Th17 cytokines are critical for respiratory syncytial virus-associated airway hyperreponsiveness through regulation by complement C3a and tachykinins

Monali M Bera, Bao Lu, Thomas R Martin, Shun Cui, Lawrence M Rhein, Craig Gerard, Norma P Gerard, Monali M Bera, Bao Lu, Thomas R Martin, Shun Cui, Lawrence M Rhein, Craig Gerard, Norma P Gerard

Abstract

Respiratory syncytial virus (RSV) infection is associated with serious lung disease in infants and immunocompromised individuals and is linked to development of asthma. In mice, acute RSV infection causes airway hyperresponsiveness (AHR), inflammation, and mucus hypersecretion. Infected cells induce complement activation, producing the anaphylatoxin C3a. In this paper, we show RSV-infected wild-type mice produce Th17 cytokines, a response not previously associated with viral infections. Mice deficient in the C3aR fail to develop AHR following acute RSV infection, and production of Th17 cytokines was significantly attenuated. Tachykinin production also has been implicated in RSV pathophysiology, and tachykinin receptor-null mice were similarly protected from developing AHR. These animals were also deficient in production of Th17 cytokines. Tachykinin release was absent in mice deficient in C3aR, whereas C3a levels were unchanged in tachykinin receptor-null animals. Thus, our data reveal a crucial sequence following acute RSV infection where initial C3a production causes tachykinin release, followed by activation of the IL-17A pathway. Deficiency of either receptor affords protection from AHR, identifying two potential therapeutic targets.

