Azithromycin treatment alters gene expression in inflammatory, lipid metabolism, and cell cycle pathways in well-differentiated human airway epithelia

Carla Maria P Ribeiro, Harry Hurd, Yichao Wu, Mary E B Martino, Lisa Jones, Brian Brighton, Richard C Boucher, Wanda K O'Neal, Carla Maria P Ribeiro, Harry Hurd, Yichao Wu, Mary E B Martino, Lisa Jones, Brian Brighton, Richard C Boucher, Wanda K O'Neal

Abstract

Prolonged macrolide antibiotic therapy at low doses improves clinical outcome in patients affected with diffuse panbronchiolitis and cystic fibrosis. Consensus is building that the therapeutic effects are due to anti-inflammatory, rather than anti-microbial activities, but the mode of action is likely complex. To gain insights into how the macrolide azithromycin (AZT) modulates inflammatory responses in airways, well-differentiated primary cultures of human airway epithelia were exposed to AZT alone, an inflammatory stimulus consisting of soluble factors from cystic fibrosis airways, or AZT followed by the inflammatory stimulus. RNA microarrays were conducted to identify global and specific gene expression changes. Analysis of gene expression changes revealed that the AZT treatment alone altered the gene profile of the cells, primarily by significantly increasing the expression of lipid/cholesterol genes and decreasing the expression of cell cycle/mitosis genes. The increase in cholesterol biosynthetic genes was confirmed by increased filipin staining, an index of free cholesterol, after AZT treatment. AZT also affected genes with inflammatory annotations, but the effect was variable (both up- and down-regulation) and gene specific. AZT pretreatment prevented the up-regulation of some genes, such as MUC5AC and MMP9, triggered by the inflammatory stimulus, but the up-regulation of other inflammatory genes, e.g., cytokines and chemokines, such as interleukin-8, was not affected. On the other hand, HLA genes were increased by AZT. Notably, secreted IL-8 protein levels did not reflect mRNA levels, and were, in fact, higher after AZT pretreatment in cultures exposed to the inflammatory stimulus, suggesting that AZT can affect inflammatory pathways other than by altering gene expression. These findings suggest that the specific effects of AZT on inflamed and non-inflamed airway epithelia are likely relevant to its clinical activity, and their apparent complexity may help explain the diverse immunomodulatory roles of macrolides.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Clustering of the expression data.
Figure 1. Clustering of the expression data.
Clustering of the data, as described in Methods, revealing the general organization of differential gene expression. Green color represents reduced gene expression relative to red color, which represents increased expression. Treatments are explained in Table 1. PBS: phosphate buffered saline. AZT: azithromycin. SMM: Supernatant of mucopurulent material from CF airways.
Figure 2. Illustration of the most significant…
Figure 2. Illustration of the most significant gene ontology (GO) groups whose expression was modulated by the various treatments.
The following comparisons were analyzed: AZT 6 hr vs. PBS 6 hr, AZT 24 hr vs. PBS 24 hr, SMM 6 hr vs. PBS 6 hr, SMM 24 hr vs. PBS 24 hr, AZT 48 hr (+SMM during last 6 hr) vs. SMM 6 hr, and AZT 72 hr (+SMM during the last 24 hr) vs. SMM 24 hr. Green = down-regulated groups; red = up-regulated groups; white = not significant. Values in boxes represent the probability that the genes within the list from each GO group would be present by chance – the more negative, the less likely genes within these categories are present by chance. See corresponding text for details.
Figure 3. Venn diagrams representing the number…
Figure 3. Venn diagrams representing the number of differentially expressed inflammation-related genes as a function of treatment.
The top Venn diagram compares SMM-induced genes to AZT up- and down-regulated genes. The bottom Venn diagram shows how AZT pretreatment affected SMM genes.
Figure 4. AZT up-regulates lipid/cholesterol metabolism and…
Figure 4. AZT up-regulates lipid/cholesterol metabolism and down-regulates cell cycle genes.
A: The cholesterol biosynthetic pathway and the fold-changes induced for each of the enzymes within the pathway are indicated. Data are expressed as fold changes in gene expression values from 24 hr AZT vs. 24 hr PBS (left numbers), 24 hr SMM vs. 24 hr PBS (center numbers) and 72 hr AZT (+addition of mucosal SMM during the last 24 hr) vs. 24 hr PBS (right numbers). Both SMM and AZT increased the enzymes involved in the cholesterol biosynthetic pathway, and their effect appears to be additive. B and C: Ingenuity Pathways Analysis™ was used to generate two top significant networks for genes regulated by AZT 24 hr vs. PBS 24 hr. The top two networks are shown. The analysis settings were as follows: only direct relationships were considered, endogenous chemicals were excluded, and only relationships where data sources = Argonaute 2 or Ingenuity curated findings were considered. The network depicted in B was labeled “Lipid Metabolism, Small Molecule Biochemistry, Nucleic Acid Metabolism” and had a score = 47. The network depicted in C was labeled “Cancer, Cell Cycle, Reproductive System Disease” and had a score = 44. Red indicates up-regulation. Green indicates down-regulation.
Figure 5. AZT decreases basal and SMM-stimulated…
Figure 5. AZT decreases basal and SMM-stimulated mRNA and protein expression levels of MUC5AC in human airway epithelia.
Well-differentiated HBE were exposed for 24 hr to mucosal PBS or mucosal SMM in the absence of presence of 72 hr pretreatment with 30 µg/ml serosal AZT. A: MUC5AC mRNA levels, expressed as fold change from mucosal PBS-exposed HBE. B: Immunocytochemical assessment of MUC5AC in HBE. Right panel depicts compiled data from MUC5AC expression as a percent of PBS-exposed HBE. C: AB-PAS staining from HBE subjected to the various treatments. Right panel shows compiled data from AB-PAS staining as a percent of PBS-exposed HBE. *p

