Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

EGFR transactivation contributes to neuroinflammation in Streptococcus suis meningitis

Authors: Xiao-Pei Yang, Ji-Yang Fu, Rui-Cheng Yang, Wen-Tong Liu, Tao Zhang, Bo Yang, Ling Miao, Bei-Bei Dou, Chen Tan, Huan-Chun Chen, Xiang-Ru Wang

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

Streptococcus suis serotype 2 (SS2) is an important zoonotic bacterial pathogen in both humans and animals, which can cause high morbidity and mortality. Meningitis is one of the major clinical manifestations of SS2 infection. However, the specific process of SS2 meningitis and its molecular mechanisms remain unclear. Epidermal growth factor receptor (EGFR) has been reported to initiate transduction of intracellular signals and regulate host inflammatory responses. Whether and how EGFR contributes to the development of S. suis meningitis are currently unknown.

Methods

The tyrosine phosphorylation of cellular proteins, the transactivation of EGFR, as well as its dimerization, and the associated signal transduction pathways were investigated by immunoprecipitation and western blotting. Real-time quantitative PCR was used to investigate the transcriptional level of the ErbB family members, EGFR-related ligands, cytokines, and chemokines. The secretion of cytokines and chemokines in the serum and brain were detected by Q-Plex™ Chemiluminescent ELISA.

Results

We found an important role of EGFR in SS2 strain SC19-induced meningitis. SC19 increasingly adhered to human brain microvascular endothelial cells (hBMEC) and caused inflammatory lesions in the brain tissues, with significant induction and secretion of proinflammatory cytokines and chemokines in the serum and brains. SC19 infection of hBMEC induced tyrosine phosphorylation of cellular EGFR in a ligand-dependent manner involving the EGF-like ligand HB-EGF, amphiregulin (AREG), and epiregulin (EREG) and led to heterodimerization of EGFR/ErbB3. The EGFR transactivation did not participate in SS2 strain SC19 adhesion of hBMEC, as well as in bacterial colonization in vivo. However, its transactivation contributed to the bacterial-induced neuroinflammation, via triggering the MAPK-ERK1/2 and NF-κB signaling pathways in hBMEC that promote the production of proinflammatory cytokines and chemokines.

Conclusions

We investigated for the first time the tyrosine phosphorylation of cellular proteins in response to SS2 strain SC19 infection of hBMEC and demonstrated the contribution of EGFR to SS2-induced neuroinflammation. These observations propose a novel mechanism involving EGFR in SS2-mediated inflammatory responses in the brain, and therefore, EGFR might be an important host target for further investigation and prevention of neuroinflammation caused by SS2 strains.
Literature
1.
go back to reference van Samkar A, Brouwer MC, Schultsz C, van der Ende A, van de Beek D. Streptococcus suis meningitis: a systematic review and meta-analysis. PLoS Negl Trop Dis. 2015;9:e0004191.CrossRefPubMedPubMedCentral van Samkar A, Brouwer MC, Schultsz C, van der Ende A, van de Beek D. Streptococcus suis meningitis: a systematic review and meta-analysis. PLoS Negl Trop Dis. 2015;9:e0004191.CrossRefPubMedPubMedCentral
2.
go back to reference Yu H, Jing H, Chen Z, Zheng H, Zhu X, Wang H, Wang S, Liu L, Zu R, Luo L, et al. Human Streptococcus suis outbreak, Sichuan, China. Emerg Infect Dis. 2006;12:914–20.CrossRefPubMedPubMedCentral Yu H, Jing H, Chen Z, Zheng H, Zhu X, Wang H, Wang S, Liu L, Zu R, Luo L, et al. Human Streptococcus suis outbreak, Sichuan, China. Emerg Infect Dis. 2006;12:914–20.CrossRefPubMedPubMedCentral
3.
go back to reference Hoa NT, Chieu TT, Do Dung S, Long NT, Hieu TQ, Luc NT, Nhuong PT, Huong VT, Trinh DT, Wertheim HF, et al. Streptococcus suis and porcine reproductive and respiratory syndrome, Vietnam. Emerg Infect Dis. 2013;19:331–3.CrossRefPubMedPubMedCentral Hoa NT, Chieu TT, Do Dung S, Long NT, Hieu TQ, Luc NT, Nhuong PT, Huong VT, Trinh DT, Wertheim HF, et al. Streptococcus suis and porcine reproductive and respiratory syndrome, Vietnam. Emerg Infect Dis. 2013;19:331–3.CrossRefPubMedPubMedCentral
4.