Figures

Figure 1. C3aR1 deficient mice are protected…
Figure 1. C3aR1 deficient mice are protected from RSV associated airway pathophysiology
A. Airway hyperresponsiveness in RSV infected C3aR1−/− and wild type Balb/c mice assessed as changes in pulmonary resistance as a function of methacholine concentration relative to PBS control. Sham infected mice were administered UV-killed RSV under identical conditions. ■ RSV infected wild type mice, ▲ RSV infected C3aR1−/− mice, □ sham infected wild type mice, △ sham infected C3aR1−/− mice. *P<0.001 relative to RSV infected C3aR1−/− mice. B. Total CD45+ cells isolated from lungs of RSV or sham infected wild type and C3aR1−/− mice. C. Differential analysis of inflammatory cells isolated from lungs of RSV or sham infected wild type and C3aR1−/− mice. D. Expression analysis of the gob5 gene, mclca3 as a function of time after infection with 106 pfu of human RSV-A2 or UV-killed (sham) virus. Data normalized to GAPDH; significance of differences is indicated. E. Assessment of the lung burden of RSV as determined by quantitative real-time PCR of RSV matrix protein RNA at 4 and 7 days following infection normalized to GAPDH. Results are representative of 3 independent experiments (mean ± SEM for 4–5 animals per group).
Figure 2. Adoptive transfer of wild type…
Figure 2. Adoptive transfer of wild type bone marrow cells to C3aR1−/− mice restores wild type AHR associated with RSV infection
Airway responsiveness as a function of methacholine concentration in RSV infected C3aR1−/− and wild type mice after bone marrow transplantation as indicated. Sham-infected mice were administered UV-killed RSV under identical conditions. Results are expressed as the pulmonary resistance normalized to PBS control. ● C3aR1−/− mice transplanted with wild type bone marrow and infected with RSV, ■ wild type mice transplanted with C3aR1−/− bone marrow and infected with RSV, ○ C3aR1−/− mice transplanted with wild type bone marrow and sham infected, □ wild type mice transplanted with C3aR1−/− bone marrow and sham infected. *P<0.001 for RSV infected C3aR1−/− mice transplanted with wild type bone marrow relative to RSV infected wild type mice transplanted with cells from C3aR1−/− animals. Results are the mean ± SEM for 6–8 animals per group.
Figure 3. TACR−/− 1 mice are protected…
Figure 3. TACR−/−1 mice are protected from RSV associated airway pathophysiology
A. Airway hyperresponsiveness in RSV infected TACR1−/− and wild type Balb/c mice assessed as changes in pulmonary resistance as a function of methacholine concentration relative to PBS control. Sham infected mice were administered UV-killed RSV under identical conditions. ● RSV infected wild type mice, ■ RSV infected TACR1−/− mice, ○ sham infected wild type mice, □ sham infected TACR1−/− mice. *P<0.001 for RSV infected wild type mice relative to RSV infected TACR1−/− animals; **P<0.01 for RSV-infected TACR1−/− mice relative to sham-infected wild type or NK1R−/− animals. B. Expression analysis of the gob5 gene, mclca3 as a function of time after infection with 106 pfu of human RSV-A2 or UV-killed (sham) virus. Data normalized to GAPDH; significance of differences is indicated. C. Total CD45+ cells isolated from RSV or sham-infected TACR1−/− or wild type mice assessed by flow cytometry. D. Differential analysis of inflammatory cells isolated from lungs of RSV or sham infected wild type and TACR1−/− mice. Results are expressed as percent of total cells. E. Assessment of the lung burden of RSV as determined by quantitative real-time PCR of RSV matrix protein RNA at 4 and 7 days following infection. Results are the mean ± SEM for 5 animals per group. Significance of differences is indicated.
Figure 4. RSV-induced expression of tachykinins is…
Figure 4. RSV-induced expression of tachykinins is reduced in C3aR1−/− mice with no concomitant alteration in C3a generation
A&B. Expression analysis of TAC1 (substance P; A) and TAC4 (hemokinin-1; B) mRNAs in the lungs of RSV and sham infected wild type and C3aR1−/− mice. Results are normalized to GAPDH; mean ± SEM for 4–5 mice per group. Significance of differences is indicated. C. ELISA determination of C3a levels in the lungs of RSV and sham infected wild type, C3aR1−/− and TACR1−/− mice. *P<0.05 for day 4-post RSV relative to sham infected mice. **P<0.001 for day 7-post RSV relative to sham infected mice; mean ± SEM for 5–9 mice per group. D. C3aR1 mRNA in the lungs of RSV and sham infected wild type and TACR1−/− mice. Results are normalized to GAPDH; mean ± SEM for 4–5 mice per group. Significance of differences is indicated.
Figure 5. Analysis of Th1 and Th2…
Figure 5. Analysis of Th1 and Th2 cytokines in the lungs of RSV and sham infected wild type, C3aR1−/− and TACR1−/− mice
A&B. IFN-γ levels in wild type and C3aR1−/− (A) or TACR1−/− (B) mice following RSV or sham infection. C&D. TNF-α in lung homogenates of RSV or sham infected C3aR1−/− (C) or TACR1−/− (D) mice determined by ELISA. E&F. IL-4 (E) and IL-5 (F) levels in wild type and C3aR1−/− mice following RSV or sham infection. G. IL-4 levels in wild type and TACR1−/− mice following RSV or sham infection. Results are the mean ± SEM for 5 mice per group. Significance of differences is indicated.
Figure 6. Mice deficient in the C3aR1…
Figure 6. Mice deficient in the C3aR1 or TACR1 fail to produce Th17 cytokines following RSV infection
A&B. IL-17A C&D. IL-6 E&F. IL-21 in lung homogenates of RSV or sham infected C3aR1−/− (A, C, E) or TACR1−/− (B, D, F) mice determined by ELISA. G&H. RORγt I&J. IL-23p19 mRNA levels in lungs of RSV or sham infected C3aR1−/− (G&I) or TACR1−/− (H&J) mice determined by quantitative real-time PCR. K&L IL-1β in lung homogenates of RSV or sham infected C3aR1−/− (K) or TACR1−/− (L) mice determined by ELISA. Results are the mean ± SEM for 5 mice per group. Significance of differences is indicated.
Figure 7. Flow cytometric identification of cells…
Figure 7. Flow cytometric identification of cells producing IL-17A, IL-6, and substance P-like immunoreactive material in the lungs following RSV infection
A&B. Intracellular cytokine staining for IL-17A in lung cells from wild type, C3aR1−/− (A) and TACR1−/− (B) mice. C&D. Intracellular staining for IL-6 in lung cells from wild type, C3aR1−/− (A) and TACR1−/− (D) mice. E. Intracellular staining for substance P-like immunoreactivity in lung cells from wild type and C3aR1−/− mice. Representative of 2 independent experiments. Results are the mean ± SEM for 3–5 mice per group. RU, relative units.

