Skip to main content
Top
Published in: Inflammation 3/2020

01-06-2020 | Cytokines | Original Article

Exogenous Hydrogen Sulfide Regulates Mycoplasma pneumoniae Lipid-Associated Membrane Proteins to Induce Expression of Heme Oxygenase-1 and Proinflammatory Cytokines

Authors: Xinnian Hu, Wuwei Zeng, Xiaoxing You, Weiyan Ding, Peng Liu, Liesong Chen, Yanhua Zeng, Cuiming Zhu

Published in: Inflammation | Issue 3/2020

Login to get access

Abstract

This study was designed to investigate the effect of exogenous hydrogen sulfide (H2S) on the secretion of Heme oxygenase (HO-1) and proinflammatory cytokines in human mononuclear cell line THP-1 stimulated by lipid-associated membrane proteins (LAMPs) prepared from Mycoplasma pneumoniae (M. pneumoniae) and explore its regulatory mechanism. Cultured cells were stimulated with M. pneumoniae LAMPs after pretreatment with H2S to analyze the production of proinflammatory cytokines and HO-1 by enzyme-linked immunosorbent assay (ELISA) and Western blot. The results showed that THP-1 cells, which were stimulated by LAMPs after pretreatment with H2S, had decreased production of interleukin-6 (IL-6) and interleukin-8 (IL-8) by inhibiting the mitogen-activated protein kinases (MAPKs)/nuclear factor-kappa B (NF-κB) signaling pathway and increased expression of HO-1 by activating the nuclear factor E2–related factor 2 (Nrf2) signaling pathway. Our results indicate that H2S may play an important role in attenuating inflammation induced by M. pneumoniae LAMPs due to its ability to decrease the production of IL-6 and IL-8 and increase the expression of the HO-1. These findings support further studies for possible clinical applications.
Appendix
Available only for authorised users
Literature
1.
go back to reference Kannan, T.R., J.J. Coalson, M. Cagle, O. Musatovova, R.D. Hardy, and J.B. Baseman. 2011. Synthesis and distribution of CARDS toxin during Mycoplasma pneumoniae infection in a murine model. Journal of Infectious Diseases 204: 1596–1604.CrossRef Kannan, T.R., J.J. Coalson, M. Cagle, O. Musatovova, R.D. Hardy, and J.B. Baseman. 2011. Synthesis and distribution of CARDS toxin during Mycoplasma pneumoniae infection in a murine model. Journal of Infectious Diseases 204: 1596–1604.CrossRef
2.
go back to reference Zhang, H., G. Lv, Y. Shang, L. Liu, Y. Xiang, J. Feng, et al. 2015. Changes of airway reactivity after Mycoplasma pneumoniae infection in children: a study for early precautions against pediatric asthma. Iranian Journal of Allergy, Asthma, and Immunology 14: 476.PubMed Zhang, H., G. Lv, Y. Shang, L. Liu, Y. Xiang, J. Feng, et al. 2015. Changes of airway reactivity after Mycoplasma pneumoniae infection in children: a study for early precautions against pediatric asthma. Iranian Journal of Allergy, Asthma, and Immunology 14: 476.PubMed
3.
go back to reference Waites, K.B., L. Xiao, Y. Liu, M.F. Balish, and T.P. Atkinson. 2017. Mycoplasma pneumoniae from the respiratory tract and beyond. Clinical Microbiology Reviews 30: 747.CrossRef Waites, K.B., L. Xiao, Y. Liu, M.F. Balish, and T.P. Atkinson. 2017. Mycoplasma pneumoniae from the respiratory tract and beyond. Clinical Microbiology Reviews 30: 747.CrossRef
4.
