期刊论文详细信息
BMC Pulmonary Medicine
Plasma Chemokine signature correlates with lung goblet cell hyperplasia in smokers with and without chronic obstructive pulmonary disease
Thomas J. Rogers3  Gerard J. Criner1  Heba Durra2  Michelle Oros2  William D. Cornwell3  Victor Kim1 
[1] Division of Pulmonary and Critical Care Medicine, Temple University School of Medicine, 3401 North Broad Street, 785 Parkinson Pavilion, Philadelphia 19140, PA, USA;Department of Pathology, Temple University School of Medicine, Philadelphia, PA, USA;Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, PA, USA
关键词: Macrophage;    Neutrophil;    Chemokine;    Goblet cell hyperplasia;    Chronic obstructive pulmonary disease;    Mucin;   
Others  :  1228329
DOI  :  10.1186/s12890-015-0103-2
 received in 2015-04-17, accepted in 2015-09-17,  发布年份 2015
PDF
【 摘 要 】

Background

Chronic Obstructive Pulmonary Disease (COPD) is characterized by lung and systemic inflammation as well as airway goblet cell hyperplasia (GCH). Mucin production is activated in part by stimulation of the epidermal growth factor (EGF) receptor pathway through neutrophils and macrophages. How circulating cytokine levels relate to GCH is not clear.

Methods

We performed phlebotomy and bronchoscopy on 25 subjects (six nonsmokers, 11 healthy smokers, and eight COPD subjects FEV 130–60 %). Six endobronchial biopsies per subject were performed. GCH was measured by measuring mucin volume density (MVD) using stereological techniques on periodic acid fast-Schiff stained samples. We measured the levels of chemokines CXCL8/IL-8, CCL2/MCP-1, CCL7/MCP-3, CCL22/MCD, CCL3/MIP-1α, and CCL4/MIP-1β, and the cytokines IL-1, IL-4, IL-6, IL-9, IL-17, EGF, and vascular endothelial growth factor (VEGF). Differences between groups were assessed using one-way ANOVA, t test, or Chi squared test. Post hoc tests after ANOVA were performed using Bonferroni correction.

Results

MVD was highest in healthy smokers (27.78 ± 10.24 μL/mm 2 ) compared to COPD subjects (16.82 ± 16.29 μL/mm 2 , p = 0.216) and nonsmokers (3.42 ± 3.07 μL/mm 2 , p <0.0001). Plasma CXCL8 was highest in healthy smokers (11.05 ± 8.92 pg/mL) compared to nonsmokers (1.20 ± 21.92 pg/mL, p = 0.047) and COPD subjects (6.01 ± 5.90 pg/mL, p = 0.366). CCL22 and CCL4 followed the same trends. There were no significant differences in the other cytokines measured. When the subjects were divided into current smokers (healthy smokers and COPD current smokers) and non/ex-smokers (nonsmokers and COPD ex-smokers), plasma CXCL8, CCL22, CCL4, and MVD were greater in current smokers. No differences in other cytokines were seen. Plasma CXCL8 moderately correlated with MVD (r = 0.552, p = 0.003).

Discussion

In this small cohort, circulating levels of the chemokines CXCL8, CCL4, and CCL22, as well as MVD, attain the highest levels in healthy smokers compared to nonsmokers and COPD subjects. These findings seem to be driven by current smoking and are independent of airflow obstruction.

Conclusions

These data suggest that smoking upregulates a systemic pattern of neutrophil and macrophage chemoattractant expression, and this correlates significantly with the development of goblet cell hyperplasia.

【 授权许可】

   
2015 Kim et al.

【 预 览 】
附件列表
Files Size Format View
20151015020505454.pdf 906KB PDF download
Fig. 4. 15KB Image download
Fig. 3. 19KB Image download
Fig. 2. 29KB Image download
Fig. 1. 78KB Image download
【 图 表 】

Fig. 1.

Fig. 2.

Fig. 3.

Fig. 4.

