Functional characterisation of bone marrow-derived mesenchymal stromal cells from COPD patients

Winifred Broekman, Helene Roelofs, Maria C Zarcone, Christian Taube, Jan Stolk, Pieter S Hiemstra, Winifred Broekman, Helene Roelofs, Maria C Zarcone, Christian Taube, Jan Stolk, Pieter S Hiemstra

Abstract

Autologous bone marrow-derived mesenchymal stromal cells (BM-MSCs) are evaluated for clinical use in chronic obstructive pulmonary disease (COPD) patients, but it is unclear whether COPD affects BM-MSCs. To investigate this, BM-MSCs from nine COPD patients and nine non-COPD age-matched controls were compared with regard to immunophenotype, growth and differentiation potential, and migration capacity. Other functional assays included the response to pro-inflammatory stimuli and inducers of the nuclear factor (erythroid derived 2)-like 2 antioxidant response element (Nrf2-ARE) pathway, and effects on NCI-H292 airway epithelial cells. No significant differences were observed in terms of morphology, proliferation and migration, except for increased adipocyte differentiation potential in the COPD group. Both groups were comparable regarding mRNA expression of growth factors and inflammatory mediators, and in their potential to induce mRNA expression of epidermal growth factor receptor ligands in NCI-H292 airway epithelial cells. MSCs from COPD patients secreted more interleukin-6 in response to pro-inflammatory stimuli. Activation of the Nrf2-ARE pathway resulted in a comparable induction of mRNA expression of four target genes, but the expression of the NAD(P)H:quinone oxidoreductase 1 gene NQO1 was lower in MSCs from COPD patients. The observation that MSCs from COPD patients are phenotypically and functionally comparable to those from non-COPD controls implies that autologous MSCs can be considered for use in the setting of clinical trials as a treatment for COPD.