go back to reference Wang J, Kong D, Zhang S, Jiang H, Zheng Y, Zang Y, Hao H, Jiang Y. Interaction of fibrinogen and muramidase-released protein promotes the development of Streptococcus suis meningitis. Front Microbiol. 2015;6:1001.PubMedPubMedCentral Wang J, Kong D, Zhang S, Jiang H, Zheng Y, Zang Y, Hao H, Jiang Y. Interaction of fibrinogen and muramidase-released protein promotes the development of Streptococcus suis meningitis. Front Microbiol. 2015;6:1001.PubMedPubMedCentral
5.
go back to reference Takeuchi D, Akeda Y, Nakayama T, Kerdsin A, Sano Y, Kanda T, Hamada S, Dejsirilert S, Oishi K. The contribution of suilysin to the pathogenesis of Streptococcus suis meningitis. J Infect Dis. 2014;209:1509–19.CrossRefPubMed Takeuchi D, Akeda Y, Nakayama T, Kerdsin A, Sano Y, Kanda T, Hamada S, Dejsirilert S, Oishi K. The contribution of suilysin to the pathogenesis of Streptococcus suis meningitis. J Infect Dis. 2014;209:1509–19.CrossRefPubMed
6.
go back to reference Wu Z, Wang W, Tang M, Shao J, Dai C, Zhang W, Fan H, Yao H, Zong J, Chen D, et al. Comparative genomic analysis shows that Streptococcus suis meningitis isolate SC070731 contains a unique 105 K genomic island. Gene. 2014;535:156–64.CrossRefPubMed Wu Z, Wang W, Tang M, Shao J, Dai C, Zhang W, Fan H, Yao H, Zong J, Chen D, et al. Comparative genomic analysis shows that Streptococcus suis meningitis isolate SC070731 contains a unique 105 K genomic island. Gene. 2014;535:156–64.CrossRefPubMed
7.
go back to reference Dominguez-Punaro Mde L, Segura M, Contreras I, Lachance C, Houde M, Lecours MP, Olivier M, Gottschalk M. In vitro characterization of the microglial inflammatory response to Streptococcus suis, an important emerging zoonotic agent of meningitis. Infect Immun. 2010;78:5074–85.CrossRefPubMed Dominguez-Punaro Mde L, Segura M, Contreras I, Lachance C, Houde M, Lecours MP, Olivier M, Gottschalk M. In vitro characterization of the microglial inflammatory response to Streptococcus suis, an important emerging zoonotic agent of meningitis. Infect Immun. 2010;78:5074–85.CrossRefPubMed
8.
go back to reference Mai NT, Hoa NT, Nga TV, le Linh D, Chau TT, Sinh DX, Phu NH, Chuong LV, Diep TS, Campbell J, et al. Streptococcus suis meningitis in adults in Vietnam. Clin Infect Dis. 2008;46:659–67.CrossRefPubMed Mai NT, Hoa NT, Nga TV, le Linh D, Chau TT, Sinh DX, Phu NH, Chuong LV, Diep TS, Campbell J, et al. Streptococcus suis meningitis in adults in Vietnam. Clin Infect Dis. 2008;46:659–67.CrossRefPubMed
9.
go back to reference Dominguez-Punaro MC, Segura M, Plante MM, Lacouture S, Rivest S, Gottschalk M. Streptococcus suis serotype 2, an important swine and human pathogen, induces strong systemic and cerebral inflammatory responses in a mouse model of infection. J Immunol. 2007;179:1842–54.CrossRefPubMed Dominguez-Punaro MC, Segura M, Plante MM, Lacouture S, Rivest S, Gottschalk M. Streptococcus suis serotype 2, an important swine and human pathogen, induces strong systemic and cerebral inflammatory responses in a mouse model of infection. J Immunol. 2007;179:1842–54.CrossRefPubMed
10.