References

    1. Hall CB. Respiratory syncytial virus and parainfluenza virus. N Engl J Med. 2001;344:1917–1928.
    1. Openshaw PJ, Dean GS, Culley FJ. Links between respiratory syncytial virus bronchiolitis and childhood asthma: clinical and research approaches. Pediatr Infect Dis J. 2003;22:S58–S64. discussion S64-55.
    1. Polack FP, Teng MN, Collins PL, Prince GA, Exner M, Regele H, Lirman DD, Rabold R, Hoffman SJ, Karp CL, Kleeberger SR, Wills-Karp M, Karron RA. A role for immune complexes in enhanced respiratory syncytial virus disease. The Journal of experimental medicine. 2002;196:859–865.
    1. Hancock GE, Speelman DJ, Heers K, Bortell E, Smith J, Cosco C. Generation of atypical pulmonary inflammatory responses in BALB/c mice after immunization with the native attachment (G) glycoprotein of respiratory syncytial virus. Journal of virology. 1996;70:7783–7791.
    1. Openshaw PJ. Immunity and immunopathology to respiratory syncytial virus. The mouse model. Am J Respir Crit Care Med. 1995;152:S59–S62.
    1. Tripp RA, Jones LP, Haynes LM, Zheng H, Murphy PM, Anderson LJ. CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein. Nature immunology. 2001;2:732–738.
    1. Rudd BD, Burstein E, Duckett CS, Li X, Lukacs NW. Differential role for TLR3 in respiratory syncytial virus-induced chemokine expression. Journal of virology. 2005;79:3350–3357.
    1. Bonville CA, Rosenberg HF, Domachowske JB. Macrophage inflammatory protein-1alpha and RANTES are present in nasal secretions during ongoing upper respiratory tract infection. Pediatr Allergy Immunol. 1999;10:39–44.
    1. Miller AL, Gerard C, Schaller M, Gruber AD, Humbles AA, Lukacs NW. Deletion of CCR1 attenuates pathophysiologic responses during respiratory syncytial virus infection. J Immunol. 2006;176:2562–2567.
    1. Miller AL, Strieter RM, Gruber AD, Ho SB, Lukacs NW. CXCR2 regulates respiratory syncytial virus-induced airway hyperreactivity and mucus overproduction. J Immunol. 2003;170:3348–3356.
    1. Tripp RA, Dakhama A, Jones LP, Barskey A, Gelfand EW, Anderson LJ. The G glycoprotein of respiratory syncytial virus depresses respiratory rates through the CX3C motif and substance P. Journal of virology. 2003;77:6580–6584.
    1. Smith TF, McIntosh K, Fishaut M, Henson PM. Activation of complement by cells infected with respiratory syncytial virus. Infection and immunity. 1981;33:43–48.
    1. Morrison PT, Thomas LH, Sharland M, Friedland JS. RSV-infected airway epithelial cells cause biphasic up-regulation of CCR1 expression on human monocytes. Journal of leukocyte biology. 2007;81:1487–1495.
    1. Haynes LM, Jones LP, Barskey A, Anderson LJ, Tripp RA. Enhanced disease and pulmonary eosinophilia associated with formalin-inactivated respiratory syncytial virus vaccination are linked to G glycoprotein CX3C-CX3CR1 interaction and expression of substance P. Journal of virology. 2003;77:9831–9844.
    1. Haynes LM, Tonkin J, Anderson LJ, Tripp RA. Neutralizing anti-F glycoprotein and anti-substance P antibody treatment effectively reduces infection and inflammation associated with respiratory syncytial virus infection. Journal of virology. 2002;76:6873–6881.
    1. Dakhama A, Park JW, Taube C, El Gazzar M, Kodama T, Miyahara N, Takeda K, Kanehiro A, Balhorn A, Joetham A, Loader JE, Larsen GL, Gelfand EW. Alteration of airway neuropeptide expression and development of airway hyperresponsiveness following respiratory syncytial virus infection. Am J Physiol Lung Cell Mol Physiol. 2005;288:L761–L770.
    1. Morteau O, Lu B, Gerard C, Gerard NP. Hemokinin 1 is a full agonist at the substance P receptor. Nature immunology. 2001;2:1088.
    1. Zhang Y, Lu L, Furlonger C, Wu GE, Paige CJ. Hemokinin is a hematopoietic-specific tachykinin that regulates B lymphopoiesis. Nature immunology. 2000;1:392–397.
    1. Humbles AA, Lu B, Nilsson CA, Lilly C, Israel E, Fujiwara Y, Gerard NP, Gerard C. A role for the C3a anaphylatoxin receptor in the effector phase of asthma. Nature. 2000;406:998–1001.
    1. Drouin SM, Corry DB, Kildsgaard J, Wetsel RA. Cutting edge: the absence of C3 demonstrates a role for complement in Th2 effector functions in a murine model of pulmonary allergy. J Immunol. 2001;167:4141–4145.
