Activation and polarization of circulating monocytes in severe chronic obstructive pulmonary disease

William D Cornwell, Victor Kim, Xiaoxuan Fan, Marie Elena Vega, Frederick V Ramsey, Gerard J Criner, Thomas J Rogers, William D Cornwell, Victor Kim, Xiaoxuan Fan, Marie Elena Vega, Frederick V Ramsey, Gerard J Criner, Thomas J Rogers

Abstract

Background: The ability of circulating monocytes to develop into lung macrophages and promote lung tissue damage depends upon their phenotypic pattern of differentiation and activation. Whether this phenotypic pattern varies with COPD severity is unknown. Here we characterize the activation and differentiation status of circulating monocytes in patients with moderate vs. severe COPD.

Methods: Blood monocytes were isolated from normal non-smokers (14), current smokers (13), patients with moderate (9), and severe COPD (11). These cells were subjected to analysis by flow cytometry to characterize the expression of activation markers, chemoattractant receptors, and surface markers characteristic of either M1- or M2-type macrophages.

Results: Patients with severe COPD had increased numbers of total circulating monocytes and non-classical patrolling monocytes, compared to normal subjects and patients with moderate COPD. In addition, while the percentage of circulating monocytes that expressed an M2-like phenotype was reduced in patients with either moderate or severe disease, the levels of expression of M2 markers on this subpopulation of monocytes in severe COPD was significantly elevated. This was particularly evident for the expression of the chemoattractant receptor CCR5.

Conclusions: Blood monocytes in severe COPD patients undergo unexpected pre-differentiation that is largely characteristic of M2-macrophage polarization, leading to the emergence of an unusual M2-like monocyte population with very high levels of CCR5. These results show that circulating monocytes in patients with severe COPD possess a cellular phenotype which may permit greater mobilization to the lung, with a pre-existing bias toward a potentially destructive inflammatory phenotype.

Keywords: COPD; Monocyte activation; Polarization; Systemic inflammation.

Conflict of interest statement

Ethics approval and consent to participate

The study was conducted in accordance with the amended Declaration of Helsinki. Institutional Review Board approval was obtained from the Temple University Institutional Review Board, and all subjects signed written informed consent.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Representative flow cytometric analyses for PBMCs. Normal (a), smoker (b), moderate COPD (COPD-M) (c), and severe COPD (COPD-S) (d) were stained for expression of CD14 and CD16. Based on staining intensity, classical monocytes (CD14 + CD16-), intermediate monocytes (CD14 + CD16+), and non-classical monocytes (CD14DIMCD16+), as well as the total numbers of monocytes were identified
Fig. 2
Fig. 2
The numbers of peripheral blood monocytes are significantly increased in severe COPD patients. The total number of blood monocytes (a), and the numbers of classical monocytes (b), intermediate monocytes (c) and non-classical monocytes (d) cells are presented. The percentages of classical monocytes and non-classical monocytes are also presented. The percentages of each population were determined relative to the total number of monocytes, and data are presented for the classical (e), non-classical (f), and intermediate (g) monocyte sub-populations. Data are presented as box plots with the mean (red line) and median (black line). The box delineates the interquartile range, and the vertical line represents the interquartile range. * = p < 0.05 and ** = p < 0.01 relative to the normal
Fig. 3
Fig. 3
Altered composition of monocyte sub-populations in smokers and COPD patients. Classical (a, c, e, and g) and non-classical (b, d, f, h) monocytes were stained for CD163 (a, b), CD25 (c, d), CD206 (e, f), and IL-13Rα1 (g, h) expression. The data are presented as the percentage of total classical or non-classical monocytes for each group. *** = p < 0.001 are relative to the normal
Fig. 4
Fig. 4
Increased CD163 and CD206 expression density in classical and non-classical monocytes in COPD patients. Classical (a, c, e) and non-classical (b, d, f) monocytes were stained for CD163 (a, b), CD206 (c, d), and CD25 (e, f) expression. The degree of expression is reported as the MFI
Fig. 5
Fig. 5
Reduced numbers of CD206 + CCR5+ monocytes with increased inflammatory phenotype in severe COPD. CD206 + CCR5+ classical (a, c, e, g) and CD206 + CCR5+ non-classical (b, d, f, h) monocytes were also stained for CD14 (c, d), CD163 (e, f) and CCR5 (g, h) expression. The degree of expression is reported as the MFI
Fig. 6
Fig. 6
Elevated expression of CCR5 in CD206 + CCR5+ monocytes in severe COPD. Panels a and b are representative histograms of CCR5 expression on CD206 + CCR5+ classical and non-classical monocytes shown in Fig. 5. Results are representative of the 11 COPD-S patients

