Cargando…

Nigericin Promotes NLRP3-Independent Bacterial Killing in Macrophages

Altered microbiota has been associated with a number of diseases, including inflammatory bowel diseases, diabetes, and cancer. This dysregulation is thought to relate the host inflammatory response to enteric pathogens. Macrophages play a key role in host response to microbes and are involved in bac...

Descripción completa

Detalles Bibliográficos
Autores principales: Armstrong, Heather, Bording-Jorgensen, Michael, Chan, Richard, Wine, Eytan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6779719/
https://www.ncbi.nlm.nih.gov/pubmed/31632394
http://dx.doi.org/10.3389/fimmu.2019.02296
_version_ 1783456954276904960
author Armstrong, Heather
Bording-Jorgensen, Michael
Chan, Richard
Wine, Eytan
author_facet Armstrong, Heather
Bording-Jorgensen, Michael
Chan, Richard
Wine, Eytan
author_sort Armstrong, Heather
collection PubMed
description Altered microbiota has been associated with a number of diseases, including inflammatory bowel diseases, diabetes, and cancer. This dysregulation is thought to relate the host inflammatory response to enteric pathogens. Macrophages play a key role in host response to microbes and are involved in bacterial killing and clearance. This process is partially mediated through the potassium efflux-dependent, cytosolic, PYCARD-containing inflammasome protein complex. Surprisingly, we discovered an alternative mechanism for bacterial killing, independent of the NLRP3 inflammasome/PYCARD. Using the NLRP3 inflammasome-deficient Raw 264.7 and PYCARD-deficient J77 macrophages, which both lack PYCARD, we found that the potassium efflux activator nigericin enhances bacterial killing. Macrophage response to nigericin was examined by RT gene profiling and subsequent qPCR, which demonstrated altered expression of a series of genes involved in the IL-18 bacterial killing pathway. Based on our results we propose a model of bacterial killing, unrelated to NLRP3 inflammasome activation in macrophage cells. Improving understanding of the molecular pathways driving bacterial clearance within macrophage cells will aid in the development of novel immune-targeted therapeutics in a number of diseases.
format Online
Article
Text
id pubmed-6779719
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-67797192019-10-18 Nigericin Promotes NLRP3-Independent Bacterial Killing in Macrophages Armstrong, Heather Bording-Jorgensen, Michael Chan, Richard Wine, Eytan Front Immunol Immunology Altered microbiota has been associated with a number of diseases, including inflammatory bowel diseases, diabetes, and cancer. This dysregulation is thought to relate the host inflammatory response to enteric pathogens. Macrophages play a key role in host response to microbes and are involved in bacterial killing and clearance. This process is partially mediated through the potassium efflux-dependent, cytosolic, PYCARD-containing inflammasome protein complex. Surprisingly, we discovered an alternative mechanism for bacterial killing, independent of the NLRP3 inflammasome/PYCARD. Using the NLRP3 inflammasome-deficient Raw 264.7 and PYCARD-deficient J77 macrophages, which both lack PYCARD, we found that the potassium efflux activator nigericin enhances bacterial killing. Macrophage response to nigericin was examined by RT gene profiling and subsequent qPCR, which demonstrated altered expression of a series of genes involved in the IL-18 bacterial killing pathway. Based on our results we propose a model of bacterial killing, unrelated to NLRP3 inflammasome activation in macrophage cells. Improving understanding of the molecular pathways driving bacterial clearance within macrophage cells will aid in the development of novel immune-targeted therapeutics in a number of diseases. Frontiers Media S.A. 2019-10-01 /pmc/articles/PMC6779719/ /pubmed/31632394 http://dx.doi.org/10.3389/fimmu.2019.02296 Text en Copyright © 2019 Armstrong, Bording-Jorgensen, Chan and Wine. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Armstrong, Heather
Bording-Jorgensen, Michael
Chan, Richard
Wine, Eytan
Nigericin Promotes NLRP3-Independent Bacterial Killing in Macrophages
title Nigericin Promotes NLRP3-Independent Bacterial Killing in Macrophages
title_full Nigericin Promotes NLRP3-Independent Bacterial Killing in Macrophages
title_fullStr Nigericin Promotes NLRP3-Independent Bacterial Killing in Macrophages
title_full_unstemmed Nigericin Promotes NLRP3-Independent Bacterial Killing in Macrophages
title_short Nigericin Promotes NLRP3-Independent Bacterial Killing in Macrophages
title_sort nigericin promotes nlrp3-independent bacterial killing in macrophages
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6779719/
https://www.ncbi.nlm.nih.gov/pubmed/31632394
http://dx.doi.org/10.3389/fimmu.2019.02296
work_keys_str_mv AT armstrongheather nigericinpromotesnlrp3independentbacterialkillinginmacrophages
AT bordingjorgensenmichael nigericinpromotesnlrp3independentbacterialkillinginmacrophages
AT chanrichard nigericinpromotesnlrp3independentbacterialkillinginmacrophages
AT wineeytan nigericinpromotesnlrp3independentbacterialkillinginmacrophages