    1. Nakano Y, Morita S, Kawamoto A, Suda T, Chida K, Nakamura H. Elevated complement C3a in plasma from patients with severe acute asthma. The Journal of allergy and clinical immunology. 2003;112:525–530.
    1. Bautsch W, Hoymann HG, Zhang Q, Meier-Wiedenbach I, Raschke U, Ames RS, Sohns B, Flemme N, Meyer zu Vilsendorf A, Grove M, Klos A, Kohl J. Cutting edge: guinea pigs with a natural C3a-receptor defect exhibit decreased bronchoconstriction in allergic airway disease: evidence for an involvement of the C3a anaphylatoxin in the pathogenesis of asthma. J Immunol. 2000;165:5401–5405.
    1. Walters DM, Breysse PN, Schofield B, Wills-Karp M. Complement factor 3 mediates particulate matter-induced airway hyperresponsiveness. Am J Respir Cell Mol Biol. 2002;27:413–418.
    1. Lajoie S, Lewkowich IP, Suzuki Y, Clark JR, Sproles AA, Dienger K, Budelsky AL, Wills-Karp M. Complement-mediated regulation of the IL-17A axis is a central genetic determinant of the severity of experimental allergic asthma. Nature immunology. 2010;11:928–935.
    1. Bozic CR, Lu B, Hopken UE, Gerard C, Gerard NP. Neurogenic amplification of immune complex inflammation. Science. 1996;273:1722–1725.
    1. Prince GA, Jenson AB, Horswood RL, Camargo E, Chanock RM. The pathogenesis of respiratory syncytial virus infection in cotton rats. The American journal of pathology. 1978;93:771–791.
    1. Rhein LM, Perkins M, Gerard NP, Gerard C. FcgammaRIII is protective against Pseudomonas aeruginosa pneumonia. Am J Respir Cell Mol Biol. 2008;38:401–406.
    1. Kuypers J, Wright N, Morrow R. Evaluation of quantitative and type-specific real-time RT-PCR assays for detection of respiratory syncytial virus in respiratory specimens from children. J Clin Virol. 2004;31:123–129.
    1. Ivanov I, McKenzie B, Zhou L, Tadokoro C, Lepelley A, Lafaille J, Cua D, Littman D. The Orphan Nuclear Receptor RORγt Directs the Differentiation Program of Proinflammatory IL-17+ T Helper Cells. Cell. 2006;126:1121–1133.
    1. Liu Z, Yadav P, Xu X, Su J, Chen C, Tang M, Lin H, Yu J, Qian J, Yang P-C, Wang X. The increased expression of IL-23 in inflammatory bowel disease promotes intraepithelial and lamina propria lymphocyte inflammatory responses and cytotoxicity. J Leukocyte Biology. 2011;89:597–606.
    1. Klassert TE, Pinto F, Hernandez M, Candenas ML, Hernandez MC, Abreu J, Almeida TA. Differential expression of neurokinin B and hemokinin-1 in human immune cells. Journal of neuroimmunology. 2008;196:27–34.
    1. Pribul PK, Harker J, Wang B, Wang H, Tregoning JS, Schwarze J, Openshaw PJ. Alveolar macrophages are a major determinant of early responses to viral lung infection but do not influence subsequent disease development. Journal of virology. 2008;82:4441–4448.
    1. Leverkoehne I, Gruber AD. The murine mCLCA3 (alias gob-5) protein is located in the mucin granule membranes of intestinal, respiratory, and uterine goblet cells. J Histochem Cytochem. 2002;50:829–838.
    1. O'Neill L. Gob genes, mucus and asthma. Trends in immunology. 2001;22:353.
    1. Drouin SM, Kildsgaard J, Haviland J, Zabner J, Jia HP, McCray PB, Jr, Tack BF, Wetsel RA. Expression of the complement anaphylatoxin C3a and C5a receptors on bronchial epithelial and smooth muscle cells in models of sepsis and asthma. J Immunol. 2001;166:2025–2032.
    1. Haviland DL, McCoy RL, Whitehead WT, Akama H, Molmenti EP, Brown A, Haviland JC, Parks WC, Perlmutter DH, Wetsel RA. Cellular expression of the C5a anaphylatoxin receptor (C5aR): demonstration of C5aR on nonmyeloid cells of the liver and lung. J Immunol. 1995;154:1861–1869.
    1. Melendi GA, Hoffman SJ, Karron RA, Irusta PM, Laham FR, Humbles A, Schofield B, Pan CH, Rabold R, Thumar B, Thumar A, Gerard NP, Mitzner W, Barnum SR, Gerard C, Kleeberger SR, Polack FP. C5 modulates airway hyperreactivity and pulmonary eosinophilia during enhanced respiratory syncytial virus disease by decreasing C3a receptor expression. Journal of virology. 2007;81:991–999.
    1. Piedimonte G, Rodriguez MM, King KA, McLean S, Jiang X. Respiratory syncytial virus upregulates expression of the substance P receptor in rat lungs. Am J Physiol. 1999;277:L831–L840.
