Skip to main content
Top
Published in: Allergy, Asthma & Clinical Immunology 1/2018

Open Access 01-12-2018 | Research

A bronchoprotective role for Rgs2 in a murine model of lipopolysaccharide-induced airways inflammation

Authors: Tresa George, Mainak Chakraborty, Mark A. Giembycz, Robert Newton

Published in: Allergy, Asthma & Clinical Immunology | Issue 1/2018

Login to get access

Abstract

Background

Asthma exacerbations are associated with the recruitment of neutrophils to the lungs. These cells release proteases and mediators, many of which act at G protein-coupled receptors (GPCRs) that couple via Gq to promote bronchoconstriction and inflammation. Common asthma therapeutics up-regulate expression of the regulator of G protein signalling (RGS), RGS2. As RGS2 reduces signaling from Gq-coupled GPCRs, we have defined role(s) for this GTPase-activating protein in an acute neutrophilic model of lung inflammation.

Methods

Wild type and Rgs2/ C57Bl6 mice were exposed to nebulized lipopolysaccharide (LPS). Lung function (respiratory system resistance and compliance) was measured using a SCIREQ flexivent small animal ventilator. Lung inflammation was assessed by histochemistry, cell counting and by cytokine and chemokine expression in bronchoalveolar lavage (BAL) fluid.

Results

Lipopolysaccharide inhalation induced transient airways hyperreactivity (AHR) and neutrophilic lung inflammation. While AHR and inflammation was greatest 3 h post-LPS exposure, BAL neutrophils persisted for 24 h. At 3 h post-LPS inhalation, multiple inflammatory cytokines (CSF2, CSF3, IL6, TNF) and chemokines (CCL3, CCL4, CXCL1, CXCL2) were highly expressed in the BAL fluid, prior to declining by 24 h. Compared to wild type counterparts, Rgs2/ mice developed significantly greater airflow resistance in response to inhaled methacholine (MCh) at 3 h post-LPS exposure. At 24 h post-LPS exposure, when lung function was recovering in the wild type animals, MCh-induced resistance was increased, and compliance decreased, in Rgs2/ mice. Thus, Rgs2/ mice show AHR and stiffer lungs 24 h post-LPS exposure. Histological markers of inflammation, total and differential cell counts, and major cytokine and chemokine expression in BAL fluid were similar between wild type and Rgs2/ mice. However, 3 and 24 h post-LPS exposure, IL12B expression was significantly elevated in BAL fluid from Rgs2/ mice compared to wild type animals.