【 参考文献 】
  • [1]Vestbo J, Hurd SS, Agusti AG, Jones PW, Vogelmeier C, Anzueto A et al.. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013; 187(4):347-65.
  • [2]Kim V, Rogers TJ, Criner GJ. New concepts in the pathobiology of chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2008; 5(4):478-85.
  • [3]Saetta M, Turato G, Baraldo S, Zanin A, Braccioni F, Mapp CE et al.. Goblet cell hyperplasia and epithelial inflammation in peripheral airways of smokers with both symptoms of chronic bronchitis and chronic airflow limitation. Am J Respir Crit Care Med. 2000; 161(3 Pt 1):1016-21.
  • [4]Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L et al.. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004; 350(26):2645-53.
  • [5]Innes AL, Woodruff PG, Ferrando RE, Donnelly S, Dolganov GM, Lazarus SC et al.. Epithelial mucin stores are increased in the large airways of smokers with airflow obstruction. Chest. 2006; 130(4):1102-8.
  • [6]Sciurba F, Martinez FJ, Rogers RM, Make B, Criner GJ, Cherniak RM et al.. The effect of small airway pathology on survival following lung volume reduction surgery (LVRS). [abstract]. Proc Am Thorac Soc. 2006; 3:A712.
  • [7]REID LM. Pathology of chronic bronchitis. Lancet. 1954; 266(6806):274-8.
  • [8]Kim V, Couturie MJ, Wang P, Gaughan JP, Williams OW, Evans CM et al.. Small airway mucous metaplasia, histomorphometry, and inflammation in patients with advanced emphysema [abstract]. Proc Am Thorac Soc. 2006; 3:A629.
  • [9]Burgel PR, Nadel JA. Roles of epidermal growth factor receptor activation in epithelial cell repair and mucin production in airway epithelium. Thorax. 2004; 59(11):992-6.
  • [10]Kim V, Kelemen SE, Abuel-Haija M, Gaughan J, Sharafkhaneh A, Evans CM et al.. Small airway mucous metaplasia and inflammation in chronic obstructive pulmonary disease. J COPD. 2008; 5(6):329-38.
  • [11]Harkema JR, Plopper CG, Hyde DM, St George JA. Regional differences in quantities of histochemically detectable mucosubstances in nasal, paranasal, and nasopharyngeal epithelium of the bonnet monkey. J Histochem Cytochem. 1987; 35(3):279-86.
  • [12]Weibel ER. Stereological methods. Academic Press Inc. Ltd., London; 1979.
  • [13]Kim V, Oros M, Durra H, Kelsen S, Aksoy M, Cornwell WD et al.. Chronic bronchitis and current smoking are associated with more goblet cells in moderate to severe COPD and smokers without airflow obstruction. PLoS One. 2015; 10(2):e0116108.
  • [14]Barnes PJ. The cytokine network in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2009; 41(6):631-8.
  • [15]Hurst JR, Perera WR, Wilkinson TM, Donaldson GC, Wedzicha JA. Systemic and upper and lower airway inflammation at exacerbation of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006; 173(1):71-8.
  • [16]Dahl M, Vestbo J, Lange P, Bojesen SE, Tybjaerg-Hansen A, Nordestgaard BG. C-reactive protein as a predictor of prognosis in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007; 175(3):250-5.
  • [17]Hurst JR, Donaldson GC, Perera WR, Wilkinson TM, Bilello JA, Hagan GW et al.. Use of plasma biomarkers at exacerbation of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006; 174(8):867-74.
  • [18]Sin DD, Lacy P, York E, Man SF. Effects of fluticasone on systemic markers of inflammation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2004; 170(7):760-5.
  • [19]Sin DD, Man SF, Marciniuk DD, Ford G, FitzGerald M, Wong E et al.. The effects of fluticasone with or without salmeterol on systemic biomarkers of inflammation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008; 177(11):1207-14.
  • [20]Richman-Eisenstat JB, Jorens PG, Hebert CA, Ueki I, Nadel JA. Interleukin-8: an important chemoattractant in sputum of patients with chronic inflammatory airway diseases. Am J Physiol. 1993; 264(4 Pt 1):L413-8.
  • [21]Park JA, He F, Martin LD, Li Y, Chorley BN, Adler KB. Human neutrophil elastase induces hypersecretion of mucin from well-differentiated human bronchial epithelial cells in vitro via a protein kinase C{delta}-mediated mechanism. Am J Pathol. 2005; 167(3):651-61.
  • [22]Bautista MV, Chen Y, Ivanova VS, Rahimi MK, Watson AM, Rose MC. IL-8 regulates mucin gene expression at the posttranscriptional level in lung epithelial cells. J Immunol. 2009; 183(3):2159-66.
  • [23]Tumkaya M, Atis S, Ozge C, Delialioglu N, Polat G, Kanik A. Relationship between airway colonization, inflammation and exacerbation frequency in COPD. Respir Med. 2007; 101(4):729-37.
  • [24]Celli BR, Locantore N, Yates J, Tal-Singer R, Miller BE, Bakke P et al.. Inflammatory biomarkers improve clinical prediction of mortality in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012; 185(10):1065-72.
