Human CD56+ cytotoxic lung lymphocytes kill autologous lung cells in chronic obstructive pulmonary disease

Christine M Freeman, Valerie R Stolberg, Sean Crudgington, Fernando J Martinez, MeiLan K Han, Stephen W Chensue, Douglas A Arenberg, Catherine A Meldrum, Lisa McCloskey, Jeffrey L Curtis, Christine M Freeman, Valerie R Stolberg, Sean Crudgington, Fernando J Martinez, MeiLan K Han, Stephen W Chensue, Douglas A Arenberg, Catherine A Meldrum, Lisa McCloskey, Jeffrey L Curtis

Abstract

CD56+ natural killer (NK) and CD56+ T cells, from sputum or bronchoalveolar lavage of subjects with chronic obstructive pulmonary disease (COPD) are more cytotoxic to highly susceptible NK targets than those from control subjects. Whether the same is true in lung parenchyma, and if NK activity actually contributes to emphysema progression are unknown. To address these questions, we performed two types of experiments on lung tissue from clinically-indicated resections (n = 60). First, we used flow cytometry on fresh single-cell suspension to measure expression of cell-surface molecules (CD56, CD16, CD8, NKG2D and NKp44) on lung lymphocytes and of the 6D4 epitope common to MICA and MICB on lung epithelial (CD326+) cells. Second, we sequentially isolated CD56+, CD8+ and CD4+ lung lymphocytes, co-cultured each with autologous lung target cells, then determined apoptosis of individual target cells using Annexin-V and 7-AAD staining. Lung NK cells (CD56+ CD3-) and CD56+ T cells (CD56+ CD3+) were present in a range of frequencies that did not differ significantly between smokers without COPD and subjects with COPD. Lung NK cells had a predominantly "cytotoxic" CD56+ CD16+ phenotype; their co-expression of CD8 was common, but the percentage expressing CD8 fell as FEV1 % predicted decreased. Greater expression by autologous lung epithelial cells of the NKG2D ligands, MICA/MICB, but not expression by lung CD56+ cells of the activating receptor NKG2D, correlated inversely with FEV1 % predicted. Lung CD56+ lymphocytes, but not CD4+ or CD8+ conventional lung T cells, rapidly killed autologous lung cells without additional stimulation. Such natural cytotoxicity was increased in subjects with severe COPD and was unexplained in multiple regression analysis by age or cancer as indication for surgery. These data show that as spirometry worsens in COPD, CD56+ lung lymphocytes exhibit spontaneous cytotoxicity of autologous structural lung cells, supporting their potential role in emphysema progression.

Trial registration: ClinicalTrials.gov NCT00281229.

Conflict of interest statement

Competing Interests: The authors have read the journal’s policy and the authors of this manuscript have the following competing interests: MKH participated in advisory boards for Boehringer Ingelheim, Pfizer, GlaxoSmithKline, Genentech, Novartis, Forest and Medimmune; participated on speaker’s bureaus for Boehringer Ingelheim, Pfizer, GlaxoSmithKline, Grifols Therapeutics, Forest and the National Association for Continuing Education, and WebMD; has consulted for Novartis and United Biosource Corporation; and has received royalties from UpToDate and ePocrates. She has served as an investigator for research sponsored by GlaxoSmithKline. FJM has been a member of advisory boards and/or consultant for Able Associates, Actelion, Almirall, Bayer, GSK, Ikaria, Janssen, MedImmune, Merck, Pearl, Pfizer and Vertex, American Institute for Research, AstraZeneca, Carden Jennings, Cardiomems, Grey Healthcare, HealthCare Research and Consulting, Merion, Nycomed/Takeda, and Sudler and Hennessey. He has been a member of steering committees for studies sponsored by Actelion, Centocor, Forest, GlaxoSmithKline, Gilead, Mpex, Nycomed/Takeda. He has participated in Food and Drug Administration mock panels for Boehringer Ingelheim, Forest and GSK. He has served on speaker’s bureaus or in continuing medical education activities sponsored by American College of Chest Physicians, American Lung Association, Astra Zeneca, Bayer, William Beaumont Hospital, Boehringer Ingelheim, Center for Health Care Education, CME Incite, Forest, France Foundation, GlaxoSmithKline, Lovelace, MedEd, MedScape/WebMD, National Association for Continuing Education, Network for Continuing Medical Education, Nycomed/Takeda, Projects in Knowledge, St Luke’s Hospital, the University of Illinois Chicago, University of Texas Southwestern, University of Virginia, UpToDate. He has served on DSMBs for Biogen and Novartis. He has received royalties from Castle Connolly and Informa. This does not alter the authors’ adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Identification and characterization of human…
Figure 1. Identification and characterization of human lung NK cells and CD56+ T cells.