Conclusions

While Rgs2 is bronchoprotective in acute neutrophilic inflammation, no clear anti-inflammatory effect was apparent. Nevertheless, elevated IL12B expression in Rgs2/ animals raises the possibility that RGS2 could dampen Th1 responses. These findings indicate that up-regulation of RGS2, as occurs in response to inhaled corticosteroids and long-acting β2-adrenoceptor agonists, may be beneficial in acute neutrophilic exacerbations of airway disease, including asthma.
Appendix
Available only for authorised users
Literature
1.
go back to reference Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. 2008;8:183–92.CrossRef Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. 2008;8:183–92.CrossRef
2.
go back to reference Penn RB, Benovic JL. Regulation of heterotrimeric G protein signaling in airway smooth muscle. Proc Am Thorac Soc. 2008;5:47–57.CrossRef Penn RB, Benovic JL. Regulation of heterotrimeric G protein signaling in airway smooth muscle. Proc Am Thorac Soc. 2008;5:47–57.CrossRef
3.
go back to reference Ammit AJ, Lazaar AL, Irani C, O’Neill GM, Gordon ND, Amrani Y, Penn RB, Panettieri RA Jr. Tumor necrosis factor-alpha-induced secretion of RANTES and interleukin-6 from human airway smooth muscle cells: modulation by glucocorticoids and beta-agonists. Am J Respir Cell Mol Biol. 2002;26:465–74.CrossRef Ammit AJ, Lazaar AL, Irani C, O’Neill GM, Gordon ND, Amrani Y, Penn RB, Panettieri RA Jr. Tumor necrosis factor-alpha-induced secretion of RANTES and interleukin-6 from human airway smooth muscle cells: modulation by glucocorticoids and beta-agonists. Am J Respir Cell Mol Biol. 2002;26:465–74.CrossRef
4.
go back to reference Holden NS, Rider CF, Bell MJ, Velayudhan J, King EM, Kaur M, Salmon M, Giembycz MA, Newton R. Enhancement of inflammatory mediator release by beta(2)-adrenoceptor agonists in airway epithelial cells is reversed by glucocorticoid action. Br J Pharmacol. 2010;160:410–20.CrossRef Holden NS, Rider CF, Bell MJ, Velayudhan J, King EM, Kaur M, Salmon M, Giembycz MA, Newton R. Enhancement of inflammatory mediator release by beta(2)-adrenoceptor agonists in airway epithelial cells is reversed by glucocorticoid action. Br J Pharmacol. 2010;160:410–20.CrossRef
5.
go back to reference Jacquet A. The role of innate immunity activation in house dust mite allergy. Trends Mol Med. 2011;17:604–11.CrossRef Jacquet A. The role of innate immunity activation in house dust mite allergy. Trends Mol Med. 2011;17:604–11.CrossRef
6.
go back to reference Gregory LG, Lloyd CM. Orchestrating house dust mite-associated allergy in the lung. Trends Immunol. 2011;32:402–11.CrossRef Gregory LG, Lloyd CM. Orchestrating house dust mite-associated allergy in the lung. Trends Immunol. 2011;32:402–11.CrossRef
7.
go back to reference Ramachandran R, Altier C, Oikonomopoulou K, Hollenberg MD. Proteinases, their extracellular targets, and inflammatory signaling. Pharmacol Rev. 2016;68:1110–42.CrossRef Ramachandran R, Altier C, Oikonomopoulou K, Hollenberg MD. Proteinases, their extracellular targets, and inflammatory signaling. Pharmacol Rev. 2016;68:1110–42.CrossRef
8.
go back to reference Johnson JR, Wiley RE, Fattouh R, Swirski FK, Gajewska BU, Coyle AJ, Gutierrez-Ramos JC, Ellis R, Inman MD, Jordana M. Continuous exposure to house dust mite elicits chronic airway inflammation and structural remodeling. Am J Respir Crit Care Med. 2004;169:378–85.CrossRef Johnson JR, Wiley RE, Fattouh R, Swirski FK, Gajewska BU, Coyle AJ, Gutierrez-Ramos JC, Ellis R, Inman MD, Jordana M. Continuous exposure to house dust mite elicits chronic airway inflammation and structural remodeling. Am J Respir Crit Care Med. 2004;169:378–85.CrossRef
9.
go back to reference Southam DS, Ellis R, Wattie J, Inman MD. Components of airway hyperresponsiveness and their associations with inflammation and remodeling in mice. J Allergy Clin Immunol. 2007;119:848–54.CrossRef Southam DS, Ellis R, Wattie J, Inman MD. Components of airway hyperresponsiveness and their associations with inflammation and remodeling in mice. J Allergy Clin Immunol. 2007;119:848–54.CrossRef
10.
go back to reference Gregory LG, Causton B, Murdoch JR, Mathie SA, O’Donnell V, Thomas CP, Priest FM, Quint DJ, Lloyd CM. Inhaled house dust mite induces pulmonary T helper 2 cytokine production. Clin Exp Allergy. 2009;39:1597–610.CrossRef Gregory LG, Causton B, Murdoch JR, Mathie SA, O’Donnell V, Thomas CP, Priest FM, Quint DJ, Lloyd CM. Inhaled house dust mite induces pulmonary T helper 2 cytokine production. Clin Exp Allergy. 2009;39:1597–610.CrossRef
12.
go back to reference Asokananthan N, Graham PT, Stewart DJ, Bakker AJ, Eidne KA, Thompson PJ, Stewart GA. House dust mite allergens induce proinflammatory cytokines from respiratory epithelial cells: the cysteine protease allergen, Der p 1, activates protease-activated receptor (PAR)-2 and inactivates PAR-1. J Immunol. 2002;169:4572–8.CrossRef Asokananthan N, Graham PT, Stewart DJ, Bakker AJ, Eidne KA, Thompson PJ, Stewart GA. House dust mite allergens induce proinflammatory cytokines from respiratory epithelial cells: the cysteine protease allergen, Der p 1, activates protease-activated receptor (PAR)-2 and inactivates PAR-1. J Immunol. 2002;169:4572–8.CrossRef