Conflict of interest statement

can be found alongside this article at openres.ersjournals.com

Figures

FIGURE 1
FIGURE 1
Mesenchymal stromal cell (MSC) phenotype from chronic obstructive pulmonary disease (COPD) and non-COPD patients. Bone marrow-derived MSCs (BM-MSCs) from COPD and non-COPD patients cultured in vitro were characterised by fluorescence-activated cell sorter (FACS) analysis and by their potential to differentiate into mesenchymal lineages. a) Morphology of MSCs from COPD and non-COPD donors: example of MSC culture. Scale bar s=100 μm. b) FACS data. Per antibody, data are shown as mean±sem. n=9 per group. c) Differentiation into adipocytes and osteoblasts, and mineralisation potential. Staining intensity of Oil red O, alkaline phosphatase and Alizarin red (calcium) was quantified on a microplate reader, and per donor optical density (OD) values were calculated as fold change compared with control (co-cultured undifferentiated MSCs). Plots represent median, interquartile range, and minimum and maximum values. *: p<0.05, n=9 per group (except osteoblast/mineralisation: n=6–8 per group).
FIGURE 2
FIGURE 2
Mesenchymal stromal cell (MSC) proliferation and migration capacity. In vitro proliferation of MSCs from chronic obstructive pulmonary disease (COPD) and non-COPD patients was followed over time. a) Time needed for cultures to become near-confluent and the number of MSCs that were obtained per passage were assessed. b) Population doubling time (PDT) was calculated by dividing the natural logarithm of 2 by the exponent of growth. Plots represent median, interquartile range, and minimum and maximum values. n=9 per group. c) Migration as assessed using electric cell-substrate impedance sensing (ECIS). MSCs were cultured in ECIS arrays in the presence of an electrical fence (EF), which prevented cell adherence across the electrode area. Resistance (at 500 Hz) and capacitance (at 40 kHz) were measured continuously and followed up to 15 h after removal of the electrical fence (t=0 h). Restoration of the resistance and capacitance to control values (corresponding to full coverage of the electrode) was used as a measure for migration capacity of MSCs. Normalised data were obtained by correcting for the resistance/capacitance values obtained in control wells without an electric fence, at the time the electric fence was removed (t=0 h). Data are presented as mean±sem. n=5–8 per group.
FIGURE 3
FIGURE 3
Induction of immune mediators and growth factors upon stimulation with pro-inflammatory cytokines. Mesenchymal stromal cells (MSCs) were stimulated with tumour necrosis factor (TNF)-α and interleukin (IL)-1β (20 ng·mL−1 each) or plain culture medium as a negative control (NC) during 6 h. a) mRNA expression of immune mediators and growth factors, normalised for housekeeping genes, shown for chronic obstructive pulmonary disease (COPD) (circles) and non-COPD donors (squares). AREG: amphiregulin; HBEGF: heparin-binding epidermal growth factor-like growth factor; FGF2: fibroblast growth factor 2; ADM1: adrenomedullin; CCL20: chemokine (C-C motif) ligand 20; IL6: IL-6; CXCL8: chemokine (C-X-C motif) ligand 8; TSG6: TNF-stimulated gene 6. Individual data are shown in graphs, horizontal bars represent mean. *: p<0.05, n=9 per group. b) IL-6 and IL-8 protein secretion by MSCs from COPD and non-COPD donors, assessed 24 h after addition of TNF-α and IL-1β. Individual data points are shown; horizontal bars represent mean. *: p<0.05, n=9 per group.
FIGURE 4
FIGURE 4
Induction of the nuclear factor (erythroid derived 2)-like 2 antioxidant response element (Nrf2-ARE) target genes in mesenchymal stromal cells (MSCs) from chronic obstructive pulmonary disease (COPD) and non-COPD donors. Gene expression of glutathione peroxidase 2 (GPX2), haem oxygenase 1 (HMOX1), NAD(P)H:quinone oxidoreductase 1 (NQO1) and smoke and cancer-associated long noncoding RNA-1 (SCAL1) was assessed in MSCs from COPD and non-COPD donors 6 h after addition of cigarette smoke extract (CSE) or sulforaphane (SFN), which are both inducers of the Nrf2-ARE pathway, or plain culture medium as a negative control (NC). a) Normalised mRNA expression in response to increasing concentrations of CSE. Data are presented as mean±sem. *: p<0.05, n=9 per group (except CSE 1.0 AU·mL−1: n=7 in COPD group). b) Normalised mRNA expression in response to 25 μM SFN, compared with housekeeping genes. Individual data points are shown (circles: COPD; squares: non-COPD); horizontal bars represent mean. *: p<0.05, n=9 per group.
FIGURE 5
FIGURE 5
Paracrine effects of mesenchymal stromal cells (MSCs) on H292 airway epithelial cells. NCI-H292 airway epithelial cells were incubated with conditioned medium obtained from MSCs (MSC-CM). mRNA expression of epidermal growth factor receptor ligands after 9 h incubation with conditioned medium from MSCs cultured in serum-free low-glucose Dulbecco's modified Eagle medium (LG-DMEM) (Ctrl, from MSC-CMCTRL) or in serum-free LG-DMEM supplemented with TNF-α and IL-1β (Stim, from MSC-CMSTIM). AREG: amphiregulin; HBEGF: heparin-binding epidermal growth factor-like growth factor; TGFA: transforming growth factor-α; CCDN1: cyclin D1. Normalised values compared with housekeeping genes are shown. Individual data points are shown (circles: chronic obstructive pulmonary disease (COPD) donor-derived MSC-CM; squares: non-COPD donor-derived MSC-CM); horizontal bars represent mean. *: p<0.05, n=9 per group. For all genes investigated, mRNA expression was lower in cells cultured in plain culture medium (LG-DMEM without additional stimulation) compared with mRNA expression in cells cultured in MSC-CM (data not shown).