Figure 6. Effect of AZT on IL-8…

Figure 6. Effect of AZT on IL-8 secretion in human airway epithelia.

A: AZT does…

Figure 6. Effect of AZT on IL-8 secretion in human airway epithelia.
A: AZT does not affect basal airway epithelial IL-8 secretion. Basal IL-8 secretion from well-differentiated primary cultures of normal human bronchial epithelia serosally exposed to different doses of AZT for 24 hr and 48 hr. B: Dose response for AZT-potentiated IL-8 secretion triggered by 24 hr mucosal exposure of HBE to supernatant from mucopurulent material (SMM) from CF airways. Well-differentiated primary cultures of normal human bronchial epithelia were pretreated with AZT for 72 hr and exposed to mucosal PBS or SMM during the last 24 hr of macrolide treatment. C: Time course for AZT-potentiated SMM-induced IL-8 secretion. Vehicle or 100 µg/ml AZT were added to the serosal surface of well-differentiated normal HBE and a 48 hr time course performed for IL-8 secretion. AZT pretreatment did not alter the basal levels of secreted IL-8, but potentiated SMM-induced IL-8 secretion at both 24 and 48 hr (corresponding to 72 and 96 hr AZT treatment, respectively).

Figure 7. Mucosal bacterial and inflammatory mediator…

Figure 7. Mucosal bacterial and inflammatory mediator challenge (SMM) or serosal AZT induces accumulation of…