go back to reference Segura M, Vanier G, Al-Numani D, Lacouture S, Olivier M, Gottschalk M. Proinflammatory cytokine and chemokine modulation by Streptococcus suis in a whole-blood culture system. FEMS Immunol Med Microbiol. 2006;47:92–106.CrossRefPubMed Segura M, Vanier G, Al-Numani D, Lacouture S, Olivier M, Gottschalk M. Proinflammatory cytokine and chemokine modulation by Streptococcus suis in a whole-blood culture system. FEMS Immunol Med Microbiol. 2006;47:92–106.CrossRefPubMed
11.
go back to reference Lv Q, Hao H, Bi L, Zheng Y, Zhou X, Jiang Y. Suilysin remodels the cytoskeletons of human brain microvascular endothelial cells by activating RhoA and Rac1 GTPase. Protein Cell. 2014;5:261–4.CrossRefPubMedPubMedCentral Lv Q, Hao H, Bi L, Zheng Y, Zhou X, Jiang Y. Suilysin remodels the cytoskeletons of human brain microvascular endothelial cells by activating RhoA and Rac1 GTPase. Protein Cell. 2014;5:261–4.CrossRefPubMedPubMedCentral
12.
go back to reference Bonifait L, Grenier D. The SspA subtilisin-like protease of Streptococcus suis triggers a pro-inflammatory response in macrophages through a non-proteolytic mechanism. BMC Microbiol. 2011;11:47.CrossRefPubMedPubMedCentral Bonifait L, Grenier D. The SspA subtilisin-like protease of Streptococcus suis triggers a pro-inflammatory response in macrophages through a non-proteolytic mechanism. BMC Microbiol. 2011;11:47.CrossRefPubMedPubMedCentral
13.
go back to reference Yan F, Cao H, Chaturvedi R, Krishna U, Hobbs SS, Dempsey PJ, Peek Jr RM, Cover TL, Washington MK, Wilson KT, Polk DB. Epidermal growth factor receptor activation protects gastric epithelial cells from Helicobacter pylori-induced apoptosis. Gastroenterology. 2009;136:1297–307. e1291-1293.CrossRefPubMedPubMedCentral Yan F, Cao H, Chaturvedi R, Krishna U, Hobbs SS, Dempsey PJ, Peek Jr RM, Cover TL, Washington MK, Wilson KT, Polk DB. Epidermal growth factor receptor activation protects gastric epithelial cells from Helicobacter pylori-induced apoptosis. Gastroenterology. 2009;136:1297–307. e1291-1293.CrossRefPubMedPubMedCentral
14.
go back to reference Zhang J, Li H, Wang J, Dong Z, Mian S, Yu FS. Role of EGFR transactivation in preventing apoptosis in Pseudomonas aeruginosa-infected human corneal epithelial cells. Invest Ophthalmol Vis Sci. 2004;45:2569–76.CrossRefPubMedPubMedCentral Zhang J, Li H, Wang J, Dong Z, Mian S, Yu FS. Role of EGFR transactivation in preventing apoptosis in Pseudomonas aeruginosa-infected human corneal epithelial cells. Invest Ophthalmol Vis Sci. 2004;45:2569–76.CrossRefPubMedPubMedCentral
15.
17.
go back to reference Wang X, Huong SM, Chiu ML, Raab-Traub N, Huang ES. Epidermal growth factor receptor is a cellular receptor for human cytomegalovirus. Nature. 2003;424:456–61.CrossRefPubMed Wang X, Huong SM, Chiu ML, Raab-Traub N, Huang ES. Epidermal growth factor receptor is a cellular receptor for human cytomegalovirus. Nature. 2003;424:456–61.CrossRefPubMed
18.
go back to reference Tao RH, Maruyama IN. All EGF(ErbB) receptors have preformed homo- and heterodimeric structures in living cells. J Cell Sci. 2008;121:3207–17.CrossRefPubMed Tao RH, Maruyama IN. All EGF(ErbB) receptors have preformed homo- and heterodimeric structures in living cells. J Cell Sci. 2008;121:3207–17.CrossRefPubMed
19.
go back to reference Hoffmann I, Eugene E, Nassif X, Couraud PO, Bourdoulous S. Activation of ErbB2 receptor tyrosine kinase supports invasion of endothelial cells by Neisseria meningitidis. J Cell Biol. 2001;155:133–43.CrossRefPubMedPubMedCentral Hoffmann I, Eugene E, Nassif X, Couraud PO, Bourdoulous S. Activation of ErbB2 receptor tyrosine kinase supports invasion of endothelial cells by Neisseria meningitidis. J Cell Biol. 2001;155:133–43.CrossRefPubMedPubMedCentral
20.
