Characterisation of lung macrophage subpopulations in COPD patients and controls

Jennifer A Dewhurst, Simon Lea, Elizabeth Hardaker, Josiah V Dungwa, Arjun K Ravi, Dave Singh, Jennifer A Dewhurst, Simon Lea, Elizabeth Hardaker, Josiah V Dungwa, Arjun K Ravi, Dave Singh

Abstract

Lung macrophage subpopulations have been identified based on size. We investigated characteristics of small and large macrophages in the alveolar spaces and lung interstitium of COPD patients and controls. Alveolar and interstitial cells were isolated from lung resection tissue from 88 patients. Macrophage subpopulation cell-surface expression of immunological markers and phagocytic ability were assessed by flow cytometry. Inflammatory related gene expression was measured. Alveolar and interstitial macrophages had subpopulations of small and large macrophages based on size and granularity. Alveolar macrophages had similar numbers of small and large cells; interstitial macrophages were mainly small. Small macrophages expressed significantly higher cell surface HLA-DR, CD14, CD38 and CD36 and lower CD206 compared to large macrophages. Large alveolar macrophages showed lower marker expression in COPD current compared to ex-smokers. Small interstitial macrophages had the highest pro-inflammatory gene expression levels, while large alveolar macrophages had the lowest. Small alveolar macrophages had the highest phagocytic ability. Small alveolar macrophage CD206 expression was lower in COPD patients compared to smokers. COPD lung macrophages include distinct subpopulations; Small interstitial and small alveolar macrophages with more pro-inflammatory and phagocytic function respectively, and large alveolar macrophages with low pro-inflammatory and phagocytic ability.

Conflict of interest statement

D. Singh has received sponsorship to attend international meetings, honoraria for lecturing or attending advisory boards and research grants from various pharmaceutical companies including Almirall, AstraZeneca, Boehringer Ingelheim, Chiesi, Genentech, GlaxoSmithKline, Glenmark, Johnson and Johnson, Merck, NAPP, Novartis, Pfizer, Skypharma, Takeda, Teva, Therevance and Verona.E. Hardaker was employed by Novartis during the undertaking of this project. S. Lea, J. Dewhurst, J. Dungwa and A. Ravi declare that they have no competing interests