Lung tissue was dispersed, stained with monoclonal antibodies against CD45, CD3, CD56, and CD16 and analyzed by flow cytometry to select a viable population comprised predominately of lung lymphocytes (CD45+, low side-scatter cells). (A, C, E) Representative staining: isotype controls on left, specific staining on right; (B, D, F) Frequency of various lung lymphocyte populations in individual subjects as a percentage of the total viable lung lymphocyte population; note difference in scale of panel B. (A) Ungated staining for CD3 and CD56 identifies four distinct populations: NK cells (CD56+ CD3−); CD56+ T cells (CD56+ CD3+); conventional T cells (CD56− CD3+); and double-negative cells (predominately B cells). (B) NK cells (blue bars) versus CD56+ T cells (orange bars). (C, D) After gating on CD3− cells, staining for CD56 and CD16 identifies two lung NK populations: CD56+ CD16+ (light blue circle & columns) and CD56+ CD16− (dark blue circle & columns). (E, F) After gating on CD3+ cells, staining for CD56 and CD16 identifies two lung CD56+ T cell populations: CD56+ CD16+ (dark orange circle & columns) and CD56+ CD16− (light orange circle & columns). By Kruskal-Wallis one-way ANOVA, there are no significant differences between subject groups for any of these three lung cell populations (B, D, F).
Figure 2. The frequency of lung NK…
Figure 2. The frequency of lung NK cells, but not lung CD56+ T cells, co-expressing CD8 correlates with FEV1 % predicted.
(A–C) NK cells; (D–F) CD56+ T cells. (A) Representative staining for CD8 on CD56+ CD3− NK cells from a smoker without COPD (left panel) and a COPD subject (right panel); blue line, CD8+ staining; grey line, isotype control. (B) Subjects were categorized by pulmonary function (x-axis) versus the percentage of NK cells that co-express CD8 (y-axis); x, smokers without COPD (n = 5); □, mild COPD (n = 9); Δ, severe COPD (n = 9); Kruskal-Wallis one-way ANOVA testing was used to determine significance. (C) The percentage of NK cells that co-express CD8 (y-axis) versus FEV1 % predicted (x-axis). Spearman correlation was used to determine the p-value. (D) Representative staining for CD8 and CD4 on CD56+ CD3+ T cells from a smoker without COPD (left panel) and a COPD subject (right panel). The numbers in the quadrants are the % of each subset among all lung CD56+ T cells. (E) CD4 single-positive (brown bars), CD8 single-positive (orange bars), and CD8/CD4 double-negative (grey bars) are shown as the percentage of lung CD56+ T cells for individual subjects. No difference was seen between groups. (F) The percentage of lung CD56+ T cells (y-axis) that express CD4 (brown symbols) or CD8 (orange symbols) versus FEV1 % predicted (x-axis); x, smokers without COPD (n = 5); □, mild COPD (n = 9); Δ, severe COPD (n = 9).
Figure 3. The percentage of epithelial cells…
Figure 3. The percentage of epithelial cells expressing MICA/MICB is increased with COPD severity.