go back to reference Yarden Y, Pines G. The ERBB network: at last, cancer therapy meets systems biology. Nat Rev Cancer. 2012;12:553–63.CrossRefPubMed Yarden Y, Pines G. The ERBB network: at last, cancer therapy meets systems biology. Nat Rev Cancer. 2012;12:553–63.CrossRefPubMed
21.
go back to reference Feigin ME, Muthuswamy SK. ErbB receptors and cell polarity: new pathways and paradigms for understanding cell migration and invasion. Exp Cell Res. 2009;315:707–16.CrossRefPubMed Feigin ME, Muthuswamy SK. ErbB receptors and cell polarity: new pathways and paradigms for understanding cell migration and invasion. Exp Cell Res. 2009;315:707–16.CrossRefPubMed
22.
go back to reference Swanson KV, Griffiss JM, Edwards VL, Stein DC, Song W. Neisseria gonorrhoeae-induced transactivation of EGFR enhances gonococcal invasion. Cell Microbiol. 2011;13:1078–90.CrossRefPubMed Swanson KV, Griffiss JM, Edwards VL, Stein DC, Song W. Neisseria gonorrhoeae-induced transactivation of EGFR enhances gonococcal invasion. Cell Microbiol. 2011;13:1078–90.CrossRefPubMed
23.
go back to reference Frank CG, Reguerio V, Rother M, Moranta D, Maeurer AP, Garmendia J, Meyer TF, Bengoechea JA. Klebsiella pneumoniae targets an EGF receptor-dependent pathway to subvert inflammation. Cell Microbiol. 2013;15:1212–33.CrossRefPubMed Frank CG, Reguerio V, Rother M, Moranta D, Maeurer AP, Garmendia J, Meyer TF, Bengoechea JA. Klebsiella pneumoniae targets an EGF receptor-dependent pathway to subvert inflammation. Cell Microbiol. 2013;15:1212–33.CrossRefPubMed
24.
go back to reference Mikami F, Gu H, Jono H, Andalibi A, Kai H, Li JD. Epidermal growth factor receptor acts as a negative regulator for bacterium nontypeable Haemophilus influenzae-induced toll-like receptor 2 expression via an Src-dependent p38 mitogen-activated protein kinase signaling pathway. J Biol Chem. 2005;280:36185–94.CrossRefPubMed Mikami F, Gu H, Jono H, Andalibi A, Kai H, Li JD. Epidermal growth factor receptor acts as a negative regulator for bacterium nontypeable Haemophilus influenzae-induced toll-like receptor 2 expression via an Src-dependent p38 mitogen-activated protein kinase signaling pathway. J Biol Chem. 2005;280:36185–94.CrossRefPubMed
25.
go back to reference Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37:13–25.CrossRefPubMed Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37:13–25.CrossRefPubMed
26.
go back to reference Zhang Y, Ding D, Liu M, Yang X, Zong B, Wang X, Chen H, Bei W, Tan C. Effect of the glycosyltransferases on the capsular polysaccharide synthesis of Streptococcus suis serotype 2. Microbiol Res. 2016;185:45–54.CrossRefPubMed Zhang Y, Ding D, Liu M, Yang X, Zong B, Wang X, Chen H, Bei W, Tan C. Effect of the glycosyltransferases on the capsular polysaccharide synthesis of Streptococcus suis serotype 2. Microbiol Res. 2016;185:45–54.CrossRefPubMed
27.
go back to reference Stins MF, Gilles F, Kim KS. Selective expression of adhesion molecules on human brain microvascular endothelial cells. J Neuroimmunol. 1997;76:81–90.CrossRefPubMed Stins MF, Gilles F, Kim KS. Selective expression of adhesion molecules on human brain microvascular endothelial cells. J Neuroimmunol. 1997;76:81–90.CrossRefPubMed
28.
go back to reference Stins MF, Badger J, Sik Kim K. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb Pathog. 2001;30:19–28.CrossRefPubMed Stins MF, Badger J, Sik Kim K. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb Pathog. 2001;30:19–28.CrossRefPubMed
29.