13.
go back to reference Ebeling C, Lam T, Gordon JR, Hollenberg MD, Vliagoftis H. Proteinase-activated receptor-2 promotes allergic sensitization to an inhaled antigen through a TNF-mediated pathway. J Immunol. 2007;179:2910–7.CrossRef Ebeling C, Lam T, Gordon JR, Hollenberg MD, Vliagoftis H. Proteinase-activated receptor-2 promotes allergic sensitization to an inhaled antigen through a TNF-mediated pathway. J Immunol. 2007;179:2910–7.CrossRef
14.
go back to reference Arizmendi NG, Abel M, Mihara K, Davidson C, Polley D, Nadeem A, El MT, Gilmore BF, Walker B, Gordon JR, Hollenberg MD, Vliagoftis H. Mucosal allergic sensitization to cockroach allergens is dependent on proteinase activity and proteinase-activated receptor-2 activation. J Immunol. 2011;186:3164–72.CrossRef Arizmendi NG, Abel M, Mihara K, Davidson C, Polley D, Nadeem A, El MT, Gilmore BF, Walker B, Gordon JR, Hollenberg MD, Vliagoftis H. Mucosal allergic sensitization to cockroach allergens is dependent on proteinase activity and proteinase-activated receptor-2 activation. J Immunol. 2011;186:3164–72.CrossRef
15.
go back to reference Davidson CE, Asaduzzaman M, Arizmendi NG, Polley D, Wu Y, Gordon JR, Hollenberg MD, Cameron L, Vliagoftis H. Proteinase-activated receptor-2 activation participates in allergic sensitization to house dust mite allergens in a murine model. Clin Exp Allergy. 2013;43:1274–85.CrossRef Davidson CE, Asaduzzaman M, Arizmendi NG, Polley D, Wu Y, Gordon JR, Hollenberg MD, Cameron L, Vliagoftis H. Proteinase-activated receptor-2 activation participates in allergic sensitization to house dust mite allergens in a murine model. Clin Exp Allergy. 2013;43:1274–85.CrossRef
16.
go back to reference Schuliga M, Royce SG, Langenbach S, Berhan A, Harris T, Keenan CR, Stewart AG. The coagulant factor Xa induces protease-activated receptor-1 and annexin A2-dependent airway smooth muscle cytokine production and cell proliferation. Am J Respir Cell Mol Biol. 2016;54:200–9.CrossRef Schuliga M, Royce SG, Langenbach S, Berhan A, Harris T, Keenan CR, Stewart AG. The coagulant factor Xa induces protease-activated receptor-1 and annexin A2-dependent airway smooth muscle cytokine production and cell proliferation. Am J Respir Cell Mol Biol. 2016;54:200–9.CrossRef
17.
go back to reference Williams JC, Lee RD, Doerschuk CM, Mackman N. Effect of PAR-2 deficiency in mice on KC expression after intratracheal LPS administration. J Signal Transduct. 2011;2011:415195.CrossRef Williams JC, Lee RD, Doerschuk CM, Mackman N. Effect of PAR-2 deficiency in mice on KC expression after intratracheal LPS administration. J Signal Transduct. 2011;2011:415195.CrossRef
18.
go back to reference Antoniak S, Owens AP III, Baunacke M, Williams JC, Lee RD, Weithauser A, Sheridan PA, Malz R, Luyendyk JP, Esserman DA, Trejo J, Kirchhofer D, Blaxall BC, Pawlinski R, Beck MA, Rauch U, Mackman N. PAR-1 contributes to the innate immune response during viral infection. J Clin Invest. 2013;123:1310–22.CrossRef Antoniak S, Owens AP III, Baunacke M, Williams JC, Lee RD, Weithauser A, Sheridan PA, Malz R, Luyendyk JP, Esserman DA, Trejo J, Kirchhofer D, Blaxall BC, Pawlinski R, Beck MA, Rauch U, Mackman N. PAR-1 contributes to the innate immune response during viral infection. J Clin Invest. 2013;123:1310–22.CrossRef
19.
go back to reference Mihara K, Ramachandran R, Renaux B, Saifeddine M, Hollenberg MD. Neutrophil elastase and proteinase-3 trigger G protein-biased signaling through proteinase-activated receptor-1 (PAR1). J Biol Chem. 2013;288:32979–90.CrossRef Mihara K, Ramachandran R, Renaux B, Saifeddine M, Hollenberg MD. Neutrophil elastase and proteinase-3 trigger G protein-biased signaling through proteinase-activated receptor-1 (PAR1). J Biol Chem. 2013;288:32979–90.CrossRef
20.
go back to reference Zhao P, Lieu T, Barlow N, Sostegni S, Haerteis S, Korbmacher C, Liedtke W, Jimenez-Vargas NN, Vanner SJ, Bunnett NW. Neutrophil elastase activates protease-activated receptor-2 (PAR2) and transient receptor potential vanilloid 4 (TRPV4) to cause inflammation and pain. J Biol Chem. 2015;290:13875–87.CrossRef Zhao P, Lieu T, Barlow N, Sostegni S, Haerteis S, Korbmacher C, Liedtke W, Jimenez-Vargas NN, Vanner SJ, Bunnett NW. Neutrophil elastase activates protease-activated receptor-2 (PAR2) and transient receptor potential vanilloid 4 (TRPV4) to cause inflammation and pain. J Biol Chem. 2015;290:13875–87.CrossRef
21.
go back to reference Muley MM, Reid AR, Botz B, Bolcskei K, Helyes Z, McDougall JJ. Neutrophil elastase induces inflammation and pain in mouse knee joints via activation of proteinase-activated receptor-2. Br J Pharmacol. 2016;173:766–77.CrossRef Muley MM, Reid AR, Botz B, Bolcskei K, Helyes Z, McDougall JJ. Neutrophil elastase induces inflammation and pain in mouse knee joints via activation of proteinase-activated receptor-2. Br J Pharmacol. 2016;173:766–77.CrossRef