Human lung tissue was dispersed and stained with monoclonal antibodies against CD45, CD56, NKG2D, CD326, and MICA/MICB. (A) Representative staining showing the expression of NKG2D on CD45+ CD56+ cells from a smoker without COPD (left panel) and a subject with COPD (right panel). Blue line, NKG2D+ staining; grey line, isotype control. (B) The percentage of CD56+ cells that express NKG2D (y-axis) versus FEV1 % predicted (x-axis). x, smokers without COPD (n = 10); □, mild COPD (n = 5); Δ, severe COPD (n = 10). N.S., not significant. (C) Representative staining showing the expression of MICA/MICB on CD45−, CD326 (EpCAM)+ epithelial cells from a smoker without COPD (left panel) and a subject with COPD (right panel). Blue line, MICA/MICB+ staining; grey line, isotype control. (D) The percentage of CD326+ epithelial cells that express MICA/MICB (y-axis) versus FEV1 % predicted (x-axis). x, smokers without COPD (n = 10); □, mild COPD (n = 5); Δ, severe COPD (n = 10). Spearman correlation was used to determine the p value.
Figure 4. Human lung CD56+ cells spontaneously…
Figure 4. Human lung CD56+ cells spontaneously kill autologous lung CD45− cells in vitro.
CD56+ cells were isolated from dispersed human lung tissue using magnetic beads. CD8+ cells were isolated from the CD56 depleted fraction and CD4+ cells were isolated from the CD56− and CD8− depleted fraction. The remaining cells were used as autologous target cells. Target cells were cultured either alone or with CD56+ cells, CD8+ cells, or CD4+ cells at a ratio of 1 target to 10 effectors. After 4 hours, all cells were collected and stained with CD45, Annexin-V, and 7-AAD for flow cytometry. Target cells were identified as CD45− with a high side scatter. (A) Representative staining of Annexin-V on target cells that were cultured with CD56+ cells (left panel), CD8+ cells (middle panel), and CD4+ cells (right panel). (B) % Cytotoxicity (y-axis) for target cells cultured with CD56+ cells (blue circles), CD8+ cells (orange circles), or CD4+ cells (green circles); n = 28. Lines represent the mean ± SEM. The Kruskal-Wallis one-way ANOVA with Dunn’s multiple comparison test was used to determine significant differences between groups.
Figure 5. Increased cytotoxicity by lung CD56+…
Figure 5. Increased cytotoxicity by lung CD56+ cells correlates with decreased pulmonary function.
Human lung CD56+ cells were co-cultured with autologous lung target cells and % cytotoxicity was determined as described in the Methods. (A) Subjects were categorized by pulmonary function (x-axis) versus % cytotoxicity (y-axis). The Kruskal-Wallis one-way ANOVA with Dunn’s multiple comparison test was used to determine significant differences between groups. (B) FEV1 % predicted (x-axis) versus % cytotoxicity (y-axis). Spearman non-parametric correlation was used to determine the p-value. (C, D) The same subjects were separated into two groups based on their CD56+ cell function: non-cytotoxic (<8.5% cytotoxicity) or cytotoxic (≥8.5% cytotoxicity) and then FEV1 % predicted (C) and DLCO % predicted (D) were analyzed. The parametric unpaired Student t-test was used to determine significant differences between the two groups. In all figures, x, smokers without COPD (n = 6); □, mild COPD (n = 12); Δ, severe COPD (n = 10). Lines represent the mean ± SEM.

References

    1. Arialdi M, Minino JX, Kochanek KD (2010) Deaths: Preliminary Data for 2008. National Vital Statistics Reports 59: 1–72.
    1. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, et al. (2004) The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 350: 2645–2653.
    1. Curtis JL, Freeman CM, Hogg JC (2007) The immunopathogenesis of chronic obstructive pulmonary disease: insights from recent research. Proc Am Thorac Soc 4: 512–521.
    1. Martinez FJ, Han MK, Andrei AC, Wise R, Murray S, et al. (2008) Longitudinal change in the BODE index predicts mortality in severe emphysema. Am J Respir Crit Care Med 178: 491–499.