References

    1. Grashoff WF, Sont JK, Sterk PJ, Hiemstra PS, de Boer WI, Stolk J, Han J, van Krieken JM. Chronic obstructive pulmonary disease: role of bronchiolar mast cells and macrophages. AmJPathol 1997; 151:1785–1790.
    1. Finkelstein R, Fraser RS, Ghezzo H, Cosio MG, Finkelstein R, Fraser RS, Ghezzo H, Cosio MG. Alveolar inflammation and its relation to emphysema in smokers. American Journal of Respiratory & Critical Care Medicine. 1995;152:1666–1672. doi: 10.1164/ajrccm.152.5.7582312.
    1. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. NEnglJMed. 2004;350:2645–2653. doi: 10.1056/NEJMoa032158.
    1. Hiemstra PS. Altered macrophage function in chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2013;10(Suppl):S180–S185. doi: 10.1513/AnnalsATS.201305-123AW.
    1. Barnes PJ. Alveolar macrophages as orchestrators of COPD. COPD: J Chron Obstruct Pulmon Dis. 2004;1:59–70. doi: 10.1081/COPD-120028701.
    1. van de Laar L, Saelens W, De Prijck S, Martens L, Scott CL, Van Isterdael G, Hoffmann E, Beyaert R, Saeys Y, Lambrecht BN, et al. Yolk sac macrophages, fetal liver, and adult monocytes can colonize an empty niche and develop into functional tissue-resident macrophages. Immunity. 2016;44:755–768. doi: 10.1016/j.immuni.2016.02.017.
    1. Yona S, Kim KW, Wolf Y, Mildner A, Varol D, Breker M, Strauss-Ayali D, Viukov S, Guilliams M, Misharin A, et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity. 2013;38:79–91. doi: 10.1016/j.immuni.2012.12.001.
    1. Kopf M, Schneider C, Nobs SP. The development and function of lung-resident macrophages and dendritic cells. Nat Immunol. 2015;16:36–44. doi: 10.1038/ni.3052.
    1. Guilliams M, De Kleer I, Henri S, Post S, Vanhoutte L, De Prijck S, Deswarte K, Malissen B, Hammad H, Lambrecht BN. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J Exp Med. 2013;210:1977–1992. doi: 10.1084/jem.20131199.
    1. Misharin AV, Morales-Nebreda L, Reyfman PA, Cuda CM, Walter JM, McQuattie-Pimentel AC, Chen CI, Anekalla KR, Joshi N, Williams KJN, et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J Exp Med. 2017;214:2387–2404. doi: 10.1084/jem.20162152.
    1. Martinez FO, Gordon S, Locati M, Mantovani A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol. 2006;177:7303–7311. doi: 10.4049/jimmunol.177.10.7303.
    1. Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol. 2009;27:451–483. doi: 10.1146/annurev.immunol.021908.132532.
    1. Rees AJ. Monocyte and macrophage biology: an overview. Semin Nephrol. 2010;30:216–233. doi: 10.1016/j.semnephrol.2010.03.002.
    1. Auffray C, Sieweke MH, Geissmann F. Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol. 2009;27:669–692. doi: 10.1146/annurev.immunol.021908.132557.
    1. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, Leenen PJ, Liu YJ, MacPherson G, Randolph GJ, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116:e74-e80. doi: 10.1182/blood-2010-02-258558.
    1. Nockher WA, Scherberich JE. Expanded CD14+ CD16+ monocyte subpopulation in patients with acute and chronic infections undergoing hemodialysis. Infect Immun. 1998;66:2782–2790.
    1. Fingerle G, Pforte A, Passlick B, Blumenstein M, Strobel M, Ziegler-Heitbrock HW. The novel subset of CD14+/CD16+ blood monocytes is expanded in sepsis patients. Blood. 1993;82:3170–3176.
