Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation

Detalhes bibliográficos
Autor(a) principal: Wegiel, Barbara
Data de Publicação: 2014
Outros Autores: Larsen, Rasmus, Gallo, David, Chin, Beek Yoke, Harris, Clair, Mannam, Praveen, Kaczmarek, Elzbieta, Lee, Patty J, Zuckerbraun, Brian S, Flavell, Richard, Soares, Miguel P, Otterbein, Leo E
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10400.7/380
Resumo: Microbial clearance by eukaryotes relies on complex and coordinated processes that remain poorly understood. The gasotransmitter carbon monoxide (CO) is generated by the stress-responsive enzyme heme oxygenase-1 (HO-1, encoded by Hmox1), which is highly induced in macrophages in response to bacterial infection. HO-1 deficiency results in inadequate pathogen clearance, exaggerated tissue damage, and increased mortality. Here, we determined that macrophage-generated CO promotes ATP production and release by bacteria, which then activates the Nacht, LRR, and PYD domains-containing protein 3 (NALP3) inflammasome, intensifying bacterial killing. Bacterial killing defects in HO-1-deficient murine macrophages were restored by administration of CO. Moreover, increased CO levels enhanced the bacterial clearance capacity of human macrophages and WT murine macrophages. CO-dependent bacterial clearance required the NALP3 inflammasome, as CO did not increase bacterial killing in macrophages isolated from NALP3-deficient or caspase-1-deficient mice. IL-1β cleavage and secretion were impaired in HO-1-deficient macrophages, and CO-dependent processing of IL-1β required the presence of bacteria-derived ATP. We found that bacteria remained viable to generate and release ATP in response to CO. The ATP then bound to macrophage nucleotide P2 receptors, resulting in activation of the NALP3/IL-1β inflammasome to amplify bacterial phagocytosis by macrophages. Taken together, our results indicate that macrophage-derived CO permits efficient and coordinated regulation of the host innate response to invading microbes.
id RCAP_f0ae98e560cde9456b0a32c23357f297
oai_identifier_str oai:arca.igc.gulbenkian.pt:10400.7/380
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activationMicrobial clearance by eukaryotes relies on complex and coordinated processes that remain poorly understood. The gasotransmitter carbon monoxide (CO) is generated by the stress-responsive enzyme heme oxygenase-1 (HO-1, encoded by Hmox1), which is highly induced in macrophages in response to bacterial infection. HO-1 deficiency results in inadequate pathogen clearance, exaggerated tissue damage, and increased mortality. Here, we determined that macrophage-generated CO promotes ATP production and release by bacteria, which then activates the Nacht, LRR, and PYD domains-containing protein 3 (NALP3) inflammasome, intensifying bacterial killing. Bacterial killing defects in HO-1-deficient murine macrophages were restored by administration of CO. Moreover, increased CO levels enhanced the bacterial clearance capacity of human macrophages and WT murine macrophages. CO-dependent bacterial clearance required the NALP3 inflammasome, as CO did not increase bacterial killing in macrophages isolated from NALP3-deficient or caspase-1-deficient mice. IL-1β cleavage and secretion were impaired in HO-1-deficient macrophages, and CO-dependent processing of IL-1β required the presence of bacteria-derived ATP. We found that bacteria remained viable to generate and release ATP in response to CO. The ATP then bound to macrophage nucleotide P2 receptors, resulting in activation of the NALP3/IL-1β inflammasome to amplify bacterial phagocytosis by macrophages. Taken together, our results indicate that macrophage-derived CO permits efficient and coordinated regulation of the host innate response to invading microbes.NIH grants: (HL-071797, HL-076167, HL-106227), American Heart Association grants: (10SDG2640091 and NIH R21CA169904-01), Julie Henry Fund, Transplant Center of the BIDMC, FCT grants: (SFRH/BPD/25436/2005, PTDC/BIO/70815/2006, PTDC/BIA-BCM/101311/2008, PTDC/SAU-FCF/100762/2008), the European Community, 6th Framework grant LSH-2005-1.2.5-1 and ERC-2011-AdG, Howard Hughes Medical Institute.American Society for Clinical InvestigationARCAWegiel, BarbaraLarsen, RasmusGallo, DavidChin, Beek YokeHarris, ClairMannam, PraveenKaczmarek, ElzbietaLee, Patty JZuckerbraun, Brian SFlavell, RichardSoares, Miguel POtterbein, Leo E2015-10-07T11:52:02Z2014-112014-11-01T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://hdl.handle.net/10400.7/380engJ Clin Invest. 