22.
go back to reference Koga H, Miyahara N, Fuchimoto Y, Ikeda G, Waseda K, Ono K, Tanimoto Y, Kataoka M, Gelfand EW, Tanimoto M, Kanehiro A. Inhibition of neutrophil elastase attenuates airway hyperresponsiveness and inflammation in a mouse model of secondary allergen challenge: neutrophil elastase inhibition attenuates allergic airway responses. Respir Res. 2013;14:8.CrossRef Koga H, Miyahara N, Fuchimoto Y, Ikeda G, Waseda K, Ono K, Tanimoto Y, Kataoka M, Gelfand EW, Tanimoto M, Kanehiro A. Inhibition of neutrophil elastase attenuates airway hyperresponsiveness and inflammation in a mouse model of secondary allergen challenge: neutrophil elastase inhibition attenuates allergic airway responses. Respir Res. 2013;14:8.CrossRef
23.
go back to reference Kimple AJ, Bosch DE, Giguere PM, Siderovski DP. Regulators of G-protein signaling and their Galpha substrates: promises and challenges in their use as drug discovery targets. Pharmacol Rev. 2011;63:728–49.CrossRef Kimple AJ, Bosch DE, Giguere PM, Siderovski DP. Regulators of G-protein signaling and their Galpha substrates: promises and challenges in their use as drug discovery targets. Pharmacol Rev. 2011;63:728–49.CrossRef
24.
go back to reference Xie Z, Chan EC, Druey KM. R4 regulator of G protein signaling (RGS) proteins in inflammation and immunity. AAPS J. 2016;18:294–304.CrossRef Xie Z, Chan EC, Druey KM. R4 regulator of G protein signaling (RGS) proteins in inflammation and immunity. AAPS J. 2016;18:294–304.CrossRef
25.
go back to reference Damera G, Druey KM, Cooper PR, Krymskaya VP, Soberman RJ, Amrani Y, Hoshi T, Brightling CE, Panettieri RA Jr. An RGS4-mediated phenotypic switch of bronchial smooth muscle cells promotes fixed airway obstruction in asthma. PLoS ONE. 2012;7:e28504.CrossRef Damera G, Druey KM, Cooper PR, Krymskaya VP, Soberman RJ, Amrani Y, Hoshi T, Brightling CE, Panettieri RA Jr. An RGS4-mediated phenotypic switch of bronchial smooth muscle cells promotes fixed airway obstruction in asthma. PLoS ONE. 2012;7:e28504.CrossRef
26.
go back to reference Holden NS, Bell MJ, Rider CF, King EM, Gaunt DD, Leigh R, Johnson M, Siderovski DP, Heximer SP, Giembycz MA, Newton R. beta2-Adrenoceptor agonist-induced RGS2 expression is a genomic mechanism of bronchoprotection that is enhanced by glucocorticoids. Proc Natl Acad Sci USA. 2011;108:19713–8.CrossRef Holden NS, Bell MJ, Rider CF, King EM, Gaunt DD, Leigh R, Johnson M, Siderovski DP, Heximer SP, Giembycz MA, Newton R. beta2-Adrenoceptor agonist-induced RGS2 expression is a genomic mechanism of bronchoprotection that is enhanced by glucocorticoids. Proc Natl Acad Sci USA. 2011;108:19713–8.CrossRef
27.
go back to reference Xie Y, Jiang H, Nguyen H, Jia S, Berro A, Panettieri RA Jr, Wolff DW, Abel PW, Casale TB, Tu Y. Regulator of G protein signaling 2 is a key modulator of airway hyperresponsiveness. J Allergy Clin Immunol. 2012;130:968–76.CrossRef Xie Y, Jiang H, Nguyen H, Jia S, Berro A, Panettieri RA Jr, Wolff DW, Abel PW, Casale TB, Tu Y. Regulator of G protein signaling 2 is a key modulator of airway hyperresponsiveness. J Allergy Clin Immunol. 2012;130:968–76.CrossRef
28.
go back to reference George T, Bell M, Chakraborty M, Siderovski DP, Giembycz MA, Newton R. Protective roles for RGS2 in a mouse model of house dust mite-induced airway inflammation. PLoS ONE. 2017;12:e0170269.CrossRef George T, Bell M, Chakraborty M, Siderovski DP, Giembycz MA, Newton R. Protective roles for RGS2 in a mouse model of house dust mite-induced airway inflammation. PLoS ONE. 2017;12:e0170269.CrossRef
29.
go back to reference Balenga NA, Jester W, Jiang M, Panettieri RA Jr, Druey KM. Loss of regulator of G protein signaling 5 promotes airway hyperresponsiveness in the absence of allergic inflammation. J Allergy Clin Immunol. 2014;134:451–9.CrossRef Balenga NA, Jester W, Jiang M, Panettieri RA Jr, Druey KM. Loss of regulator of G protein signaling 5 promotes airway hyperresponsiveness in the absence of allergic inflammation. J Allergy Clin Immunol. 2014;134:451–9.CrossRef
30.
go back to reference Yang Z, Cooper PR, Damera G, Mukhopadhyay I, Cho H, Kehrl JH, Panettieri RA Jr, Druey KM. Beta-agonist-associated reduction in RGS5 expression promotes airway smooth muscle hyper-responsiveness. J Biol Chem. 2011;286:11444–55.CrossRef Yang Z, Cooper PR, Damera G, Mukhopadhyay I, Cho H, Kehrl JH, Panettieri RA Jr, Druey KM. Beta-agonist-associated reduction in RGS5 expression promotes airway smooth muscle hyper-responsiveness. J Biol Chem. 2011;286:11444–55.CrossRef
31.
go back to reference Leigh R, Mostafa MM, King EM, Rider CF, Shah S, Dumonceaux C, Traves SL, McWhae A, Kolisnik T, Kooi C, Slater DM, Kelly MM, Bieda M, Miller-Larsson A, Newton R. An inhaled dose of budesonide induces genes involved in transcription and signaling in the human airways: enhancement of anti- and proinflammatory effector genes. Pharma Res Per. 2016;4:e00243.CrossRef Leigh R, Mostafa MM, King EM, Rider CF, Shah S, Dumonceaux C, Traves SL, McWhae A, Kolisnik T, Kooi C, Slater DM, Kelly MM, Bieda M, Miller-Larsson A, Newton R. An inhaled dose of budesonide induces genes involved in transcription and signaling in the human airways: enhancement of anti- and proinflammatory effector genes. Pharma Res Per. 2016;4:e00243.CrossRef
