Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Streptococcal M1 Protein-Provoked CXC Chemokine Formation, Neutrophil Recruitment and Lung Damage Are Regulated by Rho-Kinase Signaling.

Zhang, Songen LU ; Rahman, Milladur LU orcid ; Zhang, Su LU ; Herwald, Heiko LU orcid ; Qi, Zhongquan LU ; Jeppsson, Bengt LU and Thorlacius, Henrik LU (2012) In Journal of Innate Immunity 4(4). p.399-408
Abstract
Streptococcal toxic shock syndrome is frequently caused by Streptococcus pyogenes of the M1 serotype. The aim of this study was to determine the role of Ras-homologous (Rho)-kinase signaling in M1 protein-provoked lung damage. Male C57BL/6 mice received the Rho-kinase-specific inhibitor Y-27632 before administration of M1 protein. Edema, neutrophil accumulation and CXC chemokines were quantified in the lung 4 h after M1 protein challenge. Flow cytometry was used to determine Mac-1 expression. Quantitative RT-PCR was used to determine gene expression of CXC chemokine mRNA in alveolar macrophages. M1 protein increased neutrophil accumulation, edema and CXC chemokine formation in the lung as well as enhanced Mac-1 expression on neutrophils.... (More)
Streptococcal toxic shock syndrome is frequently caused by Streptococcus pyogenes of the M1 serotype. The aim of this study was to determine the role of Ras-homologous (Rho)-kinase signaling in M1 protein-provoked lung damage. Male C57BL/6 mice received the Rho-kinase-specific inhibitor Y-27632 before administration of M1 protein. Edema, neutrophil accumulation and CXC chemokines were quantified in the lung 4 h after M1 protein challenge. Flow cytometry was used to determine Mac-1 expression. Quantitative RT-PCR was used to determine gene expression of CXC chemokine mRNA in alveolar macrophages. M1 protein increased neutrophil accumulation, edema and CXC chemokine formation in the lung as well as enhanced Mac-1 expression on neutrophils. Inhibition of Rho-kinase signaling significantly reduced M1 protein-provoked neutrophil accumulation and edema formation in the lung. M1 protein-triggered pulmonary production of CXC chemokine and gene expression of CXC chemokines in alveolar macrophages was decreased by Y-27632. Moreover, Rho-kinase inhibition attenuated M1 protein-induced Mac-1 expression on neutrophils. We conclude that Rho-kinase-dependent neutrophil infiltration controls pulmonary tissue damage in response to streptococcal M1 protein and that Rho-kinase signaling regulates M1 protein-induced lung recruitment of neutrophils via the formation of CXC chemokines and Mac-1 expression. (Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; and
organization
publishing date
type
Contribution to journal
publication status
published
subject
in
Journal of Innate Immunity
volume
4
issue
4
pages
399 - 408
publisher
Karger
external identifiers
  • wos:000305819800009
  • pmid:22433673
  • scopus:84863309800
  • pmid:22433673
ISSN
1662-811X
DOI
10.1159/000336182
language
English
LU publication?
yes
id
db3cb0b8-7041-4fe8-8df5-4566e70f6895 (old id 2431610)
alternative location
http://www.ncbi.nlm.nih.gov/pubmed/22433673?dopt=Abstract
date added to LUP
2016-04-01 10:23:47
date last changed
2022-04-27 21:40:44
@article{db3cb0b8-7041-4fe8-8df5-4566e70f6895,
  abstract     = {{Streptococcal toxic shock syndrome is frequently caused by Streptococcus pyogenes of the M1 serotype. The aim of this study was to determine the role of Ras-homologous (Rho)-kinase signaling in M1 protein-provoked lung damage. Male C57BL/6 mice received the Rho-kinase-specific inhibitor Y-27632 before administration of M1 protein. Edema, neutrophil accumulation and CXC chemokines were quantified in the lung 4 h after M1 protein challenge. Flow cytometry was used to determine Mac-1 expression. Quantitative RT-PCR was used to determine gene expression of CXC chemokine mRNA in alveolar macrophages. M1 protein increased neutrophil accumulation, edema and CXC chemokine formation in the lung as well as enhanced Mac-1 expression on neutrophils. Inhibition of Rho-kinase signaling significantly reduced M1 protein-provoked neutrophil accumulation and edema formation in the lung. M1 protein-triggered pulmonary production of CXC chemokine and gene expression of CXC chemokines in alveolar macrophages was decreased by Y-27632. Moreover, Rho-kinase inhibition attenuated M1 protein-induced Mac-1 expression on neutrophils. We conclude that Rho-kinase-dependent neutrophil infiltration controls pulmonary tissue damage in response to streptococcal M1 protein and that Rho-kinase signaling regulates M1 protein-induced lung recruitment of neutrophils via the formation of CXC chemokines and Mac-1 expression.}},
  author       = {{Zhang, Songen and Rahman, Milladur and Zhang, Su and Herwald, Heiko and Qi, Zhongquan and Jeppsson, Bengt and Thorlacius, Henrik}},
  issn         = {{1662-811X}},
  language     = {{eng}},
  number       = {{4}},
  pages        = {{399--408}},
  publisher    = {{Karger}},
  series       = {{Journal of Innate Immunity}},
  title        = {{Streptococcal M1 Protein-Provoked CXC Chemokine Formation, Neutrophil Recruitment and Lung Damage Are Regulated by Rho-Kinase Signaling.}},
  url          = {{https://lup.lub.lu.se/search/files/1809660/3688050.pdf}},
  doi          = {{10.1159/000336182}},
  volume       = {{4}},
  year         = {{2012}},
}