References

    1. Dominici M, Le Blanc K, Mueller I, et al. . Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006; 8: 315–317.
    1. Keating A. Mesenchymal stromal cells: new directions. Cell Stem Cell 2012; 10: 709–716.
    1. Nauta AJ, Fibbe WE. Immunomodulatory properties of mesenchymal stromal cells. Blood 2007; 110: 3499–3506.
    1. Brusselle GG, Joos GF, Bracke KR. New insights into the immunology of chronic obstructive pulmonary disease. Lancet 2011; 378: 1015–1026.
    1. Weiss DJ, Casaburi R, Flannery R, et al. . A placebo-controlled, randomized trial of mesenchymal stem cells in COPD. Chest 2013; 143: 1590–1598.
    1. Stolk J, Broekman W, Mauad T, et al. . A phase I study for intravenous autologous mesenchymal stromal cell administration to patients with severe emphysema. QJM 2016; 109: 331–336.
    1. Barnes PJ, Celli BR. Systemic manifestations and comorbidities of COPD. Eur Respir J 2009; 33: 1165–1185.
    1. Jahn A., Rio C, Gigirey O, et al. . Bone marrow-derived MSCs from patients with COPD have abnormal functional capacity. Eur Respir J 2013; 42: Suppl. 57, 22s.
    1. Rankin SM. Impact of bone marrow on respiratory disease. Curr Opin Pharmacol 2008; 8: 236–241.
    1. Duijvestein M, Vos AC, Roelofs H, et al. . Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn's disease: results of a phase I study. Gut 2010; 59: 1662–1669.
    1. Ma Q. Role of Nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol 2013; 53: 401–426.
    1. Sekine T, Hirata T, Mine T, et al. . Activation of transcription factors in human bronchial epithelial cells exposed to aqueous extracts of mainstream cigarette smoke in vitro. Toxicol Mech Methods 2016; 26: 22–31.
    1. Zhang Y, Talalay P, Cho CG, et al. . A major inducer of anticarcinogenic protective enzymes from broccoli: isolation and elucidation of structure. Proc Natl Acad Sci U S A 1992; 89: 2399–2403.
    1. Vandesompele J, De Preter K, Pattyn F, et al. . Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 2002; 3: RESEARCH0034.
    1. Broekman W, Amatngalim GD, Mooij-Eijk Y, et al. . TNF-α and IL-1β-activated human mesenchymal stromal cells increase airway epithelial wound healing in vitro via activation of the epidermal growth factor receptor. Respir Res 2016; 17: 1–12.
    1. Ma S, Xie N, Li W, et al. . Immunobiology of mesenchymal stem cells. Cell Death Differ 2014; 21: 216–225.
    1. MacNee W. Pulmonary and systemic oxidant/antioxidant imbalance in chronic obstructive pulmonary disease. Proc Am Thorac Soc 2005; 2: 50–60.
    1. Reinders ME, Roemeling-van Rhijn M, Khairoun M, et al. . Bone marrow-derived mesenchymal stromal cells from patients with end-stage renal disease are suitable for autologous therapy. Cytotherapy 2013; 15: 663–672.
    1. Larghero J, Farge D, Braccini A, et al. . Phenotypical and functional characteristics of in vitro expanded bone marrow mesenchymal stem cells from patients with systemic sclerosis. Ann Rheum Dis 2008; 67: 443–449.
    1. Bocelli-Tyndall C, Bracci L, Spagnoli G, et al. . Bone marrow mesenchymal stromal cells (BM-MSCs) from healthy donors and auto-immune disease patients reduce the proliferation of autologous- and allogeneic-stimulated lymphocytes in vitro. Rheumatology 2007; 46: 403–408.
    1. Bernardo ME, Avanzini MA, Ciccocioppo R, et al. . Phenotypical/functional characterization of in vitro-expanded mesenchymal stromal cells from patients with Crohn's disease. Cytotherapy 2009; 11: 825–836.
    1. Cho GW, Noh MY, Kim HY, et al. . Bone marrow-derived stromal cells from amyotrophic lateral sclerosis patients have diminished stem cell capacity. Stem Cells Dev 2009; 19: 1035–1042.
    1. Mazzanti B, Aldinucci A, Biagioli T, et al. . Differences in mesenchymal stem cell cytokine profiles between MS patients and healthy donors: implication for assessment of disease activity and treatment. J Neuroimmunol 2008; 199: 142–150.
    1. Arnulf B, Lecourt S, Soulier J, et al. . Phenotypic and functional characterization of bone marrow mesenchymal stem cells derived from patients with multiple myeloma. Leukemia 2007; 21: 158–163.