    1. Fishman A, Martinez F, Naunheim K, Piantadosi S, Wise R, et al. (2003) A randomized trial comparing lung-volume-reduction surgery with medical therapy for severe emphysema. N Engl J Med 348: 2059–2073.
    1. Naunheim KS, Wood DE, Mohsenifar Z, Sternberg AL, Criner GJ, et al. (2006) Long-term follow-up of patients receiving lung-volume-reduction surgery versus medical therapy for severe emphysema by the National Emphysema Treatment Trial Research Group. Ann Thorac Surg 82: 431–443.
    1. Stetson DB, Mohrs M, Reinhardt RL, Baron JL, Wang ZE, et al. (2003) Constitutive cytokine mRNAs mark natural killer (NK) and NK T cells poised for rapid effector function. J Exp Med 198: 1069–1076.
    1. Hodge G, Mukaro V, Holmes M, Reynolds PN, Hodge S (2013) Enhanced cytotoxic function of natural killer and natural killer T-like cells associated with decreased CD94 (Kp43) in the chronic obstructive pulmonary disease airway. Respirology 18: 369–376.
    1. Urbanowicz RA, Lamb JR, Todd I, Corne JM, Fairclough LC (2010) Enhanced effector function of cytotoxic cells in the induced sputum of COPD patients. Respir Res 11: 76.
    1. Stolberg VR, Martin B, Mancuso P, Olszewski MA, Freeman CM, et al. (2014) Role of CC Chemokine Receptor 4 in Natural Killer cell activation during acute cigarette smoke exposure. Am J Pathol 184: 454–463.
    1. Motz GT, Eppert BL, Wortham BW, Amos-Kroohs RM, Flury JL, et al. (2010) Chronic cigarette smoke exposure primes NK cell activation in a mouse model of chronic obstructive pulmonary disease. J Immunol 184: 4460–4469.
    1. Wortham BW, Eppert BL, Motz GT, Flury JL, Orozco-Levi M, et al. (2012) NKG2D mediates NK cell hyperresponsiveness and influenza-induced pathologies in a mouse model of chronic obstructive pulmonary disease. J Immunol 188: 4468–4475.
    1. Demedts IK, Demoor T, Bracke KR, Joos GF, Brusselle GG (2006) Role of apoptosis in the pathogenesis of COPD and pulmonary emphysema. Respir Res 7: 53.
    1. Imai K, Mercer BA, Schulman LL, Sonett JR, D’Armiento JM (2005) Correlation of lung surface area to apoptosis and proliferation in human emphysema. Eur Respir J 25: 250–258.
    1. Segura-Valdez L, Pardo A, Gaxiola M, Uhal BD, Becerril C, et al. (2000) Upregulation of gelatinases A and B, collagenases 1 and 2, and increased parenchymal cell death in COPD. Chest 117: 684–694.
    1. Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S (2013) Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol 31: 227–258.
    1. Campbell KS, Hasegawa J (2013) Natural killer cell biology: an update and future directions. J Allergy Clin Immunol 132: 536–544.
    1. Raulet DH (2003) Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 3: 781–790.
    1. Borchers MT, Harris NL, Wesselkamper SC, Vitucci M, Cosman D (2006) NKG2D ligands are expressed on stressed human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 291: L222–231.
    1. Borchers MT, Wesselkamper SC, Curull V, Ramirez-Sarmiento A, Sanchez-Font A, et al. (2009) Sustained CTL activation by murine pulmonary epithelial cells promotes the development of COPD-like disease. J Clin Invest 119: 636–649.
    1. Wortham BW, Eppert BL, Flury JL, Morgado Garcia S, Borchers MT (2013) TLR and NKG2D signaling pathways mediate CS-induced pulmonary pathologies. PLoS One 8: e78735.
    1. Shi FD, Ljunggren HG, La Cava A, Van Kaer L (2011) Organ-specific features of natural killer cells. Nat Rev Immunol 11: 658–671.
    1. Michel T, Poli A, Domingues O, Mauffray M, Theresine M, et al. (2012) Mouse lung and spleen natural killer cells have phenotypic and functional differences, in part influenced by macrophages. PLoS One 7: e51230.