Figures

Figure 1
Figure 1
Flow chart describing subsets of patients. a = cells from the same COPD patients used for identifying macrophage subpopulations were used for these experiments, b = cells from the same smoking controls used for measuring cell surface markers on alveolar macrophages were used for these experiments. S: smoking controls, NS: never smokers.
Figure 2
Figure 2
Identifying macrophage subpopulations in alveolar and interstitial macrophages. Alveolar macrophage (A,B and C) and interstitial macrophage (D,E and F) subpopulations were compared and data is shown of enriched macrophages from an ex smoking COPD patient. Flow cytometry pseudocolor plots (A and D and density plots (B and E) indicating size (FSC) and granularity (SSC) were used to identify small (a) and large (b) macrophage subpopulations. H&E stained cytospins (C and F) were used to confirm the presence of small (a) and large (b) macrophages. The number of small and large macrophages present was expressed as a percentage of the total macrophage population (G). Data represents mean (SEM) of n = 18 COPD patients. Paired t test (two tailed) was performed. ***The percentage of large interstitial macrophages is significantly decreased compared to the percentage of large alveolar macrophages (p < 0.001).
Figure 3
Figure 3
The expression of macrophage markers in small macrophages compared to large macrophages. Flow cytometric analysis of enriched alveolar (A and B) and interstitial (C and D) macrophages for n = 13 and n = 11 COPD patients respectively per marker. Markers analysed were HLA-DR, CD14, CD38, CD36, CD206 and CD163. Data is expressed as the percentage of cells within each subpopulation expressing a specific marker (A and C) and the median fluorescence intensity (MFI) of each marker (B and D) and represents mean (SEM). Paired t test (two tailed) was performed for each marker. *,**,***Significantly increased between size populations within tissue compartment (p < 0.05, 0.01 and 0.001 respectively). #,##,###Significantly increased between tissue compartments for each size population (p < 0.05, 0.01 and 0.001 respectively)
Figure 4
Figure 4
The expression of macrophage markers in the large alveolar macrophage population in COPD (including smoking status). Flow cytometric analysis of large alveolar macrophages for n = 7 COPD smokers (COPDS) and n = 6 COPD ex-smokers (COPDE). Markers analysed were HLA-DR (A), CD206 (B), CD163 (C), CD14 (D), CD38 (E) and CD36 (F). Data is expressed as the median fluorescence intensity (MFI) of each marker and represents mean (SEM). Unpaired t test (Two-tailed) was performed for each marker. ***Significantly different compared to expression of the same marker on large alveolar macrophages from COPDE patients (p < 0.001).
Figure 5
Figure 5
The diameter of alveolar and interstitial macrophages in lung resected tissue. Alveolar (AM) and interstitial (IM) macrophages from never smokers (NS) (n = 11) (A), smoking controls (S) (n = 15) (B) and COPD patients (COPD) (n = 12) (C) were identified in FFPE resected lung tissue by co-expression of CX3CR1 and CD14 and the diameter of each cell was measured. Data are shown as the percentage of macrophage population measuring a certain diameter (A-C) or individual cell diameters for IMs (D) and AMs (E) with median and interquartile range. Mann-whitney t-tests were performed between IM and AMs. One way ANOVA followed by Tukey-Kramer Multiple Comparisons Test were performed between subject groups. **,***Significant difference (p < 0.01, < 0.001 respectively)
Figure 6
Figure 6
Expression of CXCL1 and TLR3 protein and phagocytosis of pHrodo E. coli BioParticles in macrophage subpopulations. Alveolar (AM) and interstitial (IM) macrophages were isolated using EasySep monocyte enrichment kit. Cells were used to generate cytospin slides (A and B) or exposed to pHrodo E. coli BioParticles for 1 h in a shaking incubator (C and D). Percentage of small alveolar, large alveolar and small interstitial macrophages expressing CXCL1 protein and TLR3 protein are shown in (A and B) respectively. Data represents mean (SEM) of n = 6 COPD patients. Phagocytosis was analysed by flow cytometry and is represented by (C): % of pHrodo bright macrophages within each subpopulation and (D): median fluorescence intensity (MFI) of each subpopulation relative to the negative control. Data are shown as mean (SEM) of n = 9 COPD patients One way ANOVA followed by Tukey-Kramer Multiple Comparisons Test was performed. *,**,***Significantly different compared to expression of the same protein on large alveolar macrophages (p < 0.05, < 0.01 and <0.001 respectively). #,##Significantly different compared to the percentage of pHrodo bright macrophages or MFI of other macrophage subpopulations (p < 0.05, <0.01 respectively).