32.
go back to reference Holden NS, George T, Rider CF, Chandrasekhar A, Shah S, Kaur M, Johnson M, Siderovski DP, Leigh R, Giembycz MA, Newton R. Induction of regulator of G-protein signaling 2 expression by long-acting beta2-adrenoceptor agonists and glucocorticoids in human airway epithelial cells. J Pharmacol Exp Ther. 2014;348:12–24.CrossRef Holden NS, George T, Rider CF, Chandrasekhar A, Shah S, Kaur M, Johnson M, Siderovski DP, Leigh R, Giembycz MA, Newton R. Induction of regulator of G-protein signaling 2 expression by long-acting beta2-adrenoceptor agonists and glucocorticoids in human airway epithelial cells. J Pharmacol Exp Ther. 2014;348:12–24.CrossRef
33.
go back to reference Knight DA, Lim S, Scaffidi AK, Roche N, Chung KF, Stewart GA, Thompson PJ. Protease-activated receptors in human airways: upregulation of PAR-2 in respiratory epithelium from patients with asthma. J Allergy Clin Immunol. 2001;108:797–803.CrossRef Knight DA, Lim S, Scaffidi AK, Roche N, Chung KF, Stewart GA, Thompson PJ. Protease-activated receptors in human airways: upregulation of PAR-2 in respiratory epithelium from patients with asthma. J Allergy Clin Immunol. 2001;108:797–803.CrossRef
34.
go back to reference Asokananthan N, Graham PT, Fink J, Knight DA, Bakker AJ, McWilliam AS, Thompson PJ, Stewart GA. Activation of protease-activated receptor (PAR)-1, PAR-2, and PAR-4 stimulates IL-6, IL-8, and prostaglandin E2 release from human respiratory epithelial cells. J Immunol. 2002;168:3577–85.CrossRef Asokananthan N, Graham PT, Fink J, Knight DA, Bakker AJ, McWilliam AS, Thompson PJ, Stewart GA. Activation of protease-activated receptor (PAR)-1, PAR-2, and PAR-4 stimulates IL-6, IL-8, and prostaglandin E2 release from human respiratory epithelial cells. J Immunol. 2002;168:3577–85.CrossRef
35.
go back to reference Jose RJ, Williams AE, Mercer PF, Sulikowski MG, Brown JS, Chambers RC. Regulation of neutrophilic inflammation by proteinase-activated receptor 1 during bacterial pulmonary infection. J Immunol. 2015;194:6024–34.CrossRef Jose RJ, Williams AE, Mercer PF, Sulikowski MG, Brown JS, Chambers RC. Regulation of neutrophilic inflammation by proteinase-activated receptor 1 during bacterial pulmonary infection. J Immunol. 2015;194:6024–34.CrossRef
36.
go back to reference Schuliga M. The inflammatory actions of coagulant and fibrinolytic proteases in disease. Mediators Inflamm. 2015;2015:437695.CrossRef Schuliga M. The inflammatory actions of coagulant and fibrinolytic proteases in disease. Mediators Inflamm. 2015;2015:437695.CrossRef
37.
go back to reference Lefort J, Motreff L, Vargaftig BB. Airway administration of Escherichia coli endotoxin to mice induces glucocorticosteroid-resistant bronchoconstriction and vasopermeation. Am J Respir Cell Mol Biol. 2001;24:345–51.CrossRef Lefort J, Motreff L, Vargaftig BB. Airway administration of Escherichia coli endotoxin to mice induces glucocorticosteroid-resistant bronchoconstriction and vasopermeation. Am J Respir Cell Mol Biol. 2001;24:345–51.CrossRef
38.
go back to reference Oliveira-Dos-Santos AJ, Matsumoto G, Snow BE, Bai D, Houston FP, Whishaw IQ, Mariathasan S, Sasaki T, Wakeham A, Ohashi PS, Roder JC, Barnes CA, Siderovski DP, Penninger JM. Regulation of T cell activation, anxiety, and male aggression by RGS2. Proc Natl Acad Sci USA. 2000;97:12272–7.CrossRef Oliveira-Dos-Santos AJ, Matsumoto G, Snow BE, Bai D, Houston FP, Whishaw IQ, Mariathasan S, Sasaki T, Wakeham A, Ohashi PS, Roder JC, Barnes CA, Siderovski DP, Penninger JM. Regulation of T cell activation, anxiety, and male aggression by RGS2. Proc Natl Acad Sci USA. 2000;97:12272–7.CrossRef
39.
go back to reference Heximer SP, Knutsen RH, Sun X, Kaltenbronn KM, Rhee MH, Peng N, Oliveira-dos-Santos A, Penninger JM, Muslin AJ, Steinberg TH, Wyss JM, Mecham RP, Blumer KJ. Hypertension and prolonged vasoconstrictor signaling in RGS2-deficient mice. J Clin Invest. 2003;111:445–52.CrossRef Heximer SP, Knutsen RH, Sun X, Kaltenbronn KM, Rhee MH, Peng N, Oliveira-dos-Santos A, Penninger JM, Muslin AJ, Steinberg TH, Wyss JM, Mecham RP, Blumer KJ. Hypertension and prolonged vasoconstrictor signaling in RGS2-deficient mice. J Clin Invest. 2003;111:445–52.CrossRef
40.
go back to reference Shalaby KH, Gold LG, Schuessler TF, Martin JG, Robichaud A. Combined forced oscillation and forced expiration measurements in mice for the assessment of airway hyperresponsiveness. Respir Res. 2010;11:82.CrossRef Shalaby KH, Gold LG, Schuessler TF, Martin JG, Robichaud A. Combined forced oscillation and forced expiration measurements in mice for the assessment of airway hyperresponsiveness. Respir Res. 2010;11:82.CrossRef
41.