    1. Urbanowicz RA, Lamb JR, Todd I, Corne JM, Fairclough LC (2009) Altered effector function of peripheral cytotoxic cells in COPD. Respir Res 10: 53.
    1. Chi SY, Ban HJ, Kwon YS, Oh IJ, Kim KS, et al. (2012) Invariant natural killer T cells in chronic obstructive pulmonary disease. Respirology 17: 486–492.
    1. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, et al. (2007) Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med 176: 532–555.
    1. Freeman CM, Curtis JL, Chensue SW (2007) CC chemokine receptor 5 and CXC chemokine receptor 6 expression by lung CD8+ cells correlates with chronic obstructive pulmonary disease severity. Am J Pathol 171: 767–776.
    1. Freeman CM, Han MK, Martinez FJ, Murray S, Liu LX, et al. (2010) Cytotoxic potential of lung CD8(+) T cells increases with chronic obstructive pulmonary disease severity and with in vitro stimulation by IL-18 or IL-15. J Immunol 184: 6504–6513.
    1. Freeman CM, Martinez FJ, Han MK, Washko GR Jr, McCubbrey AL, et al. (2013) Lung CD8+ T cells in COPD have increased expression of bacterial TLRs. Respir Res 14: 13.
    1. McCubbrey AL, Sonstein J, Ames TM, Freeman CM, Curtis JL (2012) Glucocorticoids relieve collectin-driven suppression of apoptotic cell uptake in murine alveolar macrophages through downregulation of SIRPalpha. J Immunol 189: 112–119.
    1. Culley FJ (2009) Natural killer cells in infection and inflammation of the lung. Immunology 128: 151–163.
    1. Adar T, Ya’acov A., Lalazar G. (2008) Natural Killer T-Cells: Roles, Interactions, and Interventions; Fournier NV, editor: Nova Publishers.
    1. Berzins SP, Smyth MJ, Baxter AG (2011) Presumed guilty: natural killer T cell defects and human disease. Nat Rev Immunol 11: 131–142.
    1. Morice WG (2007) The immunophenotypic attributes of NK cells and NK-cell lineage lymphoproliferative disorders. Am J Clin Pathol 127: 881–886.
    1. Beziat V, Duffy D, Quoc SN, Le Garff-Tavernier M, Decocq J, et al. (2011) CD56bright CD16+ NK cells: a functional intermediate stage of NK cell differentiation. J Immunol 186: 6753–6761.
    1. Forsslund H, Mikko M, Karimi R, Grunewald J, Wheelock AM, et al. (2014) Distribution of T-Cell Subsets in BAL fluid of patients with mild to moderate COPD depends on current smoking status and not airway obstruction. Chest 145: 711–722.
    1. Hodge S, Hodge G, Holmes M, Reynolds PN (2005) Increased airway epithelial and T-cell apoptosis in COPD remains despite smoking cessation. Eur Respir J 25: 447–454.
    1. Solana R, Mariani E (2000) NK and NK/T cells in human senescence. Vaccine 18: 1613–1620.
    1. Roos-Engstrand E, Pourazar J, Behndig AF, Blomberg A, Bucht A (2010) Cytotoxic T cells expressing the co-stimulatory receptor NKG2 D are increased in cigarette smoking and COPD. Respir Res 11: 128.
    1. Vitale M, Bottino C, Sivori S, Sanseverino L, Castriconi R, et al. (1998) NKp44, a novel triggering surface molecule specifically expressed by activated natural killer cells, is involved in non-major histocompatibility complex-restricted tumor cell lysis. J Exp Med 187: 2065–2072.
    1. Esin S, Batoni G, Counoupas C, Stringaro A, Brancatisano FL, et al. (2008) Direct binding of human NK cell natural cytotoxicity receptor NKp44 to the surfaces of mycobacteria and other bacteria. Infect Immun 76: 1719–1727.
    1. Carbone E, Terrazzano G, Ruggiero G, Zanzi D, Ottaiano A, et al. (1999) Recognition of autologous dendritic cells by human NK cells. Eur J Immunol 29: 4022–4029.