go back to reference Wang Y, Kim KS. Role of OmpA and IbeB in Escherichia coli K1 invasion of brain microvascular endothelial cells in vitro and in vivo. Pediatr Res. 2002;51:559–63.CrossRefPubMed Wang Y, Kim KS. Role of OmpA and IbeB in Escherichia coli K1 invasion of brain microvascular endothelial cells in vitro and in vivo. Pediatr Res. 2002;51:559–63.CrossRefPubMed
30.
go back to reference Benga L, Fulde M, Neis C, Goethe R, Valentin-Weigand P. Polysaccharide capsule and suilysin contribute to extracellular survival of Streptococcus suis co-cultivated with primary porcine phagocytes. Vet Microbiol. 2008;132:211–9.CrossRefPubMed Benga L, Fulde M, Neis C, Goethe R, Valentin-Weigand P. Polysaccharide capsule and suilysin contribute to extracellular survival of Streptococcus suis co-cultivated with primary porcine phagocytes. Vet Microbiol. 2008;132:211–9.CrossRefPubMed
31.
go back to reference Shin S, Paul-Satyaseela M, Lee JS, Romer LH, Kim KS. Focal adhesion kinase is involved in type III group B streptococcal invasion of human brain microvascular endothelial cells. Microb Pathog. 2006;41:168–73.CrossRefPubMed Shin S, Paul-Satyaseela M, Lee JS, Romer LH, Kim KS. Focal adhesion kinase is involved in type III group B streptococcal invasion of human brain microvascular endothelial cells. Microb Pathog. 2006;41:168–73.CrossRefPubMed
32.
go back to reference Zhu L, Maruvada R, Sapirstein A, Malik KU, Peters-Golden M, Kim KS. Arachidonic acid metabolism regulates Escherichia coli penetration of the blood–brain barrier. Infect Immun. 2010;78:4302–10.CrossRefPubMedPubMedCentral Zhu L, Maruvada R, Sapirstein A, Malik KU, Peters-Golden M, Kim KS. Arachidonic acid metabolism regulates Escherichia coli penetration of the blood–brain barrier. Infect Immun. 2010;78:4302–10.CrossRefPubMedPubMedCentral
33.
go back to reference Yan X, Prosniak M, Curtis MT, Weiss ML, Faber M, Dietzschold B, Fu ZF. Silver-haired bat rabies virus variant does not induce apoptosis in the brain of experimentally infected mice. J Neurovirol. 2001;7:518–27.CrossRefPubMed Yan X, Prosniak M, Curtis MT, Weiss ML, Faber M, Dietzschold B, Fu ZF. Silver-haired bat rabies virus variant does not induce apoptosis in the brain of experimentally infected mice. J Neurovirol. 2001;7:518–27.CrossRefPubMed
34.
go back to reference Charland N, Nizet V, Rubens CE, Kim KS, Lacouture S, Gottschalk M. Streptococcus suis serotype 2 interactions with human brain microvascular endothelial cells. Infect Immun. 2000;68:637–43.CrossRefPubMedPubMedCentral Charland N, Nizet V, Rubens CE, Kim KS, Lacouture S, Gottschalk M. Streptococcus suis serotype 2 interactions with human brain microvascular endothelial cells. Infect Immun. 2000;68:637–43.CrossRefPubMedPubMedCentral
35.
go back to reference Finlay BB, Cossart P. Exploitation of mammalian host cell functions by bacterial pathogens. Science. 1997;276:718–25.CrossRefPubMed Finlay BB, Cossart P. Exploitation of mammalian host cell functions by bacterial pathogens. Science. 1997;276:718–25.CrossRefPubMed
37.
go back to reference Graus-Porta D, Beerli RR, Daly JM, Hynes NE. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J. 1997;16:1647–55.CrossRefPubMedPubMedCentral Graus-Porta D, Beerli RR, Daly JM, Hynes NE. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J. 1997;16:1647–55.CrossRefPubMedPubMedCentral
38.