go back to reference Diederen, B.M., V.D.V. Pd, J.A. Kluytmans, M.F. Peeters, and R. Hendrix. 2007. The role of atypical respiratory pathogens in exacerbations of chronic obstructive pulmonary disease. European Respiratory Journal 30: 240–244.CrossRef Diederen, B.M., V.D.V. Pd, J.A. Kluytmans, M.F. Peeters, and R. Hendrix. 2007. The role of atypical respiratory pathogens in exacerbations of chronic obstructive pulmonary disease. European Respiratory Journal 30: 240–244.CrossRef
5.
go back to reference Susanna Esposito, M.D., M.D. Roberta Droghetti, M.D. Samantha Bosis, M.D. Laura Claut, M.D. Paola Marchisio, and M.D. Nicola Principi. 2002. Cytokine secretion in children with acute Mycoplasma pneumoniae infection and wheeze. Pediatric Pulmonology 34: 122–127.CrossRef Susanna Esposito, M.D., M.D. Roberta Droghetti, M.D. Samantha Bosis, M.D. Laura Claut, M.D. Paola Marchisio, and M.D. Nicola Principi. 2002. Cytokine secretion in children with acute Mycoplasma pneumoniae infection and wheeze. Pediatric Pulmonology 34: 122–127.CrossRef
6.
go back to reference Waites, K.B., and D.F. Talkington. 2004. Mycoplasma pneumoniae and its role as a human pathogen. Clinical Microbiology Reviews 17: 697–728.CrossRef Waites, K.B., and D.F. Talkington. 2004. Mycoplasma pneumoniae and its role as a human pathogen. Clinical Microbiology Reviews 17: 697–728.CrossRef
7.
go back to reference Hu, J., C. Chen, G. Ou, X. You, T. Tan, X. Hu, et al. 2017. Nrf2 regulates the inflammatory response, including heme oxygenase-1 induction, by Mycoplasma pneumoniae lipid associated membrane proteins in THP-1 cells. Pathogens & Disease. Hu, J., C. Chen, G. Ou, X. You, T. Tan, X. Hu, et al. 2017. Nrf2 regulates the inflammatory response, including heme oxygenase-1 induction, by Mycoplasma pneumoniae lipid associated membrane proteins in THP-1 cells. Pathogens & Disease.
8.
go back to reference Chmura, K., B.M. Nakamura, P. Kandasamy, and M. Mcgibney. 2008. Induction of IL-8 by Mycoplasma pneumoniae membrane in BEAS-2B cells. American Journal of Physiology—Lung Cellular and Molecular Physiology 295: L220.CrossRef Chmura, K., B.M. Nakamura, P. Kandasamy, and M. Mcgibney. 2008. Induction of IL-8 by Mycoplasma pneumoniae membrane in BEAS-2B cells. American Journal of Physiology—Lung Cellular and Molecular Physiology 295: L220.CrossRef
9.
go back to reference He, J., X. You, Y. Zeng, M. Yu, L. Zuo, and Y. Wu. 2009. Mycoplasma genitalium-derived lipid-associated membrane proteins activate NF-κB through toll-like receptors 1, 2, and 6 and CD14 in a MyD88-dependent pathway. Clinical and Vaccine Immunology 16: 1750–1757.CrossRef He, J., X. You, Y. Zeng, M. Yu, L. Zuo, and Y. Wu. 2009. Mycoplasma genitalium-derived lipid-associated membrane proteins activate NF-κB through toll-like receptors 1, 2, and 6 and CD14 in a MyD88-dependent pathway. Clinical and Vaccine Immunology 16: 1750–1757.CrossRef
10.
go back to reference Mishanina, T.V., M. Libiad, and R. Banerjee. 2015. Biogenesis of reactive sulfur species for signaling by hydrogen sulfide oxidation pathways. Nature Chemical Biology 11: 457–464.CrossRef Mishanina, T.V., M. Libiad, and R. Banerjee. 2015. Biogenesis of reactive sulfur species for signaling by hydrogen sulfide oxidation pathways. Nature Chemical Biology 11: 457–464.CrossRef
11.
go back to reference Li, L., P. Rose, and P.K. Moore. 2011. Hydrogen sulfide and cell signaling. Annual Review of Pharmacology and Toxicology 51: 169–187.CrossRef Li, L., P. Rose, and P.K. Moore. 2011. Hydrogen sulfide and cell signaling. Annual Review of Pharmacology and Toxicology 51: 169–187.CrossRef
12.