go back to reference Puljic R, Benediktus E, Plater-Zyberk C, Baeuerle PA, Szelenyi S, Brune K, Pahl A. Lipopolysaccharide-induced lung inflammation is inhibited by neutralization of GM-CSF. Eur J Pharmacol. 2007;557:230–5.CrossRef Puljic R, Benediktus E, Plater-Zyberk C, Baeuerle PA, Szelenyi S, Brune K, Pahl A. Lipopolysaccharide-induced lung inflammation is inhibited by neutralization of GM-CSF. Eur J Pharmacol. 2007;557:230–5.CrossRef
42.
go back to reference Chignard M, Balloy V. Neutrophil recruitment and increased permeability during acute lung injury induced by lipopolysaccharide. Am J Physiol Lung Cell Mol Physiol. 2000;279:L1083–90.CrossRef Chignard M, Balloy V. Neutrophil recruitment and increased permeability during acute lung injury induced by lipopolysaccharide. Am J Physiol Lung Cell Mol Physiol. 2000;279:L1083–90.CrossRef
43.
go back to reference Campanholle G, Landgraf RG, Borducchi E, Semedo P, Wang PH, Amano MT, Russo M, Pacheco-Silva A, Jancar S, Camara NO. Bradykinin inducible receptor is essential to lipopolysaccharide-induced acute lung injury in mice. Eur J Pharmacol. 2010;634:132–7.CrossRef Campanholle G, Landgraf RG, Borducchi E, Semedo P, Wang PH, Amano MT, Russo M, Pacheco-Silva A, Jancar S, Camara NO. Bradykinin inducible receptor is essential to lipopolysaccharide-induced acute lung injury in mice. Eur J Pharmacol. 2010;634:132–7.CrossRef
46.
go back to reference Cruz FF, Rocco PR, Pelosi P. Anti-inflammatory properties of anesthetic agents. Crit Care. 2017;21:67.CrossRef Cruz FF, Rocco PR, Pelosi P. Anti-inflammatory properties of anesthetic agents. Crit Care. 2017;21:67.CrossRef
47.
go back to reference Jiang H, Xie Y, Abel PW, Wolff DW, Toews ML, Panettieri RA Jr, Casale TB, Tu Y. Regulator of G-protein signaling 2 repression exacerbates airway hyper-responsiveness and remodeling in asthma. Am J Respir Cell Mol Biol. 2015;53:42–9.CrossRef Jiang H, Xie Y, Abel PW, Wolff DW, Toews ML, Panettieri RA Jr, Casale TB, Tu Y. Regulator of G-protein signaling 2 repression exacerbates airway hyper-responsiveness and remodeling in asthma. Am J Respir Cell Mol Biol. 2015;53:42–9.CrossRef
48.
go back to reference Akers IA, Parsons M, Hill MR, Hollenberg MD, Sanjar S, Laurent GJ, McAnulty RJ. Mast cell tryptase stimulates human lung fibroblast proliferation via protease-activated receptor-2. Am J Physiol Lung Cell Mol Physiol. 2000;278:L193–201.CrossRef Akers IA, Parsons M, Hill MR, Hollenberg MD, Sanjar S, Laurent GJ, McAnulty RJ. Mast cell tryptase stimulates human lung fibroblast proliferation via protease-activated receptor-2. Am J Physiol Lung Cell Mol Physiol. 2000;278:L193–201.CrossRef
49.
go back to reference Momen A, Afroze T, Sadi AM, Khoshbin A, Zhang H, Choi J, Gu S, Zaidi SH, Heximer SP, Husain M. Enhanced proliferation and altered calcium handling in RGS2-deficient vascular smooth muscle cells. J Recept Signal Transduct Res. 2014;34:476–83.CrossRef Momen A, Afroze T, Sadi AM, Khoshbin A, Zhang H, Choi J, Gu S, Zaidi SH, Heximer SP, Husain M. Enhanced proliferation and altered calcium handling in RGS2-deficient vascular smooth muscle cells. J Recept Signal Transduct Res. 2014;34:476–83.CrossRef
50.
go back to reference Allard B, Bara I, Gilbert G, Carvalho G, Trian T, Ozier A, Gillibert-Duplantier J, Ousova O, Maurat E, Thumerel M, Quignard JF, Girodet PO, Marthan R, Berger P. Protease activated receptor-2 expression and function in asthmatic bronchial smooth muscle. PLoS ONE. 2014;9:e86945.CrossRef Allard B, Bara I, Gilbert G, Carvalho G, Trian T, Ozier A, Gillibert-Duplantier J, Ousova O, Maurat E, Thumerel M, Quignard JF, Girodet PO, Marthan R, Berger P. Protease activated receptor-2 expression and function in asthmatic bronchial smooth muscle. PLoS ONE. 2014;9:e86945.CrossRef
51.
go back to reference Roos AB, Stampfli MR. Targeting Interleukin-17 signalling in cigarette smoke-induced lung disease: mechanistic concepts and therapeutic opportunities. Pharmacol Ther. 2017;178:123–31.CrossRef Roos AB, Stampfli MR. Targeting Interleukin-17 signalling in cigarette smoke-induced lung disease: mechanistic concepts and therapeutic opportunities. Pharmacol Ther. 2017;178:123–31.CrossRef
52.
go back to reference Togbe D, Schnyder-Candrian S, Schnyder B, Couillin I, Maillet I, Bihl F, Malo D, Ryffel B, Quesniaux VF. TLR4 gene dosage contributes to endotoxin-induced acute respiratory inflammation. J Leukoc Biol. 2006;80:451–7.CrossRef Togbe D, Schnyder-Candrian S, Schnyder B, Couillin I, Maillet I, Bihl F, Malo D, Ryffel B, Quesniaux VF. TLR4 gene dosage contributes to endotoxin-induced acute respiratory inflammation. J Leukoc Biol. 2006;80:451–7.CrossRef
53.
go back to reference Togbe D, Schnyder-Candrian S, Schnyder B, Doz E, Noulin N, Janot L, Secher T, Gasse P, Lima C, Coelho FR, Vasseur V, Erard F, Ryffel B, Couillin I, Moser R. Toll-like receptor and tumour necrosis factor dependent endotoxin-induced acute lung injury. Int J Exp Pathol. 2007;88:387–91.CrossRef Togbe D, Schnyder-Candrian S, Schnyder B, Doz E, Noulin N, Janot L, Secher T, Gasse P, Lima C, Coelho FR, Vasseur V, Erard F, Ryffel B, Couillin I, Moser R. Toll-like receptor and tumour necrosis factor dependent endotoxin-induced acute lung injury. Int J Exp Pathol. 2007;88:387–91.CrossRef