go back to reference Higashiyama S, Iwabuki H, Morimoto C, Hieda M, Inoue H, Matsushita N. Membrane-anchored growth factors, the epidermal growth factor family: beyond receptor ligands. Cancer Sci. 2008;99:214–20.CrossRefPubMed Higashiyama S, Iwabuki H, Morimoto C, Hieda M, Inoue H, Matsushita N. Membrane-anchored growth factors, the epidermal growth factor family: beyond receptor ligands. Cancer Sci. 2008;99:214–20.CrossRefPubMed
39.
go back to reference Blobel CP. ADAMs: key components in EGFR signalling and development. Nat Rev Mol Cell Biol. 2005;6:32–43.CrossRefPubMed Blobel CP. ADAMs: key components in EGFR signalling and development. Nat Rev Mol Cell Biol. 2005;6:32–43.CrossRefPubMed
40.
go back to reference Segura M, Vadeboncoeur N, Gottschalk M. CD14-dependent and -independent cytokine and chemokine production by human THP-1 monocytes stimulated by Streptococcus suis capsular type 2. Clin Exp Immunol. 2002;127:243–54.CrossRefPubMedPubMedCentral Segura M, Vadeboncoeur N, Gottschalk M. CD14-dependent and -independent cytokine and chemokine production by human THP-1 monocytes stimulated by Streptococcus suis capsular type 2. Clin Exp Immunol. 2002;127:243–54.CrossRefPubMedPubMedCentral
41.
go back to reference Graveline R, Segura M, Radzioch D, Gottschalk M. TLR2-dependent recognition of Streptococcus suis is modulated by the presence of capsular polysaccharide which modifies macrophage responsiveness. Int Immunol. 2007;19:375–89.CrossRefPubMed Graveline R, Segura M, Radzioch D, Gottschalk M. TLR2-dependent recognition of Streptococcus suis is modulated by the presence of capsular polysaccharide which modifies macrophage responsiveness. Int Immunol. 2007;19:375–89.CrossRefPubMed
42.
go back to reference Hsieh HL, Sun CC, Wu CB, Wu CY, Tung WH, Wang HH, Yang CM. Sphingosine 1-phosphate induces EGFR expression via Akt/NF-kappa B and ERK/AP-1 pathways in rat vascular smooth muscle cells. J Cell Biochem. 2008;103:1732–46.CrossRefPubMed Hsieh HL, Sun CC, Wu CB, Wu CY, Tung WH, Wang HH, Yang CM. Sphingosine 1-phosphate induces EGFR expression via Akt/NF-kappa B and ERK/AP-1 pathways in rat vascular smooth muscle cells. J Cell Biochem. 2008;103:1732–46.CrossRefPubMed
43.
go back to reference Lun ZR, Wang QP, Chen XG, Li AX, Zhu XQ. Streptococcus suis: an emerging zoonotic pathogen. Lancet Infect Dis. 2007;7:201–9.CrossRefPubMed Lun ZR, Wang QP, Chen XG, Li AX, Zhu XQ. Streptococcus suis: an emerging zoonotic pathogen. Lancet Infect Dis. 2007;7:201–9.CrossRefPubMed
44.
go back to reference Auger JP, Christodoulides M, Segura M, Xu J, Gottschalk M. Interactions of Streptococcus suis serotype 2 with human meningeal cells and astrocytes. BMC Res Notes. 2015;8:607.CrossRefPubMedPubMedCentral Auger JP, Christodoulides M, Segura M, Xu J, Gottschalk M. Interactions of Streptococcus suis serotype 2 with human meningeal cells and astrocytes. BMC Res Notes. 2015;8:607.CrossRefPubMedPubMedCentral
45.
go back to reference Sun Y, Li N, Zhang J, Liu H, Liu J, Xia X, Sun C, Feng X, Gu J, Du C, et al. Enolase of Streptococcus suis serotype 2 enhances blood-brain barrier permeability by inducing IL-8 release. Inflammation. 2016;39:718–26.CrossRefPubMed Sun Y, Li N, Zhang J, Liu H, Liu J, Xia X, Sun C, Feng X, Gu J, Du C, et al. Enolase of Streptococcus suis serotype 2 enhances blood-brain barrier permeability by inducing IL-8 release. Inflammation. 2016;39:718–26.CrossRefPubMed
46.
go back to reference Kim KS. Mechanisms of microbial traversal of the blood-brain barrier. Nat Rev Microbiol. 2008;6:625–34.CrossRefPubMed Kim KS. Mechanisms of microbial traversal of the blood-brain barrier. Nat Rev Microbiol. 2008;6:625–34.CrossRefPubMed
47.