    1. Auais A, Adkins B, Napchan G, Piedimonte G. Immunomodulatory effects of sensory nerves during respiratory syncytial virus infection in rats. Am J Physiol Lung Cell Mol Physiol. 2003;285:L105–L113.
    1. Chavez-Bueno S, Mejias A, Gomez AM, Olsen KD, Rios AM, Fonseca-Aten M, Ramilo O, Jafri HS. Respiratory syncytial virus-induced acute and chronic airway disease is independent of genetic background: an experimental murine model. Virol J. 2005;2:46.
    1. Smit JJ, Boon L, Lukacs NW. Respiratory virus-induced regulation of asthma-like responses in mice depends upon CD8 T cells and interferon-gamma production. The American journal of pathology. 2007;171:1944–1951.
    1. Shirey KA, Pletneva LM, Puche AC, Keegan AD, Prince GA, Blanco JC, Vogel SN. Control of RSV-induced lung injury by alternatively activated macrophages is IL-4R alpha-, TLR4-, and IFN-beta-dependent. Mucosal immunology. 2010;3:291–300.
    1. Hussell T, Spender LC, Georgiou A, O'Garra A, Openshaw PJ. Th1 and Th2 cytokine induction in pulmonary T cells during infection with respiratory syncytial virus. J Gen Virol. 1996;77(Pt 10):2447–2455.
    1. Schwarze J, Hamelmann E, Bradley KL, Takeda K, Gelfand EW. Respiratory syncytial virus infection results in airway hyperresponsiveness and enhanced airway sensitization to allergen. The Journal of clinical investigation. 1997;100:226–233.
    1. Zhang X, Lewkowich IP, Kohl G, Clark JR, Wills-Karp M, Kohl J. A protective role for C5a in the development of allergic asthma associated with altered levels of B7-H1 and B7-DC on plasmacytoid dendritic cells. J Immunol. 2009;182:5123–5130.
    1. Saunders V, Breysse P, Clark J, Sproles A, Davila M, Wills-Karp M. Particulate matter-induced airway hyperresponsiveness is lymphocyte dependent. Environmental health perspectives. 2010;118:640–646.
    1. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 Cells. Annual review of immunology. 2009;27:485–517.
    1. Zhou L, Ivanov, Spolski R, Min R, Shenderov K, Egawa T, Levy DE, Leonard WJ, Littman DR. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nature immunology. 2007;8:967–974.
    1. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, McClanahan T, Kastelein RA, Cua DJ. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. The Journal of experimental medicine. 2005;201:233–240.
    1. Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annual review of immunology. 1989;7:145–173.
    1. Barczyk A, Pierzchala W, Sozanska E. Interleukin-17 in sputum correlates with airway hyperresponsiveness to methacholine. Respiratory medicine. 2003;97:726–733.
    1. Chakir J, Shannon J, Molet S, Fukakusa M, Elias J, Laviolette M, Boulet LP, Hamid Q. Airway remodeling-associated mediators in moderate to severe asthma: effect of steroids on TGF-beta, IL-11, IL-17, and type I and type III collagen expression. The Journal of allergy and clinical immunology. 2003;111:1293–1298.
    1. He R, Oyoshi MK, Jin H, Geha RS. Epicutaneous antigen exposure induces a Th17 response that drives airway inflammation after inhalation challenge. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:15817–15822.
    1. Al-Ramli W, Prefontaine D, Chouiali F, Martin JG, Olivenstein R, Lemiere C, Hamid Q. T(H)17-associated cytokines (IL-17A and IL-17F) in severe asthma. The Journal of allergy and clinical immunology. 2009;123:1185–1187.
    1. Monaco-Shawver L, Schwartz L, Tuluc F, Guo CJ, Lai JP, Gunnam SM, Kilpatrick LE, Banerjee PP, Douglas SD, Orange JS. Substance P inhibits natural killer cell cytotoxicity through the neurokinin-1 receptor. Journal of leukocyte biology. 89:113–125.
    1. Rutigliano JA, Johnson TR, Hollinger TN, Fischer JE, Aung S, Graham BS. Treatment with anti-LFA-1 delays the CD8+ cytotoxic-T-lymphocyte response and viral clearance in mice with primary respiratory syncytial virus infection. Journal of virology. 2004;78:3014–3023.
    1. Mukherjee S, Lindell DM, Berlin AA, Morris SB, Shanley TP, Hershenson MB, Lukacs NW. IL-17-Induced Pulmonary Pathogenesis during Respiratory Viral Infection and Exacerbation of Allergic Disease. The American journal of pathology. 179:248–258.

Source: PubMed

Подписаться