Figure 7. Mucosal bacterial and inflammatory mediator challenge (SMM) or serosal AZT induces accumulation of cholesterol in human airway epithelia.
A: Quantitative RT-PCR confirmation of increased SREBP1 levels in AZT-treated cultures. B: Representative filipin stain in WD HBE exposed to 24 hr mucosal PBS (I), 24 hr mucosal SMM (II) or 24 hr 30 µg/ml serosal AZT (III). (IV–VI): Same conditions as in I–III, except that cultures were pretreated with 50 µM mevastatin, as described in Methods. Bar: 10 µm. C: Compiled data from filipin fluorescence from the treatments illustrated in 7B. *p
All figures (7)
Similar articles
Cited by
References
    1. Jaffe A, Bush A. Anti-inflammatory effects of macrolides in lung disease. Pediatr Pulmonol. 2001;31:464–473. - PubMed
    1. Pechere JC. New perspectives on macrolide antibiotics. Int J Antimicrob Agents. 2001;18(Suppl 1):S93–S97. - PubMed
    1. Shinkai M, Henke MO, Rubin BK. Macrolide antibiotics as immunomodulatory medications: proposed mechanisms of action. Pharmacol Ther. 2008;117:393–405. - PubMed
    1. Wagner T, Soong G, Sokol S, Saiman L, Prince A. Effects of azithromycin on clinical isolates of Pseudomonas aeruginosa from cystic fibrosis patients. Chest. 2005;128:912–919. - PubMed
    1. Hoffmann N, Lee B, Hentzer M, Rasmussen TB, Song Z, et al. 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;51:3677–3687. - PMC - PubMed
Show all 60 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6. Effect of AZT on IL-8…
Figure 6. Effect of AZT on IL-8 secretion in human airway epithelia.
A: AZT does not affect basal airway epithelial IL-8 secretion. Basal IL-8 secretion from well-differentiated primary cultures of normal human bronchial epithelia serosally exposed to different doses of AZT for 24 hr and 48 hr. B: Dose response for AZT-potentiated IL-8 secretion triggered by 24 hr mucosal exposure of HBE to supernatant from mucopurulent material (SMM) from CF airways. Well-differentiated primary cultures of normal human bronchial epithelia were pretreated with AZT for 72 hr and exposed to mucosal PBS or SMM during the last 24 hr of macrolide treatment. C: Time course for AZT-potentiated SMM-induced IL-8 secretion. Vehicle or 100 µg/ml AZT were added to the serosal surface of well-differentiated normal HBE and a 48 hr time course performed for IL-8 secretion. AZT pretreatment did not alter the basal levels of secreted IL-8, but potentiated SMM-induced IL-8 secretion at both 24 and 48 hr (corresponding to 72 and 96 hr AZT treatment, respectively).
Figure 7. Mucosal bacterial and inflammatory mediator…
Figure 7. Mucosal bacterial and inflammatory mediator challenge (SMM) or serosal AZT induces accumulation of cholesterol in human airway epithelia.
A: Quantitative RT-PCR confirmation of increased SREBP1 levels in AZT-treated cultures. B: Representative filipin stain in WD HBE exposed to 24 hr mucosal PBS (I), 24 hr mucosal SMM (II) or 24 hr 30 µg/ml serosal AZT (III). (IV–VI): Same conditions as in I–III, except that cultures were pretreated with 50 µM mevastatin, as described in Methods. Bar: 10 µm. C: Compiled data from filipin fluorescence from the treatments illustrated in 7B. *p
All figures (7)