    1. Chao YH, Peng CT, Harn HJ, et al. . Poor potential of proliferation and differentiation in bone marrow mesenchymal stem cells derived from children with severe aplastic anemia. Ann Hematol 2010; 89: 715–723.
    1. Chen L, Xu Y, Zhao J, et al. . Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice. PLoS One 2014; 9: e96161.
    1. Hsiao ST, Asgari A, Lokmic Z, et al. . Comparative analysis of paracrine factor expression in human adult mesenchymal stem cells derived from bone marrow, adipose, and dermal tissue. Stem Cells Dev 2011; 21: 2189–2203.
    1. Lappalainen U, Whitsett JA, Wert SE, et al. . Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am J Respir Cell Mol Biol 2005; 32: 311–318.
    1. Lucey EC, Keane J, Kuang PP, et al. . Severity of elastase-induced emphysema is decreased in tumor necrosis factor-alpha and interleukin-1 beta receptor-deficient mice. Lab Invest 2002; 82: 79–85.
    1. Geisler S, Textor M, Schmidt-Bleek K, et al. . In serum veritas—in serum sanitas? Cell non-autonomous aging compromises differentiation and survival of mesenchymal stromal cells via the oxidative stress pathway. Cell Death Dis 2013; 4: e970.
    1. Kim M, Kim C, Choi YS, et al. . Age-related alterations in mesenchymal stem cells related to shift in differentiation from osteogenic to adipogenic potential: implication to age-associated bone diseases and defects. Mech Ageing Dev 2012; 133: 215–225.
    1. Moerman EJ, Teng K, Lipschitz DA, et al. . Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR-γ2 transcription factor and TGF-β/BMP signaling pathways. Aging Cell 2004; 3: 379–389.
    1. Sethe S, Scutt A, Stolzing A. Aging of mesenchymal stem cells. Age Res Rev 2006; 5: 91–116.
    1. Ito K, Barnes PJ. COPD as a disease of accelerated lung aging. Chest 2009; 135: 173–180.
    1. Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature 2000; 408: 239–247.
    1. Kasper G, Mao L, Geissler S, et al. . Insights into mesenchymal stem cell aging: involvement of antioxidant defense and actin cytoskeleton. Stem Cells 2009; 27: 1288–1297.
    1. Rahman I, Swarska E, Henry M, et al. . Is there any relationship between plasma antioxidant capacity and lung function in smokers and in patients with chronic obstructive pulmonary disease? Thorax 2000; 55: 189–193.
    1. Pierrou S, Broberg P, O'Donnell RA, et al. . Expression of genes involved in oxidative stress responses in airway epithelial cells of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2007; 175: 577–586.
    1. Balan M, Teran EM, Waaga-Gasser AM, et al. . Novel roles of c-Met in the survival of renal cancer cells through the regulation of HO-1 and PD-L1 expression. J Biol Chem 2015; 290: 8110–8120.
    1. Yan W, Chen X. GPX2, a direct target of p63, inhibits oxidative stress-induced apoptosis in a p53-dependent manner. J Biol Chem 2006; 281: 7856–7862.
    1. Korashy HM, El-Kadi AO. Transcriptional regulation of the NAD(P)H:quinone oxidoreductase 1 and glutathione S-transferase YA genes by mercury, lead, and copper. Drug Metab Dispos 2006; 34: 152–165.
    1. Thai P, Statt S, Chen CH, et al. . Characterization of a novel long noncoding RNA, SCAL1, induced by cigarette smoke and elevated in lung cancer cell lines. Am J Respir Cell Mol Biol 2013; 49: 204–211.
    1. Ng TK, Carballosa CM, Pelaez D, et al. . Nicotine alters microRNA expression and hinders human adult stem cell regenerative potential. Stem Cells Dev 2012; 22: 781–790.
    1. Le Blanc K, Rasmusson I, Sundberg B, et al. . Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004; 363: 1439–1441.
    1. Hare JM, Fishman JE, Gerstenblith G, et al. . Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA 2012; 308: 2369–2379.
    1. Huang XP, Sun Z, Miyagi Y, et al. . Differentiation of allogeneic mesenchymal stem cells induces immunogenicity and limits their long-term benefits for myocardial repair. Circulation 2010; 122: 2419–2429.

Source: PubMed

3
Sottoscrivi