References

    1. Vogelmeier CF, et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease 2017 Report. GOLD Executive Summary. Am J Respir Crit Care Med. 2017;195:557–582. doi: 10.1164/rccm.201701-0218PP.
    1. Hogg JC, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:2645–2653. doi: 10.1056/NEJMoa032158.
    1. Di Stefano A, et al. Severity of airflow limitation is associated with severity of airway inflammation in smokers. Am J Respir Crit Care Med. 1998;158:1277–1285. doi: 10.1164/ajrccm.158.4.9802078.
    1. Fels AO, Cohn ZA. The alveolar macrophage. J Appl Physiol. 1986;60:353–369.
    1. Hoppstadter, J. et al. Differential cell reaction upon Toll-like receptor 4 and 9 activation in human alveolar and lung interstitial macrophages. Respir Res 11, doi:10.1186/1465-9921-11-124 (2010).
    1. Porcheray F, et al. Macrophage activation switching: an asset for the resolution of inflammation. Clin Exp Immunol. 2005;142:481–489.
    1. Murray PJ, et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41:14–20. doi: 10.1016/j.immuni.2014.06.008.
    1. Savage ND, et al. Human anti-inflammatory macrophages induce Foxp3+ GITR+ CD25+ regulatory T cells, which suppress via membrane-bound TGFbeta-1. J Immunol. 2008;181:2220–2226. doi: 10.4049/jimmunol.181.3.2220.
    1. Cao H, Wolff RG, Meltzer MS, Crawford RM. Differential regulation of class II MHC determinants on macrophages by IFN-gamma and IL-4. J Immunol. 1989;143:3524–3531.
    1. Muller U, et al. IL-13 induces disease-promoting type 2 cytokines, alternatively activated macrophages and allergic inflammation during pulmonary infection of mice with Cryptococcus neoformans. J Immunol. 2007;179:5367–5377. doi: 10.4049/jimmunol.179.8.5367.
    1. Martinez, F. O. & Gordon, S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000prime reports6, doi:10.12703/P6-13 (2014).
    1. Hume, D. A. The Many Alternative Faces of Macrophage Activation. Frontiers in immunology6, doi:10.3389/fimmu.2015.00370 (2015).
    1. Gomez Perdiguero E, et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 2015;518:547–551. doi: 10.1038/nature13989.
    1. Ravi, A. K. et al. COPD monocytes demonstrate impaired migratory ability. Respir Res18, doi:10.1186/s12931-017-0569-y (2017).
    1. Janssen WJ, et al. Fas determines differential fates of resident and recruited macrophages during resolution of acute lung injury. Am J Respir Crit Care Med. 2011;184:547–560. doi: 10.1164/rccm.201011-1891OC.
    1. Desch AN, et al. Flow Cytometric Analysis of Mononuclear Phagocytes in Nondiseased Human Lung and Lung-Draining Lymph Nodes. Am J Respir Crit Care Med. 2016;193:614–626. doi: 10.1164/rccm.201507-1376OC.
    1. Chen H, Cowan MJ, Hasday JD, Vogel SN, Medvedev AE. Tobacco smoking inhibits expression of proinflammatory cytokines and activation of IL-1R-associated kinase, p38, and NF-kappaB in alveolar macrophages stimulated with TLR2 and TLR4 agonists. J Immunol. 2007;179:6097–6106. doi: 10.4049/jimmunol.179.9.6097.
    1. Kent LM, et al. Inhibition of lipopolysaccharide-stimulated chronic obstructive pulmonary disease macrophage inflammatory gene expression by dexamethasone and the p38 mitogen-activated protein kinase inhibitor N-cyano-N’-(2-{[8-(2,6-difluorophenyl)-4-(4-fluoro-2-methylphenyl)-7-oxo-7,8-dihydropyrido[2,3-d] pyrimidin-2-yl]amino}ethyl)guanidine (SB706504) J Pharmacol Exp Ther. 2009;328:458–468. doi: 10.1124/jpet.108.142950.
    1. Shaykhiev R, et al. 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. Hodge S, Hodge G, Scicchitano R, Reynolds PN, Holmes M. Alveolar macrophages from subjects with chronic obstructive pulmonary disease are deficient in their ability to phagocytose apoptotic airway epithelial cells. Immunology and cell biology. 2003;81:289–296. doi: 10.1046/j.1440-1711.2003.t01-1-01170.x.