References

    1. Jaffe A, Bush A. Anti-inflammatory effects of macrolides in lung disease. Pediatr Pulmonol. 2001;31:464–473.
    1. Pechere JC. New perspectives on macrolide antibiotics. Int J Antimicrob Agents. 2001;18(Suppl 1):S93–S97.
    1. Shinkai M, Henke MO, Rubin BK. Macrolide antibiotics as immunomodulatory medications: proposed mechanisms of action. Pharmacol Ther. 2008;117:393–405.
    1. Wagner T, Soong G, Sokol S, Saiman L, Prince A. Effects of azithromycin on clinical isolates of Pseudomonas aeruginosa from cystic fibrosis patients. Chest. 2005;128:912–919.
    1. Hoffmann N, Lee B, Hentzer M, Rasmussen TB, Song Z, et al. 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;51:3677–3687.
    1. Sakito O, Kadota J, Kohno S, Abe K, Shirai R, et al. Interleukin 1 beta, tumor necrosis factor alpha, and interleukin 8 in bronchoalveolar lavage fluid of patients with diffuse panbronchiolitis: a potential mechanism of macrolide therapy. Respiration. 1996;63:42–48.
    1. Tsai WC, Rodriguez ML, Young KS, Deng JC, Thannickal VJ, et al. Azithromycin blocks neutrophil recruitment in Pseudomonas endobronchial infection. Am J Respir Crit Care Med. 2004;170:1331–1339.
    1. Khair OA, Devalia JL, Abdelaziz MM, Sapsford RJ, Davies RJ. Effect of erythromycin on Haemophilus influenzae endotoxin-induced release of IL-6, IL-8 and sICAM-1 by cultured human bronchial epithelial cells. Eur Respir J. 1995;8:1451–1457.
    1. Takizawa H, Desaki M, Ohtoshi T, Kawasaki S, Kohyama T, et al. Erythromycin modulates IL-8 expression in normal and inflamed human bronchial epithelial cells. Am J Respir Crit Care Med. 1997;156:266–271.
    1. Kawasaki S, Takizawa H, Ohtoshi T, Takeuchi N, Kohyama T, et al. Roxithromycin inhibits cytokine production by and neutrophil attachment to human bronchial epithelial cells in vitro. Antimicrob Agents Chemother. 1998;42:1499–1502.
    1. Kadota J, Sakito O, Kohno S, Sawa H, Mukae H, et al. A mechanism of erythromycin treatment in patients with diffuse panbronchiolitis. Am Rev Respir Dis. 1993;147:153–159.
    1. Oda H, Kadota J, Kohno S, Hara K. Erythromycin inhibits neutrophil chemotaxis in bronchoalveoli of diffuse panbronchiolitis. Chest. 1994;106:1116–1123.
    1. Ichikawa Y, Ninomiya H, Koga H, Tanaka M, Kinoshita M, et al. Erythromycin reduces neutrophils and neutrophil-derived elastolytic-like activity in the lower respiratory tract of bronchiolitis patients. Am Rev Respir Dis. 1992;146:196–203.
    1. Yamasawa H, Oshikawa K, Ohno S, Sugiyama Y. Macrolides inhibit epithelial cell-mediated neutrophil survival by modulating granulocyte macrophage colony-stimulating factor release. Am J Respir Cell Mol Biol. 2004;30:569–575.
    1. Koch CC, Esteban DJ, Chin AC, Olson ME, Read RR, et al. Apoptosis, oxidative metabolism and interleukin-8 production in human neutrophils exposed to azithromycin: effects of Streptococcus pneumoniae. J Antimicrob Chemother. 2000;46:19–26.
    1. Culic O, Erakovic V, Cepelak I, Barisic K, Brajsa K, et al. Azithromycin modulates neutrophil function and circulating inflammatory mediators in healthy human subjects. Eur J Pharmacol. 2002;450:277–289.
    1. Hodge S, Hodge G, Brozyna S, Jersmann H, Holmes M, et al. Azithromycin increases phagocytosis of apoptotic bronchial epithelial cells by alveolar macrophages. Eur Respir J. 2006;28:486–495.
    1. Mankin AS. Macrolide myths. Curr Opin Microbiol. 2008;11:414–421.
    1. Imamura Y, Yanagihara K, Mizuta Y, Seki M, Ohno H, et al. Azithromycin inhibits MUC5AC production induced by the Pseudomonas aeruginosa autoinducer N-(3-Oxododecanoyl) homoserine lactone in NCI-H292 Cells. Antimicrob Agents Chemother. 2004;48:3457–3461.
    1. Shimizu T, Shimizu S, Hattori R, Gabazza EC, Majima Y. In vivo and in vitro effects of macrolide antibiotics on mucus secretion in airway epithelial cells. Am J Respir Crit Care Med. 2003;168:581–587.