go back to reference Gottschalk M, Xu J, Calzas C, Segura M. Streptococcus suis: a new emerging or an old neglected zoonotic pathogen? Future Microbiol. 2010;5:371–91.CrossRefPubMed Gottschalk M, Xu J, Calzas C, Segura M. Streptococcus suis: a new emerging or an old neglected zoonotic pathogen? Future Microbiol. 2010;5:371–91.CrossRefPubMed
48.
go back to reference Vadeboncoeur N, Segura M, Al-Numani D, Vanier G, Gottschalk M. Pro-inflammatory cytokine and chemokine release by human brain microvascular endothelial cells stimulated by Streptococcus suis serotype 2. FEMS Immunol Med Microbiol. 2003;35:49–58.CrossRefPubMed Vadeboncoeur N, Segura M, Al-Numani D, Vanier G, Gottschalk M. Pro-inflammatory cytokine and chemokine release by human brain microvascular endothelial cells stimulated by Streptococcus suis serotype 2. FEMS Immunol Med Microbiol. 2003;35:49–58.CrossRefPubMed
49.
go back to reference Mook-Kanamori BB, Geldhoff M, van der Poll T, van de Beek D. Pathogenesis and pathophysiology of Pneumococcal meningitis. Clin Microbiol Rev. 2011;24:557–91.CrossRefPubMedPubMedCentral Mook-Kanamori BB, Geldhoff M, van der Poll T, van de Beek D. Pathogenesis and pathophysiology of Pneumococcal meningitis. Clin Microbiol Rev. 2011;24:557–91.CrossRefPubMedPubMedCentral
50.
go back to reference Doran KS, Fulde M, Gratz N, Kim BJ, Nau R, Prasadarao N, Schubert-Unkmeir A, Tuomanen EI, Valentin-Weigand P. Host-pathogen interactions in bacterial meningitis. Acta Neuropathol. 2016;131:185–209.CrossRefPubMedPubMedCentral Doran KS, Fulde M, Gratz N, Kim BJ, Nau R, Prasadarao N, Schubert-Unkmeir A, Tuomanen EI, Valentin-Weigand P. Host-pathogen interactions in bacterial meningitis. Acta Neuropathol. 2016;131:185–209.CrossRefPubMedPubMedCentral
51.
go back to reference Dominguez-Punaro Mde L, Segura M, Radzioch D, Rivest S, Gottschalk M. Comparison of the susceptibilities of C57BL/6 and A/J mouse strains to Streptococcus suis serotype 2 infection. Infect Immun. 2008;76:3901–10.CrossRefPubMed Dominguez-Punaro Mde L, Segura M, Radzioch D, Rivest S, Gottschalk M. Comparison of the susceptibilities of C57BL/6 and A/J mouse strains to Streptococcus suis serotype 2 infection. Infect Immun. 2008;76:3901–10.CrossRefPubMed
52.
go back to reference Suzuki T, Yoshinaga N, Tanabe S. Interleukin-6 (IL-6) regulates claudin-2 expression and tight junction permeability in intestinal epithelium. J Biol Chem. 2011;286:31263–71.CrossRefPubMedPubMedCentral Suzuki T, Yoshinaga N, Tanabe S. Interleukin-6 (IL-6) regulates claudin-2 expression and tight junction permeability in intestinal epithelium. J Biol Chem. 2011;286:31263–71.CrossRefPubMedPubMedCentral
53.
go back to reference Song L, Pachter JS. Monocyte chemoattractant protein-1 alters expression of tight junction-associated proteins in brain microvascular endothelial cells. Microvasc Res. 2004;67:78–89.CrossRefPubMed Song L, Pachter JS. Monocyte chemoattractant protein-1 alters expression of tight junction-associated proteins in brain microvascular endothelial cells. Microvasc Res. 2004;67:78–89.CrossRefPubMed
54.
