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

Winifred Broekman, Gimano D Amatngalim, Yvonne de Mooij-Eijk, Jaap Oostendorp, Helene Roelofs, Christian Taube, Jan Stolk, Pieter S Hiemstra, Winifred Broekman, Gimano D Amatngalim, Yvonne de Mooij-Eijk, Jaap Oostendorp, Helene Roelofs, Christian Taube, Jan Stolk, Pieter S Hiemstra

Abstract

Background: Mesenchymal stromal cells (MSCs) are investigated for their potential to reduce inflammation and to repair damaged tissue. Inflammation and tissue damage are hallmarks of chronic obstructive pulmonary disease (COPD) and MSC infusion is a promising new treatment for COPD. Inflammatory mediators attract MSCs to sites of inflammation and affect their immune-modulatory properties, but little is known about their effect on regenerative properties of MSCs. This study investigates the effect of the pro-inflammatory cytokines TNF-α and IL-1β on the regenerative potential of MSCs, using an in vitro wound healing model of airway epithelial cells.

Methods: Standardized circular wounds were created by scraping cultures of the airway epithelial cell line NCI-H292 and primary bronchial epithelial cells cultured at the air-liquid interface (ALI-PBEC), and subsequently incubated with MSC conditioned medium (MSC-CM) that was generated in presence or absence of TNF-α/IL-1β. Remaining wound size was measured up to 72 h. Phosphorylation of ERK1/2 by MSC-CM was assessed using Western blot. Inhibitors for EGFR and c-Met signaling were used to investigate the contribution of these receptors to wound closure and to ERK1/2 phosphorylation. Transactivation of EGFR by MSC-CM was investigated using a TACE inhibitor, and RT-PCR was used to quantify mRNA expression of several growth factors in MSCs and NCI-H292.

Results: Stimulation of MSCs with the pro-inflammatory cytokines TNF-α and IL-1β increased the mRNA expression of various growth factors by MCSs and enhanced the regenerative potential of MSCs in an in vitro model of airway epithelial injury using NCI-H292 airway epithelial cells. Conditioned medium from cytokine stimulated MSCs induced ERK1/2 phosphorylation in NCI-H292, predominantly via EGFR; it induced ADAM-mediated transactivation of EGFR, and it induced airway epithelial expression of several EGFR ligands. The contribution of activation of c-Met via HGF to increased repair could not be confirmed by inhibitor experiments.

Conclusion: Our data imply that at sites of tissue damage, when inflammatory mediators are present, for example in lungs of COPD patients, MSCs become more potent inducers of repair, in addition to their well-known immune-modulatory properties.