    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. Frankenberger M, et al. Chemokine expression by small sputum macrophages in COPD. Molecular medicine. 2011;17:762–770. doi: 10.2119/molmed.2010.00202.
    1. Frankenberger M, et al. Characterization of a population of small macrophages in induced sputum of patients with chronic obstructive pulmonary disease and healthy volunteers. Clin Exp Immunol. 2004;138:507–516. doi: 10.1111/j.1365-2249.2004.02637.x.
    1. Krombach F, et al. Characterization and quantification of alveolar monocyte-like cells in human chronic inflammatory lung disease. Eur Respir J. 1996;9:984–991. doi: 10.1183/09031936.96.09050984.
    1. Duan M, et al. Distinct macrophage subpopulations characterize acute infection and chronic inflammatory lung disease. J Immunol. 2012;189:946–955. doi: 10.4049/jimmunol.1200660.
    1. Wright AK, et al. Pivotal Advance: Expansion of small sputum macrophages in CF: failure to express MARCO and mannose receptors. J Leukoc Biol. 2009;86:479–489. doi: 10.1189/jlb.1108699.
    1. Chana, K. K., Fenwick, P. S., Nicholson, A. G., Barnes, P. J. & Donnelly, L. E. Identification of a distinct glucocorticosteroid-insensitive pulmonary macrophage phenotype in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol133, 207-216.e201-211 (2014).
    1. Armstrong J, Sargent C, Singh D. Glucocorticoid sensitivity of lipopolysaccharide-stimulated chronic obstructive pulmonary disease alveolar macrophages. Clin Exp Immunol. 2009;158:74–83. doi: 10.1111/j.1365-2249.2009.03986.x.
    1. Lea S, et al. The effect of PPAR-gamma ligands on in vitro and in vivo models of COPD. Eur Respir J. 2013;43(2):409–20. doi: 10.1183/09031936.00187812.
    1. Dalman MR, Deeter A, Nimishakavi G, Duan ZH. Fold change and p-value cutoffs significantly alter microarray interpretations. BMC bioinformatics. 2012;13(Suppl):2.
    1. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–969. doi: 10.1038/nri2448.
    1. Mantovani A, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends in immunology. 2004;25:677–686. doi: 10.1016/j.it.2004.09.015.
    1. Lipscomb MF, et al. Human alveolar macrophages: HLA-DR-positive macrophages that are poor stimulators of a primary mixed leukocyte reaction. J Immunol. 1986;136:497–504.
    1. Costabel U, Bross KJ, Andreesen R, Matthys H. HLA-DR antigens on human macrophages from bronchoalveolar lavage fluid. Thorax. 1986;41:261–265. doi: 10.1136/thx.41.4.261.
    1. Wright SD, Ramos RA, Tobias PS, Ulevitch RJ, Mathison JC. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science. 1990;249:1431–1433. doi: 10.1126/science.1698311.
    1. Kang J, et al. The role of CD38 in Fcgamma receptor (FcgammaR)-mediated phagocytosis in murine macrophages. J Biol Chem. 2012;287:14502–14514. doi: 10.1074/jbc.M111.329003.
    1. Savill J, Hogg N, Ren Y, Haslett C. Thrombospondin cooperates with CD36 and the vitronectin receptor in macrophage recognition of neutrophils undergoing apoptosis. J Clin Invest. 1992;90:1513–1522. doi: 10.1172/JCI116019.
    1. Gibbings SL, et al. Transcriptome analysis highlights the conserved difference between embryonic and postnatal-derived alveolar macrophages. Blood. 2015;126:1357–1366. doi: 10.1182/blood-2015-01-624809.
    1. Kojima, J. et al. Apoptosis inhibitor of macrophage (AIM) expression in alveolar macrophages in COPD. Respir Res14, doi:10.1186/1465-9921-14-30 (2013).
    1. Fabriek BO, Dijkstra CD, van den Berg TK. The macrophage scavenger receptor CD163. Immunobiology. 2005;210:153–160. doi: 10.1016/j.imbio.2005.05.010.
    1. Kaku, Y. et al. Overexpression of CD163, CD204 and CD206 on alveolar macrophages in the lungs of patients with severe chronic obstructive pulmonary disease. PloS One9, doi:10.1371/journal.pone.0087400 (2014).