    1. Kanai K, Asano K, Hisamitsu T, Suzaki H. Suppression of matrix metalloproteinase production from nasal fibroblasts by macrolide antibiotics in vitro. Eur Respir J. 2004;23:671–678.
    1. Hashimoto N, Kawabe T, Hara T, Imaizumi K, Wakayama H, et al. Effect of erythromycin on matrix metalloproteinase-9 and cell migration. J Lab Clin Med. 2001;137:176–183.
    1. Ribeiro CMP, Paradiso AM, Carew MA, Shears SB, Boucher RC. Cystic fibrosis airway epithelial Ca2+i signaling. The mechanism for the larger agonist-mediated Ca2+i signals in human cystic fibrosis airway epithelia. J Biol Chem. 2005;280:10202–10209.
    1. Ribeiro CMP, Paradiso AM, Schwab U, Perez-Vilar J, Jones L, et al. Chronic airway infection/Inflammation Induces a Ca2+i-dependent hyperinflammatory response in human cystic fibrosis airway epithelia. J Biol Chem. 2005;280:17798–17806.
    1. Wilms EB, Touw DJ, Heijerman HG. Pharmacokinetics of azithromycin in plasma, blood, polymorphonuclear neutrophils and sputum during long-term therapy in patients with cystic fibrosis. Ther Drug Monit. 2006;28:219–225.
    1. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4:249–264.
    1. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001;29:2002–2007.
    1. White NM, Corey DA, Kelley TJ. Mechanistic similarities between cultured cell models of cystic fibrosis and niemann-pick type C. Am J Respir Cell Mol Biol. 2004;31:538–543.
    1. Ribeiro CM, Paradiso AM, Livraghi A, Boucher RC. The mitochondrial barriers segregate agonist-induced calcium-dependent functions in human airway epithelia. J Gen Physiol. 2003;122:377–387.
    1. Kurdowska A, Noble JM, Griffith DE. The effect of azithromycin and clarithromycin on ex vivo interleukin-8 (IL-8) release from whole blood and IL-8 production by human alveolar macrophages. J Antimicrob Chemother. 2001;47:867–870.
    1. Shinkai M, Foster GH, Rubin BK. Macrolide antibiotics modulate ERK phosphorylation and IL-8 and GM-CSF production by human bronchial epithelial cells. Am J Physiol. 2006;290:L75–L85.
    1. Parnham MJ, Culic O, Erakovic V, Munic V, Popovic-Grle S, et al. Modulation of neutrophil and inflammation markers in chronic obstructive pulmonary disease by short-term azithromycin treatment. Eur J Pharmacol. 2005;517:132–143.
    1. Verleden GM, Vanaudenaerde BM, Dupont LJ, Van Raemdonck DE. Azithromycin reduces airway neutrophilia and interleukin-8 in patients with bronchiolitis obliterans syndrome. Am J Respir Crit Care Med. 2006;174:566–570.
    1. Janowski BA. The hypocholesterolemic agent LY295427 upregulates INSIG-1, identifying the INSIG-1 protein as a mediator of cholesterol homeostasis through SREBP. Proc Natl Acad Sci U S A. 2002;99:12675–12680.
    1. Kanai K, Asano K, Hisamitsu T, Suzaki H. Suppression of matrix metalloproteinase-9 production from neutrophils by a macrolide antibiotic, roxithromycin, in vitro. Mediators Inflamm. 2004;13:313–319.
    1. Ishimoto H, Mukae H, Sakamoto N, Amenomori M, Kitazaki T, et al. Different effects of telithromycin on MUC5AC production induced by human neutrophil peptide-1 or lipopolysaccharide in NCI-H292 cells compared with azithromycin and clarithromycin. J Antimicrob Chemother. 2009;63:109–114.
    1. Tamaoki J, Takeyama K, Tagaya E, Konno K. Effect of clarithromycin on sputum production and its rheological properties in chronic respiratory tract infections. Antimicrob Agents Chemother. 1995;39:1688–1690.
    1. Van den Steen PE, Proost P, Wuyts A, Van Damme J, Opdenakker G. Neutrophil gelatinase B potentiates interleukin-8 tenfold by aminoterminal processing, whereas it degrades CTAP-III, PF-4, and GRO-alpha and leaves RANTES and MCP-2 intact. Blood. 2000;96:2673–2681.
    1. Yu Q, Stamenkovic I. Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis. Genes Dev. 2000;14:163–176.
    1. Kelly EA, Busse WW, Jarjour NN. Increased matrix metalloproteinase-9 in the airway after allergen challenge. Am J Respir Crit Care Med. 2000;162:1157–1161.