    1. Spaggiari GM, Carosio R, Pende D, Marcenaro S, Rivera P, et al. (2001) NK cell-mediated lysis of autologous antigen-presenting cells is triggered by the engagement of the phosphatidylinositol 3-kinase upon ligation of the natural cytotoxicity receptors NKp30 and NKp46. Eur J Immunol 31: 1656–1665.
    1. Poggi A, Prevosto C, Massaro AM, Negrini S, Urbani S, et al. (2005) Interaction between human NK cells and bone marrow stromal cells induces NK cell triggering: role of NKp30 and NKG2D receptors. J Immunol 175: 6352–6360.
    1. Gao B, Bertola A (2011) Natural killer cells take two tolls to destruct bile ducts. Hepatology 53: 1076–1079.
    1. Shimoda S, Harada K, Niiro H, Shirabe K, Taketomi A, et al. (2011) Interaction between Toll-like receptors and natural killer cells in the destruction of bile ducts in primary biliary cirrhosis. Hepatology 53: 1270–1281.
    1. Saetta M, Di Stefano A, Turato G, Facchini FM, Corbino L, et al. (1998) CD8+ T-lymphocytes in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 157: 822–826.
    1. Majo J, Ghezzo H, Cosio MG (2001) Lymphocyte population and apoptosis in the lungs of smokers and their relation to emphysema. Eur Respir J 17: 946–953.
    1. O’Shaughnessy TC, Ansari TW, Barnes NC, Jeffery PK (1997) Inflammation in bronchial biopsies of subjects with chronic bronchitis: inverse relationship of CD8+ T lymphocytes with FEV1. Am J Respir Crit Care Med 155: 852–857.
    1. Maeno T, Houghton AM, Quintero PA, Grumelli S, Owen CA, et al. (2007) CD8+ T Cells are required for inflammation and destruction in cigarette smoke-induced emphysema in mice. J Immunol 178: 8090–8096.
    1. Borchers MT, Wesselkamper SC, Harris NL, Deshmukh H, Beckman E, et al. (2007) CD8+ T cells contribute to macrophage accumulation and airspace enlargement following repeated irritant exposure. Exp Mol Pathol 83: 301–310.
    1. Sullivan AK, Simonian PL, Falta MT, Mitchell JD, Cosgrove GP, et al. (2005) Oligoclonal CD4+ T cells in the lungs of patients with severe emphysema. Am J Respir Crit Care Med 172: 590–596.
    1. Motz GT, Eppert BL, Sun G, Wesselkamper SC, Linke MJ, et al. (2008) Persistence of lung CD8 T cell oligoclonal expansions upon smoking cessation in a mouse model of cigarette smoke-induced emphysema. J Immunol 181: 8036–8043.
    1. Motz GT, Eppert BL, Wesselkamper SC, Flury JL, Borchers MT (2010) Chronic cigarette smoke exposure generates pathogenic T cells capable of driving COPD-like disease in Rag2−/− mice. Am J Respir Crit Care Med 181: 1223–1233.
    1. Wang Z, Zheng T, Zhu Z, Homer RJ, Riese RJ, et al. (2000) Interferon gamma induction of pulmonary emphysema in the adult murine lung. J Exp Med 192: 1587–1600.
    1. Grumelli S, Corry DB, Song LZ, Song L, Green L, et al. (2004) An immune basis for lung parenchymal destruction in chronic obstructive pulmonary disease and emphysema. PLOS Med 1: e8.
    1. Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD (1997) Requirement for macrophage elastase for cigarette smoke-induced emphysema in mice. Science 277: 2002–2004.
    1. Piet B, de Bree GJ, Smids-Dierdorp BS, van der Loos CM, Remmerswaal EB, et al. (2011) CD8(+) T cells with an intraepithelial phenotype upregulate cytotoxic function upon influenza infection in human lung. J Clin Invest 121: 2254–2263.

Source: PubMed

Подписаться