immunotherapeutic strategies for treatment of aspergillosis that target the NOD1 receptor, to enhance the efficiency of host immune cells to clear the infection by increasing fungal kil
publishDate 2017
dc.date.none.fl_str_mv 2017-12-13
2017-12-13T00:00:00Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/1822/50601
url http://hdl.handle.net/1822/50601
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv Gresnigt, M. S., Jaeger, M., Malireddi, R. S., Rasid, O., et. al. (2017). The absence of nOD1 enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 expression. Frontiers in immunology, 8
1664-3224
10.3389/fimmu.2017.01777
https://www.frontiersin.org/articles/10.3389/fimmu.2017.01777/full
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.publisher.none.fl_str_mv Frontiers Media
publisher.none.fl_str_mv Frontiers Media
dc.source.none.fl_str_mv reponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron:RCAAP
instname_str Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron_str RCAAP
institution RCAAP
reponame_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
collection Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository.name.fl_str_mv Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
repository.mail.fl_str_mv
_version_ 1799132558707916800