    1. Gaggar A, Li Y, Weathington N, Winkler M, Kong M, et al. Matrix metalloprotease-9 dysregulation in lower airway secretions of cystic fibrosis patients. Am J Physiol Lung Cell Mol Physiol. 2007;293:L96–L104.
    1. Sagel SD, Kapsner RK, Osberg I. Induced sputum matrix metalloproteinase-9 correlates with lung function and airway inflammation in children with cystic fibrosis. Pediatr Pulmonol. 2005;39:224–232.
    1. Askew DJ, Silverman GA. Intracellular and extracellular serpins modulate lung disease. J Perinatol. 2008;28(Suppl 3):S127–S135.
    1. Liu K, Xu Q. Roxithromycin inhibits the effector phase of delayed-type hypersensitivity. Int Immunopharmacol. 2008;8:126–131.
    1. Simpson JL, Powell H, Boyle MJ, Scott RJ, Gibson PG. Clarithromycin targets neutrophilic airway inflammation in refractory asthma. Am J Respir Crit Care Med. 2008;177:148–155.
    1. Ogawa M, Suzuki J, Takayama K, Isobe M. Matrix metalloproteinase suppression induced by clarithromycin in murine cardiac allografts. Transplant Proc. 2009;41:395–397.
    1. Maxfield FR, Tabas I. Role of cholesterol and lipid organization in disease. Nature. 2005;438:612–621.
    1. D'Avila H, Maya-Monteiro CM, Bozza PT. Lipid bodies in innate immune response to bacterial and parasite infections. Int Immunopharmacol. 2008;8:1308–1315.
    1. Van Bambeke F, Gerbaux C, Michot JM, d'Yvoire MB, Montenez JP, et al. Lysosomal alterations induced in cultured rat fibroblasts by long-term exposure to low concentrations of azithromycin. J Antimicrob Chemother. 1998;42:761–767.
    1. Bengoechea-Alonso MT, Punga T, Ericsson J. Hyperphosphorylation regulates the activity of SREBP1 during mitosis. Proc Natl Acad Sci U S A. 2005;102:11681–11686.
    1. Arito M, Horiba T, Hachimura S, Inoue J, Sato R. Growth factor-induced phosphorylation of sterol regulatory element-binding proteins inhibits sumoylation, thereby stimulating the expression of their target genes, low density lipoprotein uptake, and lipid synthesis. J Biol Chem. 2008;283:15224–15231.
    1. Tyteca D, Schanck A, Dufrene YF, Deleu M, Courtoy PJ, et al. The macrolide antibiotic azithromycin interacts with lipids and affects membrane organization and fluidity: studies on Langmuir-Blodgett monolayers, liposomes and J774 macrophages. J Membr Biol. 2003;192:203–215.
    1. Fa N, Lins L, Courtoy PJ, Dufrene Y, Van Der SP, et al. Decrease of elastic moduli of DOPC bilayers induced by a macrolide antibiotic, azithromycin. Biochim Biophys Acta. 2007;1768:1830–1838.
    1. Matsumori N, Morooka A, Murata M. Detailed description of the conformation and location of membrane-bound erythromycin a using isotropic bicelles. J Med Chem. 2006;49:3501–3508.
    1. Baronas ET, Lee JW, Alden C, Hsieh FY. Biomarkers to monitor drug-induced phospholipidosis. Toxicol Appl Pharmacol. 2007;218:72–78.
    1. Dobrosotskaya IY, Seegmiller AC, Brown MS, Goldstein JL, Rawson RB. Regulation of SREBP processing and membrane lipid production by phospholipids in Drosophila. Science. 2002;296:879–883.
    1. Abe S, Nakamura H, Inoue S, Takeda H, Saito H, et al. Interleukin-8 gene repression by clarithromycin is mediated by the activator protein-1 binding site in human bronchial epithelial cells. Am J Respir Cell Mol Biol. 2000;22:51–60.
    1. Chen SZ, Jiang M, Zhen YS. HERG K+ channel expression-related chemosensitivity in cancer cells and its modulation by erythromycin. Cancer Chemother Pharmacol. 2005;56:212–220.
    1. Shinkai M, Tamaoki J, Kobayashi H, Kanoh S, Motoyoshi K, et al. Clarithromycin delays progression of bronchial epithelial cells from G1 phase to S phase and delays cell growth via extracellular signal-regulated protein kinase suppression. Antimicrob Agents Chemother. 2006;50:1738–1744.
    1. Shinkai M, Lopez-Boado YS, Rubin BK. Clarithromycin has an immunomodulatory effect on ERK-mediated inflammation induced by Pseudomonas aeruginosa flagellin. J Antimicrob Chemother. 2007;59:1096–1101.

Source: PubMed

Подписаться