go back to reference Stamatovic SM, Keep RF, Kunkel SL, Andjelkovic AV. Potential role of MCP-1 in endothelial cell tight junction ‘opening’: signaling via Rho and Rho kinase. J Cell Sci. 2003;116:4615–28.CrossRefPubMed Stamatovic SM, Keep RF, Kunkel SL, Andjelkovic AV. Potential role of MCP-1 in endothelial cell tight junction ‘opening’: signaling via Rho and Rho kinase. J Cell Sci. 2003;116:4615–28.CrossRefPubMed
55.
go back to reference Sawyer AJ, Tian W, Saucier-Sawyer JK, Rizk PJ, Saltzman WM, Bellamkonda RV, Kyriakides TR. The effect of inflammatory cell-derived MCP-1 loss on neuronal survival during chronic neuroinflammation. Biomaterials. 2014;35:6698–706.CrossRefPubMedPubMedCentral Sawyer AJ, Tian W, Saucier-Sawyer JK, Rizk PJ, Saltzman WM, Bellamkonda RV, Kyriakides TR. The effect of inflammatory cell-derived MCP-1 loss on neuronal survival during chronic neuroinflammation. Biomaterials. 2014;35:6698–706.CrossRefPubMedPubMedCentral
56.
go back to reference Shim JW, Sandlund J, Hameed MQ, Blazer-Yost B, Zhou FC, Klagsbrun M, Madsen JR. Excess HB-EGF, which promotes VEGF signaling, leads to hydrocephalus. Sci Rep. 2016;6:26794.CrossRefPubMedPubMedCentral Shim JW, Sandlund J, Hameed MQ, Blazer-Yost B, Zhou FC, Klagsbrun M, Madsen JR. Excess HB-EGF, which promotes VEGF signaling, leads to hydrocephalus. Sci Rep. 2016;6:26794.CrossRefPubMedPubMedCentral
57.
go back to reference Sugiyama S, Nakabayashi K, Baba I, Sasazuki T, Shirasawa S. Role of epiregulin in peptidoglycan-induced proinflammatory cytokine production by antigen presenting cells. Biochem Biophys Res Commun. 2005;337:271–4.CrossRefPubMed Sugiyama S, Nakabayashi K, Baba I, Sasazuki T, Shirasawa S. Role of epiregulin in peptidoglycan-induced proinflammatory cytokine production by antigen presenting cells. Biochem Biophys Res Commun. 2005;337:271–4.CrossRefPubMed
58.
go back to reference Val S, Belade E, George I, Boczkowski J, Baeza-Squiban A. Fine PM induce airway MUC5AC expression through the autocrine effect of amphiregulin. Arch Toxicol. 2012;86:1851–9.CrossRefPubMed Val S, Belade E, George I, Boczkowski J, Baeza-Squiban A. Fine PM induce airway MUC5AC expression through the autocrine effect of amphiregulin. Arch Toxicol. 2012;86:1851–9.CrossRefPubMed
59.
go back to reference Taylor SR, Markesbery MG, Harding PA. Heparin-binding epidermal growth factor-like growth factor (HB-EGF) and proteolytic processing by a disintegrin and metalloproteinases (ADAM): a regulator of several pathways. Semin Cell Dev Biol. 2014;28:22–30.CrossRefPubMed Taylor SR, Markesbery MG, Harding PA. Heparin-binding epidermal growth factor-like growth factor (HB-EGF) and proteolytic processing by a disintegrin and metalloproteinases (ADAM): a regulator of several pathways. Semin Cell Dev Biol. 2014;28:22–30.CrossRefPubMed
60.
go back to reference Edwards VL, Wang LC, Dawson V, Stein DC, Song W. Neisseria gonorrhoeae breaches the apical junction of polarized epithelial cells for transmigration by activating EGFR. Cell Microbiol. 2013;15:1042–57.CrossRefPubMed Edwards VL, Wang LC, Dawson V, Stein DC, Song W. Neisseria gonorrhoeae breaches the apical junction of polarized epithelial cells for transmigration by activating EGFR. Cell Microbiol. 2013;15:1042–57.CrossRefPubMed
Metadata
Title
EGFR transactivation contributes to neuroinflammation in Streptococcus suis meningitis
Authors
Xiao-Pei Yang
Ji-Yang Fu
Rui-Cheng Yang
Wen-Tong Liu
Tao Zhang
Bo Yang
Ling Miao
Bei-Bei Dou
Chen Tan
Huan-Chun Chen
Xiang-Ru Wang
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0734-0

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue