Azithromycin attenuates airway inflammation in a mouse model of viral bronchiolitis

Avraham Beigelman, Cassandra L Mikols, Sean P Gunsten, Carolyn L Cannon, Steven L Brody, Michael J Walter, Avraham Beigelman, Cassandra L Mikols, Sean P Gunsten, Carolyn L Cannon, Steven L Brody, Michael J Walter

Abstract

Background: Viral bronchiolitis is the leading cause of hospitalization in young infants. It is associated with the development of childhood asthma and contributes to morbidity and mortality in the elderly. Currently no therapies effectively attenuate inflammation during the acute viral infection, or prevent the risk of post-viral asthma. We hypothesized that early treatment of a paramyxoviral bronchiolitis with azithromycin would attenuate acute and chronic airway inflammation.

Methods: Mice were inoculated with parainfluenza type 1, Sendai Virus (SeV), and treated daily with PBS or azithromycin for 7 days post-inoculation. On day 8 and 21 we assessed airway inflammation in lung tissue, and quantified immune cells and inflammatory mediators in bronchoalveolar lavage (BAL).

Results: Compared to treatment with PBS, azithromycin significantly attenuated post-viral weight loss. During the peak of acute inflammation (day 8), azithromycin decreased total leukocyte accumulation in the lung tissue and BAL, with the largest fold-reduction in BAL neutrophils. This decreased inflammation was independent of changes in viral load. Azithromycin significantly attenuated the concentration of BAL inflammatory mediators and enhanced resolution of chronic airway inflammation evident by decreased BAL inflammatory mediators on day 21.

Conclusions: In this mouse model of paramyxoviral bronchiolitis, azithromycin attenuated acute and chronic airway inflammation. These findings demonstrate anti-inflammatory effects of azithromycin that are not related to anti-viral activity. Our findings support the rationale for future prospective randomized clinical trials that will evaluate the effects of macrolides on acute viral bronchiolitis and their long-term consequences.

Figures

Figure 1
Figure 1
Azithromycin attenuated viral-dependent weight loss. (A) Experiment time line. Seven-week-old C57BL/6J female mice were inoculated with Sendai virus 5,000 egg infectious dose 50% (SeV 5 K). Mice were treated daily with PBS or azithromycin day 0 (one hour after SeV inoculation) through day 7. On days 8 and 21, bronchoalveolar lavage (BAL) fluid and lungs and were harvested. (B) Percentage of weight change from baseline (day 0) in PBS (black square) versus azithromycin (black triangle) treated mice. Values are the mean ± SEM (n = 23 in each group). A significant decrease between PBS and azithromycin treatment is indicated (*, p < 0.05). (C) Kaplan-Meier analysis of survival. No statistical difference between treatment groups (n = 23 in each group) was determined by log-rank test.
Figure 2
Figure 2
Azithromycin attenuated viral-dependent airway inflammation. Mice were inoculated with SeV and treated as in Figure 1. Eight days post-viral inoculation, lung sections were obtained from naive mice (Naive, left column); SeV infected mice treated with PBS (SeV + PBS, middle column), and SeV infected mice treated with azithromycin (SeV + Azithro, right column). Representative photomicrographs of hematoxylin and eosin stained lung sections are shown (n = 11, Azithro; n = 12, PBS). Inflammatory cells within the airway are indicated (arrow). Bar = 50 μm (top) and 25 μm (bottom).
Figure 3
Figure 3
Azithromycin attenuated viral-dependent accumulation of total cells, macrophages, lymphocytes and neutrophils in the BAL. BAL from mice treated as in Figure 1 was analyzed for total and differential cell number eight days post-inoculation. Groups are labeled as in Figure 2A and values are the mean ± SEM (n = 11, Azithro; n = 12, PBS) of total BAL cells (A), macrophages (B), lymphocytes (C) and neutrophils (D). A significant decrease between PBS and azithromycin treatment is indicated (*, p < 0.05).
Figure 4
Figure 4
Azithromycin attenuated viral-dependent airway inflammation is associated with decreased concentrations of BAL inflammatory mediators. BAL from mice treated as in Figure 1 was analyzed for inflammatory mediators eight days post-inoculation. Concentrations of inflammatory mediators in the cell-free BAL supernatant were determined using a multiplex flow-cytometry based assay (Bio-Rad Laboratories). Groups are labeled as in Figure 2B and values are the mean ± SEM (n = 11, Azithro; n = 12, PBS) of G-CSF (A), CCL2/JE (B), CCL4/MIP-1β (C), CCL5/RANTES (D). A significant decrease between PBS and azithromycin treatment is indicated (*, p < 0.05).
Figure 5
Figure 5
Azithromycin attenuated viral-dependent airway inflammation is independent of Sendai virus load. Mice were inoculated with SeV and treated as in Figure 1. Five and eight days post-inoculation, whole-lung RNA was analyzed for Sendai virus-specific and GAPDH RNA by one-step fluorogenic reverse transcriptase-polymerase chain reaction (RT-PCR). The mean of duplicate measurements of SeV-specific RNA was normalized to GAPDH and reported as the SeV to GAPDH ratio. Values are the mean ± SEM (n = 6, day 5; n = 4, day 8).
Figure 6
Figure 6
Azithromycin treatment attenuated chronic viral-dependent airway inflammation. Mice were inoculated with SeV and treated as in Figure 1. Twenty-one days post-inoculation, lung sections and BAL fluid were harvested. Values are the mean ± SEM (n = 10, Azithro; n = 8, PBS) of total BAL cells (A), macrophages, lymphocytes and neutrophils in the BAL (B), and concentrations of the chemokines: G-CSF (C) and CCL1/KC (D). A significant decrease between PBS and azithromycin treated mice is indicated (*, p < 0.05).

References

    1. Glezen WP, Taber LH, Frank AL, Kasel JA. Risk of primary infection and reinfection with respiratory syncytial virus. Am J Dis Child. 1986;140(6):543–546.
    1. Boyce TG, Mellen BG, Mitchel EF Jr, Wright PF, Griffin MR. Rates of hospitalization for respiratory syncytial virus infection among children in medicaid. J Pediatr. 2000;137(6):865–870. doi: 10.1067/mpd.2000.110531.
    1. Shay DK, Holman RC, Newman RD, Liu LL, Stout JW, Anderson LJ. Bronchiolitis-associated hospitalizations among US children, 1980-1996. JAMA. 1999;282(15):1440–1446. doi: 10.1001/jama.282.15.1440.
    1. Leader S, Kohlhase K. Respiratory syncytial virus-coded pediatric hospitalizations, 1997 to 1999. Pediatr Infect Dis J. 2002;21(7):629–632. doi: 10.1097/00006454-200207000-00005.
    1. Sigurs N. Epidemiologic and clinical evidence of a respiratory syncytial virus-reactive airway disease link. Am J Respir Crit Care Med. 2001;163(3 Pt 2):S2–6.
    1. Sigurs N, Bjarnason R, Sigurbergsson F, Kjellman B. Respiratory syncytial virus bronchiolitis in infancy is an important risk factor for asthma and allergy at age 7. Am J Respir Crit Care Med. 2000;161(5):1501–1507.
    1. Sigurs N, Gustafsson PM, Bjarnason R, Lundberg F, Schmidt S, Sigurbergsson F, Kjellman B. Severe respiratory syncytial virus bronchiolitis in infancy and asthma and allergy at age 13. Am J Respir Crit Care Med. 2005;171(2):137–141. doi: 10.1164/rccm.200406-730OC.
    1. Castro M, Schweiger T, Yin-Declue H, Ramkumar TP, Christie C, Zheng J, Cohen R, Schechtman KB, Strunk R, Bacharier LB. Cytokine response after severe respiratory syncytial virus bronchiolitis in early life. J Allergy Clin Immunol. 2008;122(4):726–733. doi: 10.1016/j.jaci.2008.07.010.
    1. Carroll KN, Wu P, Gebretsadik T, Griffin MR, Dupont WD, Mitchel EF, Hartert TV. The severity-dependent relationship of infant bronchiolitis on the risk and morbidity of early childhood asthma. J Allergy Clin Immunol. 2009;123(5):1055–1061. doi: 10.1016/j.jaci.2009.02.021.
    1. Glezen WP, Greenberg SB, Atmar RL, Piedra PA, Couch RB. Impact of respiratory virus infections on persons with chronic underlying conditions. JAMA. 2000;283(4):499–505. doi: 10.1001/jama.283.4.499.
    1. Diagnosis and management of bronchiolitis. Pediatrics. 2006;118(4):1774–1793. doi: 10.1542/peds.2006-2223.
    1. Shinkai M, Rubin BK. Macrolides and airway inflammation in children. Paediatr Respir Rev. 2005;6(3):227–235. doi: 10.1016/j.prrv.2005.06.005.
    1. Koyama H, Geddes DM. Erythromycin and diffuse panbronchiolitis. Thorax. 1997;52(10):915–918. doi: 10.1136/thx.52.10.915.
    1. Equi A, Balfour-Lynn IM, Bush A, Rosenthal M. Long term azithromycin in children with cystic fibrosis: a randomised, placebo-controlled crossover trial. Lancet. 2002;360(9338):978–984. doi: 10.1016/S0140-6736(02)11081-6.
    1. Miyatake H, Taki F, Taniguchi H, Suzuki R, Takagi K, Satake T. Erythromycin reduces the severity of bronchial hyperresponsiveness in asthma. Chest. 1991;99(3):670–673. doi: 10.1378/chest.99.3.670.
    1. Shimizu T, Kato M, Mochizuki H, Tokuyama K, Morikawa A, Kuroume T. Roxithromycin reduces the degree of bronchial hyperresponsiveness in children with asthma. Chest. 1994;106(2):458–461. doi: 10.1378/chest.106.2.458.
    1. Kamoi H, Kurihara N, Fujiwara H, Hirata K, Takeda T. The macrolide antibacterial roxithromycin reduces bronchial hyperresponsiveness and superoxide anion production by polymorphonuclear leukocytes in patients with asthma. J Asthma. 1995;32(3):191–197. doi: 10.3109/02770909509089507.
    1. Shimizu T, Kato M, Mochizuki H, Takei K, Maeda S, Tokuyama K, Morikawa A. Roxithromycin attenuates acid-induced cough and water-induced bronchoconstriction in children with asthma. J Asthma. 1997;34(3):211–217. doi: 10.3109/02770909709068191.
    1. Amayasu H, Yoshida S, Ebana S, Yamamoto Y, Nishikawa T, Shoji T, Nakagawa H, Hasegawa H, Nakabayashi M, Ishizaki Y. Clarithromycin suppresses bronchial hyperresponsiveness associated with eosinophilic inflammation in patients with asthma. Ann Allergy Asthma Immunol. 2000;84(6):594–598. doi: 10.1016/S1081-1206(10)62409-X.
    1. Black PN, Blasi F, Jenkins CR, Scicchitano R, Mills GD, Rubinfeld AR, Ruffin RE, Mullins PR, Dangain J, Cooper BC, David DB, Allegra L. Trial of roxithromycin in subjects with asthma and serological evidence of infection with Chlamydia pneumoniae. Am J Respir Crit Care Med. 2001;164(4):536–541.
    1. Ekici A, Ekici M, Erdemoglu AK. Effect of azithromycin on the severity of bronchial hyperresponsiveness in patients with mild asthma. J Asthma. 2002;39(2):181–185. doi: 10.1081/JAS-120002199.
    1. Kraft M, Cassell GH, Pak J, Martin RJ. Mycoplasma pneumoniae and Chlamydia pneumoniae in asthma: effect of clarithromycin. Chest. 2002;121(6):1782–1788. doi: 10.1378/chest.121.6.1782.
    1. Kostadima E, Tsiodras S, Alexopoulos EI, Kaditis AG, Mavrou I, Georgatou N, Papamichalopoulos A. Clarithromycin reduces the severity of bronchial hyperresponsiveness in patients with asthma. Eur Respir J. 2004;23(5):714–717. doi: 10.1183/09031936.04.00118404.
    1. Johnston SL, Blasi F, Black PN, Martin RJ, Farrell DJ, Nieman RB. The effect of telithromycin in acute exacerbations of asthma. N Engl J Med. 2006;354(15):1589–1600. doi: 10.1056/NEJMoa044080.
    1. Piacentini GL, Peroni DG, Bodini A, Pigozzi R, Costella S, Loiacono A, Boner AL. Azithromycin reduces bronchial hyperresponsiveness and neutrophilic airway inflammation in asthmatic children: a preliminary report. Allergy Asthma Proc. 2007;28(2):194–198. doi: 10.2500/aap.2007.28.2958.
    1. Kneyber MC, van Woensel JB, Uijtendaal E, Uiterwaal CS, Kimpen JL. Azithromycin does not improve disease course in hospitalized infants with respiratory syncytial virus (RSV) lower respiratory tract disease: a randomized equivalence trial. Pediatr Pulmonol. 2008;43(2):142–149. doi: 10.1002/ppul.20748.
    1. Tahan F, Ozcan A, Koc N. Clarithromycin in the treatment of RSV bronchiolitis: a double-blind, randomised, placebo-controlled trial. Eur Respir J. 2007;29(1):91–97. doi: 10.1183/09031936.00029206.
    1. Kim EY, Battaile JT, Patel AC, You Y, Agapov E, Grayson MH, Benoit LA, Byers DE, Alevy Y, Tucker J, Swanson S, Tidwell R, Tyner JW, Morton JD, Castro M, Polineni D, Patterson GA, Schwendener RA, Allard JD, Peltz G, Holtzman MJ. Persistent activation of an innate immune response translates respiratory viral infection into chronic lung disease. Nat Med. 2008;14(6):633–640. doi: 10.1038/nm1770.
    1. Walter MJ, Morton JD, Kajiwara N, Agapov E, Holtzman MJ. Viral induction of a chronic asthma phenotype and genetic segregation from the acute response. J Clin Invest. 2002;110(2):165–175.
    1. Beigelman A, Gunsten S, Mikols CL, Vidavsky I, Cannon CL, Brody SL, Walter MJ. Azithromycin Attenuates Airway Inflammation in a Noninfectious Mouse Model of Allergic Asthma. Chest. 2009;136(2):498–506. doi: 10.1378/chest.08-3056.
    1. Gunsten S, Mikols CL, Grayson MH, Schwendener RA, Agapov E, Tidwell RM, Cannon CL, Brody SL, Walter MJ. IL-12 p80-dependent macrophage recruitment primes the host for increased survival following a lethal respiratory viral infection. Immunology. 2009;126(4):500–513. doi: 10.1111/j.1365-2567.2008.02923.x.
    1. Mikols CL, Yan L, Norris JY, Russell TD, Khalifah AP, Hachem RR, Chakinala MM, Yusen RD, Castro M, Kuo E, Patterson GA, Mohanakumar T, Trulock EP, Walter MJ. IL-12 p80 is an innate epithelial cell effector that mediates chronic allograft dysfunction. Am J Respir Crit Care Med. 2006;174(4):461–470. doi: 10.1164/rccm.200512-1886OC.
    1. Russell TD, Yan Q, Fan G, Khalifah AP, Bishop DK, Brody SL, Walter MJ. IL-12 p40 homodimer-dependent macrophage chemotaxis and respiratory viral inflammation are mediated through IL-12 receptor beta 1. J Immunol. 2003;171(12):6866–6874.
    1. Hoffmann N, Lee B, Hentzer M, Rasmussen TB, Song Z, Johansen HK, Givskov M, Hoiby N. Azithromycin blocks quorum sensing and alginate polymer formation and increases the sensitivity to serum and stationary-growth-phase killing of Pseudomonas aeruginosa and attenuates chronic P. aeruginosa lung infection in Cftr(-/-) mice. Antimicrob Agents Chemother. 2007;51(10):3677–3687. doi: 10.1128/AAC.01011-06.
    1. Conte JE Jr, Golden J, Duncan S, McKenna E, Lin E, Zurlinden E. Single-dose intrapulmonary pharmacokinetics of azithromycin, clarithromycin, ciprofloxacin, and cefuroxime in volunteer subjects. Antimicrob Agents Chemother. 1996;40(7):1617–1622.
    1. Bao Z, Lim S, Liao W, Lin Y, Thiemermann C, Leung BP, Wong WS. Glycogen synthase kinase-3beta inhibition attenuates asthma in mice. Am J Respir Crit Care Med. 2007;176(5):431–438. doi: 10.1164/rccm.200609-1292OC.
    1. Duan W, Chan JH, Wong CH, Leung BP, Wong WS. Anti-inflammatory effects of mitogen-activated protein kinase kinase inhibitor U0126 in an asthma mouse model. J Immunol. 2004;172(11):7053–7059.
    1. Look DC, Walter MJ, Williamson MR, Pang L, You Y, Sreshta JN, Johnson JE, Zander DS, Brody SL. Effects of paramyxoviral infection on airway epithelial cell Foxj1 expression, ciliogenesis, and mucociliary function. Am J Pathol. 2001;159(6):2055–2069.
    1. Sato K, Suga M, Akaike T, Fujii S, Muranaka H, Doi T, Maeda H, Ando M. Therapeutic effect of erythromycin on influenza virus-induced lung injury in mice. Am J Respir Crit Care Med. 1998;157(3 Pt 1):853–857.
    1. Asada M, Yoshida M, Suzuki T, Hatachi Y, Sasaki T, Yasuda H, Nakayama K, Nishimura H, Nagatomi R, Kubo H, Yamaya M. Macrolide antibiotics inhibit respiratory syncytial virus infection in human airway epithelial cells. Antiviral Res. 2009;83(2):191–200. doi: 10.1016/j.antiviral.2009.05.003.
    1. Ivetic Tkalcevic V, Bosnjak B, Hrvacic B, Bosnar M, Marjanovic N, Ferencic Z, Situm K, Culic O, Parnham MJ, Erakovic V. Anti-inflammatory activity of azithromycin attenuates the effects of lipopolysaccharide administration in mice. Eur J Pharmacol. 2006;539(1-2):131–138. doi: 10.1016/j.ejphar.2006.03.074.
    1. Leiva M, Ruiz-Bravo A, Jimenez-Valera M. Effects of telithromycin in in vitro and in vivo models of lipopolysaccharide-induced airway inflammation. Chest. 2008;134(1):20–29. doi: 10.1378/chest.07-3056.
    1. Tsai WC, Rodriguez ML, Young KS, Deng JC, Thannickal VJ, Tateda K, Hershenson MB, Standiford TJ. Azithromycin blocks neutrophil recruitment in Pseudomonas endobronchial infection. Am J Respir Crit Care Med. 2004;170(12):1331–1339. doi: 10.1164/rccm.200402-200OC.
    1. Harcourt JL, Karron RA, Tripp RA. Anti-G protein antibody responses to respiratory syncytial virus infection or vaccination are associated with inhibition of G protein CX3C-CX3CR1 binding and leukocyte chemotaxis. J Infect Dis. 2004;190(11):1936–1940. doi: 10.1086/425516.
    1. Zhang W, Choi Y, Haynes LM, Harcourt JL, Anderson LJ, Jones LP, Tripp RA. Vaccination to induce antibodies blocking the CX3C-CX3CR1 interaction of respiratory syncytial virus G protein reduces pulmonary inflammation and virus replication in mice. J Virol. pp. 1148–1157.
    1. Haynes LM, Caidi H, Radu GU, Miao C, Harcourt JL, Tripp RA, Anderson LJ. Therapeutic monoclonal antibody treatment targeting respiratory syncytial virus (RSV) G protein mediates viral clearance and reduces the pathogenesis of RSV infection in BALB/c mice. J Infect Dis. 2009;200(3):439–447. doi: 10.1086/600108.
    1. Kawasaki S, Takizawa H, Ohtoshi T, Takeuchi N, Kohyama T, Nakamura H, Kasama T, Kobayashi K, Nakahara K, Morita Y, Yamamoto K. Roxithromycin inhibits cytokine production by and neutrophil attachment to human bronchial epithelial cells in vitro. Antimicrob Agents Chemother. 1998;42(6):1499–1502.
    1. Adachi T, Motojima S, Hirata A, Fukuda T, Kihara N, Kosaku A, Ohtake H, Makino S. Eosinophil apoptosis caused by theophylline, glucocorticoids, and macrolides after stimulation with IL-5. J Allergy Clin Immunol. 1996;98(6 Pt 2):S207–215. doi: 10.1016/S0091-6749(96)70068-4.
    1. Hatipoglu U, Rubinstein I. Low-dose, long-term macrolide therapy in asthma: An overview. Clin Mol Allergy. 2004;2(1):4. doi: 10.1186/1476-7961-2-4.
    1. Jenkins SG, Farrell DJ. Increase in pneumococcus macrolide resistance, United States. Emerg Infect Dis. 2009;15(8):1260–1264. doi: 10.3201/eid1508.081187.

Source: PubMed

3
Předplatit