go back to reference Zhang, Y., Z.H. Tang, Z. Ren, S.L. Qu, M.H. Liu, L.S. Liu, and Z.S. Jiang. 2013. Hydrogen sulfide: next potent preventive and therapeutic agent in aging and age-associated diseases. Molecular and Cellular Biology 33: 1104–1113.CrossRef Zhang, Y., Z.H. Tang, Z. Ren, S.L. Qu, M.H. Liu, L.S. Liu, and Z.S. Jiang. 2013. Hydrogen sulfide: next potent preventive and therapeutic agent in aging and age-associated diseases. Molecular and Cellular Biology 33: 1104–1113.CrossRef
13.
go back to reference Predmore, B.L., D.J. Lefer, and G. Gojon. 2012. Hydrogen sulfide in biochemistry and medicine. Antioxidants & Redox Signaling 17: 119.CrossRef Predmore, B.L., D.J. Lefer, and G. Gojon. 2012. Hydrogen sulfide in biochemistry and medicine. Antioxidants & Redox Signaling 17: 119.CrossRef
14.
go back to reference Whiteman, M., and P.G. Winyard. 2011. Hydrogen sulfide and inflammation: the good, the bad, the ugly and the promising. Expert Review of Clinical Pharmacology 4: 13–32.CrossRef Whiteman, M., and P.G. Winyard. 2011. Hydrogen sulfide and inflammation: the good, the bad, the ugly and the promising. Expert Review of Clinical Pharmacology 4: 13–32.CrossRef
15.
go back to reference Szabó, C. 2007. Hydrogen sulphide and its therapeutic potential. Nature Reviews Drug Discovery 6: 917–935.CrossRef Szabó, C. 2007. Hydrogen sulphide and its therapeutic potential. Nature Reviews Drug Discovery 6: 917–935.CrossRef
16.
go back to reference Benedetti, F., S. Davinelli, S. Krishnan, R.C. Gallo, G. Scapagnini, D. Zella, et al. 2014. Sulfur compounds block MCP-1 production by Mycoplasma fermentans -infected macrophages through NF-κB inhibition. Journal of Translational Medicine 12: 145.CrossRef Benedetti, F., S. Davinelli, S. Krishnan, R.C. Gallo, G. Scapagnini, D. Zella, et al. 2014. Sulfur compounds block MCP-1 production by Mycoplasma fermentans -infected macrophages through NF-κB inhibition. Journal of Translational Medicine 12: 145.CrossRef
17.
go back to reference Kaspar, J.W., and Niture SKJaiswal AK. 2009. Nrf2:INrf2 (Keap1) signaling in oxidative stress. Free Radical Biology & Medicine 47: 1304–1309.CrossRef Kaspar, J.W., and Niture SKJaiswal AK. 2009. Nrf2:INrf2 (Keap1) signaling in oxidative stress. Free Radical Biology & Medicine 47: 1304–1309.CrossRef
18.
go back to reference Wu, Y., D. Li, Y. Wang, X. Liu, Y. Zhang, W. Qu, et al. 2018. Beta-defensin 2 and 3 promote bacterial clearance of Pseudomonas aeruginosa by inhibiting macrophage autophagy through downregulation of early growth response gene-1 and c-FOS. Frontiers in Immunology 9. Wu, Y., D. Li, Y. Wang, X. Liu, Y. Zhang, W. Qu, et al. 2018. Beta-defensin 2 and 3 promote bacterial clearance of Pseudomonas aeruginosa by inhibiting macrophage autophagy through downregulation of early growth response gene-1 and c-FOS. Frontiers in Immunology 9.
Metadata
Title
Exogenous Hydrogen Sulfide Regulates Mycoplasma pneumoniae Lipid-Associated Membrane Proteins to Induce Expression of Heme Oxygenase-1 and Proinflammatory Cytokines
Authors
Xinnian Hu
Wuwei Zeng
Xiaoxing You
Weiyan Ding
Peng Liu
Liesong Chen
Yanhua Zeng
Cuiming Zhu
Publication date
01-06-2020
Publisher
Springer US
Keyword
Cytokines
Published in
Inflammation / Issue 3/2020
Print ISSN: 0360-3997
Electronic ISSN: 1573-2576
DOI
https://doi.org/10.1007/s10753-019-01170-3

Other articles of this Issue 3/2020

Inflammation 3/2020 Go to the issue