Cargando…

Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition

BACKGROUND AND AIMS: Hydrogen sulfide (H(2)S), together with nitric oxide (NO) and carbon monoxide (CO), belongs to a family of endogenous signaling mediators termed “gasotransmitters”. Recent studies suggest that H(2)S modulates many cellular processes and it has been recognized to play a central r...

Descripción completa

Detalles Bibliográficos
Autores principales: Benedetti, Francesca, Davinelli, Sergio, Krishnan, Selvi, Gallo, Robert C, Scapagnini, Giovanni, Zella, Davide, Curreli, Sabrina
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4046042/
https://www.ncbi.nlm.nih.gov/pubmed/24886588
http://dx.doi.org/10.1186/1479-5876-12-145
_version_ 1782319441053220864
author Benedetti, Francesca
Davinelli, Sergio
Krishnan, Selvi
Gallo, Robert C
Scapagnini, Giovanni
Zella, Davide
Curreli, Sabrina
author_facet Benedetti, Francesca
Davinelli, Sergio
Krishnan, Selvi
Gallo, Robert C
Scapagnini, Giovanni
Zella, Davide
Curreli, Sabrina
author_sort Benedetti, Francesca
collection PubMed
description BACKGROUND AND AIMS: Hydrogen sulfide (H(2)S), together with nitric oxide (NO) and carbon monoxide (CO), belongs to a family of endogenous signaling mediators termed “gasotransmitters”. Recent studies suggest that H(2)S modulates many cellular processes and it has been recognized to play a central role in inflammation, in the cardiovascular and nervous systems. By infecting monocytes/macrophages with Mycoplasma fermentans (M.F.), a well-known pro-inflammatory agent, we evaluated the effects of H(2)S. METHODS: M.F.-infected cells were analyzed by ELISA and real time RT-PCR to detect the M.F. effects on MCP-1 and on MMP-12 expression. The role of two different H(2)S donors (NaHS and GYY4137) on MF-infected cells was determined by treating infected cells with H(2)S and then testing the culture supernatants for MCP-1 and on MMP-12 production by ELISA assay. In order to identify the pathway/s mediating H(2)S- anti-inflammatory activity, cells were also treated with specific pharmaceutical inhibitors. Cytoplasmic and nuclear accumulation of NF-κB heterodimers was analyzed. RESULTS: We show that H(2)S was able to reduce the production of pro-inflammatory cytokine MCP-1, that was induced in monocytes/macrophages during M.F. infection. Moreover, MCP-1 was induced by M.F. through Toll-like receptor (TLR)-mediated nuclear factor-κB (NF-κB) activation, as demonstrated by the fact that TLR inhibitors TIRAP and MyD88 and NF-κB inhibitor IKK were able to block the cytokine production. In contrast H(2)S treatment of M.F. infected macrophages reduced nuclear accumulation of NF-κB heterodimer p65/p52. CONCLUSIONS: Our data demonstrate that under the present conditions H(2)S is effective in reducing Mycoplasma-induced inflammation by targeting the NF-κB pathway. This supports further studies for possible clinical applications.
format Online
Article
Text
id pubmed-4046042
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-40460422014-06-06 Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition Benedetti, Francesca Davinelli, Sergio Krishnan, Selvi Gallo, Robert C Scapagnini, Giovanni Zella, Davide Curreli, Sabrina J Transl Med Research BACKGROUND AND AIMS: Hydrogen sulfide (H(2)S), together with nitric oxide (NO) and carbon monoxide (CO), belongs to a family of endogenous signaling mediators termed “gasotransmitters”. Recent studies suggest that H(2)S modulates many cellular processes and it has been recognized to play a central role in inflammation, in the cardiovascular and nervous systems. By infecting monocytes/macrophages with Mycoplasma fermentans (M.F.), a well-known pro-inflammatory agent, we evaluated the effects of H(2)S. METHODS: M.F.-infected cells were analyzed by ELISA and real time RT-PCR to detect the M.F. effects on MCP-1 and on MMP-12 expression. The role of two different H(2)S donors (NaHS and GYY4137) on MF-infected cells was determined by treating infected cells with H(2)S and then testing the culture supernatants for MCP-1 and on MMP-12 production by ELISA assay. In order to identify the pathway/s mediating H(2)S- anti-inflammatory activity, cells were also treated with specific pharmaceutical inhibitors. Cytoplasmic and nuclear accumulation of NF-κB heterodimers was analyzed. RESULTS: We show that H(2)S was able to reduce the production of pro-inflammatory cytokine MCP-1, that was induced in monocytes/macrophages during M.F. infection. Moreover, MCP-1 was induced by M.F. through Toll-like receptor (TLR)-mediated nuclear factor-κB (NF-κB) activation, as demonstrated by the fact that TLR inhibitors TIRAP and MyD88 and NF-κB inhibitor IKK were able to block the cytokine production. In contrast H(2)S treatment of M.F. infected macrophages reduced nuclear accumulation of NF-κB heterodimer p65/p52. CONCLUSIONS: Our data demonstrate that under the present conditions H(2)S is effective in reducing Mycoplasma-induced inflammation by targeting the NF-κB pathway. This supports further studies for possible clinical applications. BioMed Central 2014-05-24 /pmc/articles/PMC4046042/ /pubmed/24886588 http://dx.doi.org/10.1186/1479-5876-12-145 Text en Copyright © 2014 Benedetti et al.; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/4.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Benedetti, Francesca
Davinelli, Sergio
Krishnan, Selvi
Gallo, Robert C
Scapagnini, Giovanni
Zella, Davide
Curreli, Sabrina
Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition
title Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition
title_full Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition
title_fullStr Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition
title_full_unstemmed Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition
title_short Sulfur compounds block MCP-1 production by Mycoplasma fermentans-infected macrophages through NF-κB inhibition
title_sort sulfur compounds block mcp-1 production by mycoplasma fermentans-infected macrophages through nf-κb inhibition
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4046042/
https://www.ncbi.nlm.nih.gov/pubmed/24886588
http://dx.doi.org/10.1186/1479-5876-12-145
work_keys_str_mv AT benedettifrancesca sulfurcompoundsblockmcp1productionbymycoplasmafermentansinfectedmacrophagesthroughnfkbinhibition
AT davinellisergio sulfurcompoundsblockmcp1productionbymycoplasmafermentansinfectedmacrophagesthroughnfkbinhibition
AT krishnanselvi sulfurcompoundsblockmcp1productionbymycoplasmafermentansinfectedmacrophagesthroughnfkbinhibition
AT gallorobertc sulfurcompoundsblockmcp1productionbymycoplasmafermentansinfectedmacrophagesthroughnfkbinhibition
AT scapagninigiovanni sulfurcompoundsblockmcp1productionbymycoplasmafermentansinfectedmacrophagesthroughnfkbinhibition
AT zelladavide sulfurcompoundsblockmcp1productionbymycoplasmafermentansinfectedmacrophagesthroughnfkbinhibition
AT currelisabrina sulfurcompoundsblockmcp1productionbymycoplasmafermentansinfectedmacrophagesthroughnfkbinhibition