54.
go back to reference Tintinger GR, Anderson R, Feldman C. Pharmacological approaches to regulate neutrophil activity. Semin Immunopathol. 2013;35:395–409.CrossRef Tintinger GR, Anderson R, Feldman C. Pharmacological approaches to regulate neutrophil activity. Semin Immunopathol. 2013;35:395–409.CrossRef
55.
go back to reference Vergnolle N. Protease-activated receptors as drug targets in inflammation and pain. Pharmacol Ther. 2009;123:292–309.CrossRef Vergnolle N. Protease-activated receptors as drug targets in inflammation and pain. Pharmacol Ther. 2009;123:292–309.CrossRef
56.
go back to reference Vliagoftis H, Schwingshackl A, Milne CD, Duszyk M, Hollenberg MD, Wallace JL, Befus AD, Moqbel R. Proteinase-activated receptor-2-mediated matrix metalloproteinase-9 release from airway epithelial cells. J Allergy Clin Immunol. 2000;106:537–45.CrossRef Vliagoftis H, Schwingshackl A, Milne CD, Duszyk M, Hollenberg MD, Wallace JL, Befus AD, Moqbel R. Proteinase-activated receptor-2-mediated matrix metalloproteinase-9 release from airway epithelial cells. J Allergy Clin Immunol. 2000;106:537–45.CrossRef
57.
go back to reference Vliagoftis H, Befus AD, Hollenberg MD, Moqbel R. Airway epithelial cells release eosinophil survival-promoting factors (GM-CSF) after stimulation of proteinase-activated receptor 2. J Allergy Clin Immunol. 2001;107:679–85.CrossRef Vliagoftis H, Befus AD, Hollenberg MD, Moqbel R. Airway epithelial cells release eosinophil survival-promoting factors (GM-CSF) after stimulation of proteinase-activated receptor 2. J Allergy Clin Immunol. 2001;107:679–85.CrossRef
58.
go back to reference Kimple AJ, Soundararajan M, Hutsell SQ, Roos AK, Urban DJ, Setola V, Temple BR, Roth BL, Knapp S, Willard FS, Siderovski DP. Structural determinants of G-protein alpha subunit selectivity by regulator of G-protein signaling 2 (RGS2). J Biol Chem. 2009;284:19402–11.CrossRef Kimple AJ, Soundararajan M, Hutsell SQ, Roos AK, Urban DJ, Setola V, Temple BR, Roth BL, Knapp S, Willard FS, Siderovski DP. Structural determinants of G-protein alpha subunit selectivity by regulator of G-protein signaling 2 (RGS2). J Biol Chem. 2009;284:19402–11.CrossRef
59.
go back to reference Heximer SP. A “new twist” on RGS protein selectivity. Structure. 2013;21:319–20.CrossRef Heximer SP. A “new twist” on RGS protein selectivity. Structure. 2013;21:319–20.CrossRef
60.
go back to reference Karakoula A, Tovey SC, Brighton PJ, Willars GB. Lack of receptor-selective effects of either RGS2, RGS3 or RGS4 on muscarinic M3- and gonadotropin-releasing hormone receptor-mediated signalling through G alpha q/11. Eur J Pharmacol. 2008;587:16–24.CrossRef Karakoula A, Tovey SC, Brighton PJ, Willars GB. Lack of receptor-selective effects of either RGS2, RGS3 or RGS4 on muscarinic M3- and gonadotropin-releasing hormone receptor-mediated signalling through G alpha q/11. Eur J Pharmacol. 2008;587:16–24.CrossRef
61.
go back to reference Chen B, Siderovski DP, Neubig RR, Lawson MA, Trejo J. Regulation of protease-activated receptor 1 signaling by the adaptor protein complex 2 and R4 subfamily of regulator of G protein signaling proteins. J Biol Chem. 2014;289:1580–91.CrossRef Chen B, Siderovski DP, Neubig RR, Lawson MA, Trejo J. Regulation of protease-activated receptor 1 signaling by the adaptor protein complex 2 and R4 subfamily of regulator of G protein signaling proteins. J Biol Chem. 2014;289:1580–91.CrossRef
62.
go back to reference Ghil S, McCoy KL, Hepler JR. Regulator of G protein signaling 2 (RGS2) and RGS4 form distinct G protein-dependent complexes with protease activated-receptor 1 (PAR1) in live cells. PLoS ONE. 2014;9:e95355.CrossRef Ghil S, McCoy KL, Hepler JR. Regulator of G protein signaling 2 (RGS2) and RGS4 form distinct G protein-dependent complexes with protease activated-receptor 1 (PAR1) in live cells. PLoS ONE. 2014;9:e95355.CrossRef
63.
go back to reference Peters T, Henry PJ. Protease-activated receptors and prostaglandins in inflammatory lung disease. Br J Pharmacol. 2009;158:1017–33.CrossRef Peters T, Henry PJ. Protease-activated receptors and prostaglandins in inflammatory lung disease. Br J Pharmacol. 2009;158:1017–33.CrossRef
64.
go back to reference Newton R, Eddleston J, Haddad E, Hawisa S, Mak J, Lim S, Fox AJ, Donnelly LE, Chung KF. Regulation of kinin receptors in airway epithelial cells by inflammatory cytokines and dexamethasone. Eur J Pharmacol. 2002;441:193–202.CrossRef Newton R, Eddleston J, Haddad E, Hawisa S, Mak J, Lim S, Fox AJ, Donnelly LE, Chung KF. Regulation of kinin receptors in airway epithelial cells by inflammatory cytokines and dexamethasone. Eur J Pharmacol. 2002;441:193–202.CrossRef
65.