  • [25]Pinto-Plata V, Toso J, Lee K, Park D, Bilello J, Mullerova H et al.. Profiling serum biomarkers in patients with COPD: associations with clinical parameters. Thorax. 2007; 62(7):595-601.
  • [26]Agusti A, Edwards LD, Rennard SI, MacNee W, Tal-Singer R, Miller BE et al.. Persistent systemic inflammation is associated with poor clinical outcomes in COPD: a novel phenotype. PLoS One. 2012; 7(5):e37483.
  • [27]Ji J, von Scheele I, Bergstrom J, Billing B, Dahlen B, Lantz AS et al.. Compartment differences of inflammatory activity in chronic obstructive pulmonary disease. Respir Res. 2014; 15:104,014-0104-3.
  • [28]Brozyna S, Ahern J, Hodge G, Nairn J, Holmes M, Reynolds PN et al.. Chemotactic mediators of Th1 T-cell trafficking in smokers and COPD patients. COPD. 2009; 6(1):4-16.
  • [29]Tanino M, Betsuyaku T, Takeyabu K, Tanino Y, Yamaguchi E, Miyamoto K et al.. Increased levels of interleukin-8 in BAL fluid from smokers susceptible to pulmonary emphysema. Thorax. 2002; 57(5):405-11.
  • [30]Mio T, Romberger DJ, Thompson AB, Robbins RA, Heires A, Rennard SI. Cigarette smoke induces interleukin-8 release from human bronchial epithelial cells. Am J Respir Crit Care Med. 1997; 155(5):1770-6.
  • [31]Hoonhorst S, Timens W, Koenderman L, Lo Tam Loi AT, Lammers JW, Boezen H et al.. Increased activation of blood neutrophils after cigarette smoking in young individuals susceptible to COPD. Respir Res. 2014; 15(1):121. BioMed Central Full Text
  • [32]Riise GC, Ahlstedt S, Larsson S, Enander I, Jones I, Larsson P et al.. Bronchial inflammation in chronic bronchitis assessed by measurement of cell products in bronchial lavage fluid. Thorax. 1995; 50(4):360-5.
  • [33]Khurana S, Ravi A, Sutula J, Milone R, Williamson R, Plumb J, et al. Clinical characteristics and airway inflammation profile in COPD persistent sputum producers. Respir Med. 2014. doi:. 10. 1016/j.rmed.2014.09.020 webcite
  • [34]Monzon ME, Forteza RM, Casalino-Matsuda SM. MCP-1/CCR2B-dependent loop upregulates MUC5AC and MUC5B in human airway epithelium. Am J Physiol Lung Cell Mol Physiol. 2011; 300(2):L204-15.
  • [35]de Moraes MR, da Costa AC, Correa Kde S, Junqueira-Kipnis AP, Rabahi MF. Interleukin-6 and interleukin-8 blood levels’ poor association with the severity and clinical profile of ex-smokers with COPD. Int J Chron Obstruct Pulmon Dis. 2014; 9:735-43.
  • [36]Yamashita U, Kuroda E. Regulation of macrophage-derived chemokine (MDC, CCL22) production. Crit Rev Immunol. 2002; 22(2):105-14.
  • [37]Ying S, O’Connor B, Ratoff J, Meng Q, Fang C, Cousins D et al.. Expression and cellular provenance of thymic stromal lymphopoietin and chemokines in patients with severe asthma and chronic obstructive pulmonary disease. J Immunol. 2008; 181(4):2790-8.
  • [38]Yogo Y, Fujishima S, Inoue T, Saito F, Shiomi T, Yamaguchi K et al.. Macrophage derived chemokine (CCL22), thymus and activation-regulated chemokine (CCL17), and CCR4 in idiopathic pulmonary fibrosis. Respir Res. 2009; 10:80,9921-10-80. BioMed Central Full Text
  • [39]Frankenberger M, Eder C, Hofer TP, Heimbeck I, Skokann K, Kassner G et al.. Chemokine expression by small sputum macrophages in COPD. Mol Med. 2011; 17(7–8):762-70.
  • [40]Belperio JA, Dy M, Murray L, Burdick MD, Xue YY, Strieter RM et al.. The role of the Th2 CC chemokine ligand CCL17 in pulmonary fibrosis. J Immunol. 2004; 173(7):4692-8.
  • [41]Inoue T, Fujishima S, Ikeda E, Yoshie O, Tsukamoto N, Aiso S et al.. CCL22 and CCL17 in rat radiation pneumonitis and in human idiopathic pulmonary fibrosis. Eur Respir J. 2004; 24(1):49-56.
  • [42]Ma B, Kang MJ, Lee CG, Chapoval S, Liu W, Chen Q et al.. Role of CCR5 in IFN-gamma-induced and cigarette smoke-induced emphysema. J Clin Invest. 2005; 115(12):3460-72.
  • [43]Bracke KR, D’hulst AI, Maes T, Demedts IK, Moerloose KB, Kuziel WA et al.. Cigarette smoke-induced pulmonary inflammation, but not airway remodelling, is attenuated in chemokine receptor 5-deficient mice. Clin Exp Allergy. 2007; 37(10):1467-79.
  • [44]Meuronen A, Majuri ML, Alenius H, Mantyla T, Wolff H, Piirila P et al.. Decreased cytokine and chemokine mRNA expression in bronchoalveolar lavage in asymptomatic smoking subjects. Respiration. 2008; 75(4):450-8.
  • [45]Capelli A, Di Stefano A, Gnemmi I, Balbo P, Cerutti CG, Balbi B et al.. Increased MCP-1 and MIP-1beta in bronchoalveolar lavage fluid of chronic bronchitics. Eur Respir J. 1999; 14(1):160-5.
  • [46]D’Ambrosio D, Panina-Bordignon P, Sinigaglia F. Chemokine receptors in inflammation: an overview. J Immunol Methods. 2003; 273(1–2):3-13.
  文献评价指标  
  下载次数:4次 浏览次数:2次