2014;124(11):4926-4940. doi:10.1172/JCI72853.10.1172/JCI72853info: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:RCAAP2022-11-29T14:34:46Zoai:arca.igc.gulbenkian.pt:10400.7/380Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-19T16:11:40.832620Repositório 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 Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation
title Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation
spellingShingle Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation
Wegiel, Barbara
title_short Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation
title_full Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation
title_fullStr Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation
title_full_unstemmed Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation
title_sort Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation
author Wegiel, Barbara
author_facet Wegiel, Barbara
Larsen, Rasmus
Gallo, David
Chin, Beek Yoke
Harris, Clair
Mannam, Praveen
Kaczmarek, Elzbieta
Lee, Patty J
Zuckerbraun, Brian S
Flavell, Richard
Soares, Miguel P
Otterbein, Leo E
author_role author
author2 Larsen, Rasmus
Gallo, David
Chin, Beek Yoke
Harris, Clair
Mannam, Praveen
Kaczmarek, Elzbieta
Lee, Patty J
Zuckerbraun, Brian S
Flavell, Richard
Soares, Miguel P
Otterbein, Leo E
author2_role author
author
author
author
author
author
author
author
author
author
author
dc.contributor.none.fl_str_mv ARCA
dc.contributor.author.fl_str_mv Wegiel, Barbara
Larsen, Rasmus
Gallo, David
Chin, Beek Yoke
Harris, Clair
Mannam, Praveen
Kaczmarek, Elzbieta
Lee, Patty J
Zuckerbraun, Brian S
Flavell, Richard
Soares, Miguel P
Otterbein, Leo E
description Microbial clearance by eukaryotes relies on complex and coordinated processes that remain poorly understood. The gasotransmitter carbon monoxide (CO) is generated by the stress-responsive enzyme heme oxygenase-1 (HO-1, encoded by Hmox1), which is highly induced in macrophages in response to bacterial infection. HO-1 deficiency results in inadequate pathogen clearance, exaggerated tissue damage, and increased mortality. Here, we determined that macrophage-generated CO promotes ATP production and release by bacteria, which then activates the Nacht, LRR, and PYD domains-containing protein 3 (NALP3) inflammasome, intensifying bacterial killing. Bacterial killing defects in HO-1-deficient murine macrophages were restored by administration of CO. Moreover, increased CO levels enhanced the bacterial clearance capacity of human macrophages and WT murine macrophages. CO-dependent bacterial clearance required the NALP3 inflammasome, as CO did not increase bacterial killing in macrophages isolated from NALP3-deficient or caspase-1-deficient mice. IL-1β cleavage and secretion were impaired in HO-1-deficient macrophages, and CO-dependent processing of IL-1β required the presence of bacteria-derived ATP. We found that bacteria remained viable to generate and release ATP in response to CO. The ATP then bound to macrophage nucleotide P2 receptors, resulting in activation of the NALP3/IL-1β inflammasome to amplify bacterial phagocytosis by macrophages. Taken together, our results indicate that macrophage-derived CO permits efficient and coordinated regulation of the host innate response to invading microbes.
publishDate 2014
dc.date.none.fl_str_mv 2014-11
2014-11-01T00:00:00Z
2015-10-07T11:52:02Z
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/10400.7/380
url http://hdl.handle.net/10400.7/380
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv J Clin Invest. 2014;124(11):4926-4940. doi:10.1172/JCI72853.
10.1172/JCI72853
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 American Society for Clinical Investigation
publisher.none.fl_str_mv American Society for Clinical Investigation
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_ 1799130572205850624