go back to reference Slater DM, Astle S, Woodcock N, Chivers JE, de Wit NC, Thornton S, Vatish M, Newton R. Anti-inflammatory and relaxatory effects of prostaglandin E2 in myometrial smooth muscle. Mol Hum Reprod. 2006;12:89–97.CrossRef Slater DM, Astle S, Woodcock N, Chivers JE, de Wit NC, Thornton S, Vatish M, Newton R. Anti-inflammatory and relaxatory effects of prostaglandin E2 in myometrial smooth muscle. Mol Hum Reprod. 2006;12:89–97.CrossRef
66.
go back to reference Knight DA, Stewart GA, Thompson PJ. Prostaglandin E2, but not prostacyclin inhibits histamine-induced contraction of human bronchial smooth muscle. Eur J Pharmacol. 1995;272:13–9.CrossRef Knight DA, Stewart GA, Thompson PJ. Prostaglandin E2, but not prostacyclin inhibits histamine-induced contraction of human bronchial smooth muscle. Eur J Pharmacol. 1995;272:13–9.CrossRef
67.
go back to reference Lan RS, Knight DA, Stewart GA, Henry PJ. Role of PGE(2) in protease-activated receptor-1, -2 and -4 mediated relaxation in the mouse isolated trachea. Br J Pharmacol. 2001;132:93–100.CrossRef Lan RS, Knight DA, Stewart GA, Henry PJ. Role of PGE(2) in protease-activated receptor-1, -2 and -4 mediated relaxation in the mouse isolated trachea. Br J Pharmacol. 2001;132:93–100.CrossRef
68.
go back to reference De Campo BA, Henry PJ. Stimulation of protease-activated receptor-2 inhibits airway eosinophilia, hyperresponsiveness and bronchoconstriction in a murine model of allergic inflammation. Br J Pharmacol. 2005;144:1100–8.CrossRef De Campo BA, Henry PJ. Stimulation of protease-activated receptor-2 inhibits airway eosinophilia, hyperresponsiveness and bronchoconstriction in a murine model of allergic inflammation. Br J Pharmacol. 2005;144:1100–8.CrossRef
69.
go back to reference Yamaguchi R, Yamamoto T, Sakamoto A, Narahara S, Sugiuchi H, Yamaguchi Y. Neutrophil elastase enhances IL-12p40 production by lipopolysaccharide-stimulated macrophages via transactivation of the PAR-2/EGFR/TLR4 signaling pathway. Blood Cells Mol Dis. 2016;59:1–7.CrossRef Yamaguchi R, Yamamoto T, Sakamoto A, Narahara S, Sugiuchi H, Yamaguchi Y. Neutrophil elastase enhances IL-12p40 production by lipopolysaccharide-stimulated macrophages via transactivation of the PAR-2/EGFR/TLR4 signaling pathway. Blood Cells Mol Dis. 2016;59:1–7.CrossRef
70.
go back to reference Lewkowich IP, Lajoie S, Stoffers SL, Suzuki Y, Richgels PK, Dienger K, Sproles AA, Yagita H, Hamid Q, Wills-Karp M. PD-L2 modulates asthma severity by directly decreasing dendritic cell IL-12 production. Mucosal Immunol. 2013;6:728–39.CrossRef Lewkowich IP, Lajoie S, Stoffers SL, Suzuki Y, Richgels PK, Dienger K, Sproles AA, Yagita H, Hamid Q, Wills-Karp M. PD-L2 modulates asthma severity by directly decreasing dendritic cell IL-12 production. Mucosal Immunol. 2013;6:728–39.CrossRef
71.
go back to reference Randolph AG, Lange C, Silverman EK, Lazarus R, Silverman ES, Raby B, Brown A, Ozonoff A, Richter B, Weiss ST. The IL12B gene is associated with asthma. Am J Hum Genet. 2004;75:709–15.CrossRef Randolph AG, Lange C, Silverman EK, Lazarus R, Silverman ES, Raby B, Brown A, Ozonoff A, Richter B, Weiss ST. The IL12B gene is associated with asthma. Am J Hum Genet. 2004;75:709–15.CrossRef
72.
go back to reference Li X, Hawkins GA, Ampleford EJ, Moore WC, Li H, Hastie AT, Howard TD, Boushey HA, Busse WW, Calhoun WJ, Castro M, Erzurum SC, Israel E, Lemanske RF Jr, Szefler SJ, Wasserman SI, Wenzel SE, Peters SP, Meyers DA, Bleecker ER. Genome-wide association study identifies TH1 pathway genes associated with lung function in asthmatic patients. J Allergy Clin Immunol. 2013;132:313–20.CrossRef Li X, Hawkins GA, Ampleford EJ, Moore WC, Li H, Hastie AT, Howard TD, Boushey HA, Busse WW, Calhoun WJ, Castro M, Erzurum SC, Israel E, Lemanske RF Jr, Szefler SJ, Wasserman SI, Wenzel SE, Peters SP, Meyers DA, Bleecker ER. Genome-wide association study identifies TH1 pathway genes associated with lung function in asthmatic patients. J Allergy Clin Immunol. 2013;132:313–20.CrossRef
73.
go back to reference Newton R, Giembycz MA. Understanding how long-acting beta2 -adrenoceptor agonists enhance the clinical efficacy of inhaled corticosteroids in asthma—an update. Br J Pharmacol. 2016;173:3405–30.CrossRef Newton R, Giembycz MA. Understanding how long-acting beta2 -adrenoceptor agonists enhance the clinical efficacy of inhaled corticosteroids in asthma—an update. Br J Pharmacol. 2016;173:3405–30.CrossRef
74.
go back to reference Newton R. Regulators of G-protein signaling as asthma therapy? Am J Respir Cell Mol Biol. 2018;58:7–9.CrossRef Newton R. Regulators of G-protein signaling as asthma therapy? Am J Respir Cell Mol Biol. 2018;58:7–9.CrossRef
Metadata
Title
A bronchoprotective role for Rgs2 in a murine model of lipopolysaccharide-induced airways inflammation
Authors
Tresa George
Mainak Chakraborty
Mark A. Giembycz
Robert Newton
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Allergy, Asthma & Clinical Immunology / Issue 1/2018
Electronic ISSN: 1710-1492
DOI
https://doi.org/10.1186/s13223-018-0266-5

Other articles of this Issue 1/2018

Allergy, Asthma & Clinical Immunology 1/2018 Go to the issue