    1. Skinner NA, MacIsaac CM, Hamilton JA, Visvanathan K. Regulation of toll-like receptor (TLR)2 and TLR4 on CD14dimCD16+ monocytes in response to sepsis-related antigens. Clin Exp Immunol. 2005;141:270–278. doi: 10.1111/j.1365-2249.2005.02839.x.
    1. Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003;19:71–82. doi: 10.1016/S1074-7613(03)00174-2.
    1. Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, Sarnacki S, Cumano A, Lauvau G, Geissmann F, et al. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science. 2007;317:666–670. doi: 10.1126/science.1142883.
    1. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nature Reviews. Immunology. 2008;8:183–192.
    1. Vlahos R, Bozinovski S. Role of alveolar macrophages in chronic obstructive pulmonary disease. Front Immunol. 2014;5:435. doi: 10.3389/fimmu.2014.00435.
    1. Bozinovski S, Cross M, Vlahos R, Jones JE, Hsuu K, Tessier PA, Reynolds EC, Hume DA, Hamilton JA, Geczy CL, et al. S100A8 chemotactic protein is abundantly increased, but only a minor contributor to LPS-induced, steroid resistant neutrophilic lung inflammation in vivo. J Proteome Res. 2005;4:136–145. doi: 10.1021/pr049829t.
    1. Kaku Y, Imaoka H, Morimatsu Y, Komohara Y, Ohnishi K, Oda H, Takenaka S, Matsuoka M, Kawayama T, Takeya M, et al. Overexpression of CD163, CD204 and CD206 on alveolar macrophages in the lungs of patients with severe chronic obstructive pulmonary disease. PLoS One. 2014;9:e87400. doi: 10.1371/journal.pone.0087400.
    1. Shapiro SD. The macrophage in chronic obstructive pulmonary disease. AmJRespirCrit Care Med. 1999;160:S29–S32. doi: 10.1164/ajrccm.160.supplement_1.9.
    1. Pesci A, Balbi B, Majori M, Cacciani G, Bertacco S, Alciato P, Donner CF. Inflammatory cells and mediators in bronchial lavage of patients with chronic obstructive pulmonary disease. EurRespirJ. 1998;12:380–386.
    1. Keatings VM, Collins PD, Scott DM, Barnes PJ. Differences in interleukin-8 and tumor necrosis factor-alpha in induced sputum from patients with chronic obstructive pulmonary disease or asthma. Am J Respir Crit Care Med. 1996;153:530–534. doi: 10.1164/ajrccm.153.2.8564092.
    1. Retamales I, Elliott WM, Meshi B, Coxson HO, Pare PD, Sciurba FC, Rogers RM, Hayashi S, Hogg JC, Retamales I, et al. Amplification of inflammation in emphysema and its association with latent adenoviral infection. American Journal of Respiratory & Critical Care Medicine. 2001;164:469–473. doi: 10.1164/ajrccm.164.3.2007149.
    1. Xiong Z, Leme AS, Ray P, Shapiro SD, Lee JS. CX3CR1+ lung mononuclear phagocytes spatially confined to the interstitium produce TNF-alpha and IL-6 and promote cigarette smoke-induced emphysema. J Immunol. 2011;186:3206–3214. doi: 10.4049/jimmunol.1003221.
    1. Landsman L, Varol C, Jung S. Distinct differentiation potential of blood monocyte subsets in the lung. J Immunol 2007; 178:2000–2007.
    1. Carlin LM, Stamatiades EG, Auffray C, Hanna RN, Glover L, Vizcay-Barrena G, Hedrick CC, Cook HT, Diebold S, Geissmann F. Nr4a1-dependent Ly6C(low) monocytes monitor endothelial cells and orchestrate their disposal. Cell. 2013;153:362–375. doi: 10.1016/j.cell.2013.03.010.