    1. Viksman MY, Liu MC, Bickel CA, Schleimer RP, Bochner BS. Phenotypic analysis of alveolar macrophages and monocytes in allergic airway inflammation. I. Evidence for activation of alveolar macrophages, but not peripheral blood monocytes, in subjects with allergic rhinitis and asthma. Am J Respir Crit Care Med. 1997;155:858–863. doi: 10.1164/ajrccm.155.3.9117017.
    1. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–461. doi: 10.2741/2692.
    1. Kent L, et al. Cigarette smoke extract induced cytokine and chemokine gene expression changes in COPD macrophages. Cytokine. 2008;42:205–216. doi: 10.1016/j.cyto.2008.02.001.
    1. Samet JM, et al. Lung cancer in never smokers: clinical epidemiology and environmental risk factors. Clinical cancer research: an official journal of the American Association for Cancer Research. 2009;15:5626–5645. doi: 10.1158/1078-0432.CCR-09-0376.
    1. Wilson R. Bacteria, antibiotics and COPD. Eur Respir J. 2001;17:995–1007. doi: 10.1183/09031936.01.17509950.
    1. Berenson CS, Kruzel RL, Eberhardt E, Sethi S. Phagocytic dysfunction of human alveolar macrophages and severity of chronic obstructive pulmonary disease. J Infect Dis. 2013;208:2036–2045. doi: 10.1093/infdis/jit400.
    1. Pons AR, et al. Phenotypic characterisation of alveolar macrophages and peripheral blood monocytes in COPD. Eur Respir J. 2005;25:647–652. doi: 10.1183/09031936.05.00062304.
    1. Droemann, D. et al. Toll-like receptor 2 expression is decreased on alveolar macrophages in cigarette smokers and COPD patients. Respir Res6, doi:10.1186/1465-9921-6-68 (2005).
    1. Domagala-Kulawik J, Maskey-Warzechowska M, Kraszewska I, Chazan R. The cellular composition and macrophage phenotype in induced sputum in smokers and ex-smokers with COPD. Chest. 2003;123:1054–1059. doi: 10.1378/chest.123.4.1054.
    1. Kunz, L.I. et al. Smoking status and anti-inflammatory macrophages in bronchoalveolar lavage and induced sputum in COPD. Respir Res12, doi:10.1186/1465-9921-12-34 (2011).
    1. Demedts, I. K., Demoor, T., Bracke, K. R., Joos, G. F. & Brusselle, G. G. Role of apoptosis in the pathogenesis of COPD and pulmonary emphysema. Respir Res7, doi:10.1186/1465-9921-7-53 (2006).
    1. Singh D, et al. Induced sputum genes associated with spirometric and radiological disease severity in COPD ex-smokers. Thorax. 2011;66:489–495. doi: 10.1136/thx.2010.153767.
    1. Bowler RP, et al. Plasma sphingolipids associated with chronic obstructive pulmonary disease phenotypes. Am J Respir Crit Care Med. 2015;191:275–284. doi: 10.1164/rccm.201410-1771OC.
    1. Boraschi D, Tagliabue A. The interleukin-1 receptor family. Seminars in immunology. 2013;25:394–407. doi: 10.1016/j.smim.2013.10.023.
    1. Riabov, V. et al. Role of tumor associated macrophages in tumor angiogenesis and lymphangiogenesis. Frontiers in physiology5, doi:10.3389/fphys.2014.00075 (2014).
    1. Ancelin M, et al. Vascular endothelial growth factor VEGF189 induces human neutrophil chemotaxis in extravascular tissue via an autocrine amplification mechanism. Laboratory investigation; a journal of technical methods and pathology. 2004;84:502–512. doi: 10.1038/labinvest.3700053.
    1. Vainer B, Nielsen OH. Chemotactic properties of ICAM-1 and PECAM-1 on neutrophil granulocytes in ulcerative colitis: effects of prednisolone and mesalazine. Aliment Pharmacol Ther. 2000;14:1023–1031. doi: 10.1046/j.1365-2036.2000.00797.x.
    1. Higham, A. et al. The effects of corticosteroids on COPD lung macrophages: a pooled analysis. Respir Res16, doi:10.1186/s12931-015-0260-0 (2015).
    1. Heasman SJ, et al. Interferon gamma suppresses glucocorticoid augmentation of macrophage clearance of apoptotic cells. Eur J Immunol. 2004;34:1752–1761. doi: 10.1002/eji.200324698.

Source: PubMed

3
Prenumerera