    1. Jakubzick C, Gautier EL, Gibbings SL, Sojka DK, Schlitzer A, Johnson TE, Ivanov S, Duan Q, Bala S, Condon T, et al. Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes. Immunity. 2013;39:599–610. doi: 10.1016/j.immuni.2013.08.007.
    1. Landsman L, Jung S. Lung macrophages serve as obligatory intermediate between blood monocytes and alveolar macrophages. J Immunol. 2007;179:3488–3494. doi: 10.4049/jimmunol.179.6.3488.
    1. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41:14–20. doi: 10.1016/j.immuni.2014.06.008.
    1. Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol. 2014;14:81–93. doi: 10.1038/nri3600.
    1. Abdullah M, Kahler D, Vock C, Reiling N, Kugler C, Dromann D, Rupp J, Hauber HP, Fehrenbach H, Zabel P, et al. Pulmonary haptoglobin and CD163 are functional immunoregulatory elements in the human lung. Respiration. 2012;83:61–73. doi: 10.1159/000329868.
    1. Fabriek BO, van Bruggen R, Deng DM, Ligtenberg AJ, Nazmi K, Schornagel K, Vloet RP, Dijkstra CD, van den Berg TK. The macrophage scavenger receptor CD163 functions as an innate immune sensor for bacteria. Blood. 2009;113:887–892. doi: 10.1182/blood-2008-07-167064.
    1. Baraldo S, Bazzan E, Zanin ME, Turato G, Garbisa S, Maestrelli P, Papi A, Miniati M, Fabbri LM, Zuin R, et al. Matrix metalloproteinase-2 protein in lung periphery is related to COPD progression. Chest. 2007;132:1733–1740. doi: 10.1378/chest.06-2819.
    1. Shaykhiev R, Krause A, Salit J, Strulovici-Barel Y, Harvey BG, O'Connor TP, Crystal RG. Smoking-dependent reprogramming of alveolar macrophage polarization: implication for pathogenesis of chronic obstructive pulmonary disease. J Immunol. 2009;183:2867–2883. doi: 10.4049/jimmunol.0900473.
    1. Taylor AE, Finney-Hayward TK, Quint JK, Thomas CM, Tudhope SJ, Wedzicha JA, Barnes PJ, Donnelly LE. Defective macrophage phagocytosis of bacteria in COPD. European Respiratory J. 2010;35:1039–1047. doi: 10.1183/09031936.00036709.
    1. Berenson CS, Garlipp MA, Grove LJ, Maloney J, Sethi S. Impaired phagocytosis of nontypeable Haemophilus influenzae by human alveolar macrophages in chronic obstructive pulmonary disease. J Infect Dis. 2006;194:1375–1384. doi: 10.1086/508428.
    1. Price JV, Vance RE. The macrophage paradox. Immunity. 2014;41:685–693. doi: 10.1016/j.immuni.2014.10.015.
    1. Ma B, Kang MJ, Lee CG, Chapoval S, Liu W, Chen Q, Coyle AJ, Lora JM, Picarella D, Homer RJ, et al. Role of CCR5 in IFN-gamma-induced and cigarette smoke-induced emphysema. J Clin Invest. 2005;115:3460–3472. doi: 10.1172/JCI24858.
    1. Bracke KR, D'Hulst AI, Maes T, Demedts IK, Moerloose KB, Kuziel WA, Joos GF, Brusselle GG. Cigarette smoke-induced pulmonary inflammation, but not airway remodelling, is attenuated in chemokine receptor 5-deficient mice. Clin Exp Allergy. 2007;37:1467–1479.
    1. Costa C, Traves SL, Tudhope SJ, Fenwick PS, Belchamber KB, Russell RE, Barnes PJ, Donnelly LE. Enhanced monocyte migration to CXCR3 and CCR5 chemokines in COPD. Eur Respir J. 2016;47:1093–1102. doi: 10.1183/13993003.01642-2015.
    1. Costa C, Rufino R, Traves SL, Lapa ESJR, Barnes PJ, Donnelly LE. CXCR3 and CCR5 chemokines in induced sputum from patients with COPD. Chest. 2008;133:26–33. doi: 10.1378/chest.07-0393.

Source: PubMed

3
Abonner