Figures

Fig. 1
Fig. 1
Stimulation of MSCs with TNF-α and IL-1β increases the expression of several growth factors. MSCs were stimulated with TNF-α and IL-1β 20 ng/ml each and harvested for RNA extraction after 6 h. mRNA expression of various growth factors (AREG, EGF, HBEGF (all EGFR ligands), FGF2, HGF, IL6, PDGFA, and VEGF) was determined by qPCR, which showed a significant increase of FGF2, HBEGF, HGF and of IL6, and an increase of AREG and EGF. Values were normalized to ACTB and GAPDH reference genes. Box and whiskers represent median, interquartile range and minimum and maximum for n = 4 obtained from three different donors; (*) p < 0.05
Fig. 2
Fig. 2
MSC-CM increases wound closure. a, b: NCI-H292 cells were injured by making a circular wound with a diameter of 3 mm, and subsequently incubated with MSC-CMCTRL, MSC-CMSTIM, DMEMCTRL and DMEMSTIM, and a negative control (NC, RPMI only) and positive control (RPMI supplemented with TGF-α 20 ng/ml). The wound size was measured at 0, 24, 48 and 72 h after wounding. MSC-CMSTIM significantly increased wound closure. The effect of MSC-CMSTIM was dose-dependent. Error bars represent standard error of the mean (SEM). n = 4-6; (*) p < 0.05 TGF-α compared to NC and (**) compared to DMEM; (***) p < 0.05 MSC-CM compared to NC and (****) compared to DMEM; (*****) p < 0.05 DMEMSTIM compared to NC. c: Morphology of the closure of the wound: photos in the upper panel are taken at t = 0 h, the lower panel shows the same wounds photographed 48 h later. The two photos on the left side are obtained from MSC-CMSTIM stimulated cells, whereas photos at the right represent its control, DMEMSTIM. d: Dose response. Box and whiskers represent median, interquartile range and minimum and maximum. n = 4-6; (*) p < 0.05. e: Wound closure in ALI-PBEC measured at 6 and 24 h (at 48 h all wounds were closed). At 24 h, MSC-CMSTIM significantly enhanced wound healing compared to the NC, but no significant differences were observed compared to its control, DMEMSTIM. Error bars represent SEM, n = 4; (*) p < 0.05 for MSC-CMSTIM compared to NC. f. Wound closure in ALI-PBEC at 24 h. Box and whiskers represent median, interquartile range and minimum and maximum, for n = 4 as in Fig. 2f; (*) p < 0.05
Fig. 3
Fig. 3
MSC-CMSTIM increases ERK1/2 phosphorylation. NCI-H292 cells were incubated with MSC-CMSTIM, DMEMSTIM, DMEMCTRL or TGF-α 20 ng/ml (pos ctrl). ERK1/2 phosphorylation and total ERK was determined in cell lysates using Western blot. a: After 15 min incubation time MSC-CMSTIM increased ERK1/2 phosphorylation compared to its control. b: Densitometry for Fig. 3a. Error bars represent SEM, n = 6; (*) p < 0.05. c: Time course experiment, demonstrating that the effect of MSC-CMSTIM was most prominent up to 30 min, but could still be observed up to 6 h. DMEMCTRL was obtained at 15 min. d: Densitometry for Fig. 3c. Error bars represent SEM, n = 4-5; (*) p < 0.05
Fig. 4
Fig. 4
MSC-CMSTIM induced ERK1/2 phosphorylation is mediated via (trans)activation of EGFR. a: 1 h pre-incubation of NCI-H292 with 0.05 μM PF04217903 and/or 10 μM AG1478, followed by 15 min stimulation with MSC-CMSTIM or DMEMSTIM, or DMEMCTRL showed that inhibition of EGFR decreased ERK1/2 phosphorylation as determined using Western blot. b: Densitometry for Fig. 4a. Error bars represent SEM, n = 5; (*) p < 0.05. c: 1 h pre-incubation with 2 μg/ml neutralizing anti-EGFR antibodies and 10 μM TAPI-1 followed by incubation with MSC-CMSTIM during 15 min showed that these agents decreased ERK1/2 phosphorylation. d: Densitometry for Fig. 4c. Error bars represent SEM, n = 3; (*) p < 0.05. e: NCI-H292 cells were incubated with MSC-CMSTIM or DMEMSTIM and harvested after 9 h for mRNA analysis. mRNA expression of the EGFR ligands AREG, HBEGF and TGFA was determined by qPCR. MSC-CMSTIM significantly increased the expression of AREG and HBEGF. Expression of TGFA increased but this was not significant. f: mRNA expression of the proliferation marker CCDN1 was significantly increased. Values were normalized against RPL13A and ATP5B reference genes. Box and whiskers represent median, interquartile range and minimum and maximum. n = 4; (*) p < 0.05
Fig. 5
Fig. 5
Blocking of EGFR reduces the stimulatory effect of MSC-CMSTIM on wound closure. a: NCI-H292 wounded cell layers were stimulated with MSC-CMSTIM or DMEMSTIM, DMEMCTRL, or DMEM supplemented with TGF-α 20 ng/ml or HGF 20 ng/ml (positive controls). Inhibitors of EGFR (0.2 μM AG1478) or c-Met (0.05 μM PF04217903) were added during the full culture period in assigned conditions. At 48 h, HGF and TGF-α both significantly enhanced wound healing in NCI-H292 cells, which was averted to a level comparable to the negative control by their respective inhibitors. b: In the presence of the EGFR inhibitor, the wound healing capacity of MSC-CMSTIM decreased to values below those observed in the negative control as determined after 48 h. Inhibition of c-Met alone had no effect on wound healing induced by MSC-CMSTIM. Box and whiskers represent median, interquartile range and minimum and maximum. n = 4-7; (*) p < 0.05
Fig. 6
Fig. 6
Proposed model of enhanced wound closure in airway epithelial cells by MSC-CMSTIM. In MSCs, exposure to TNF-α and IL-1β increases the mRNA expression of various growth factors. Conditioned medium from these MSCs contributes to wound healing of airway epithelial cells via three mechanisms: direct activation of EGFR; activation of matrix metalloproteases which results in shedding of membrane bound EGFR ligands; induction of mRNA expression of EGFR ligands by the airway epithelium. Activation of the downstream MAP kinase ERK1/2, which is known as a regulator of cell proliferation (a.o. assessed by increased Cyclin D1) and differentiation, subsequently results in increased wound healing

References

    1. Barnes PJ. Chronic obstructive pulmonary disease. N Engl J Med. 2000;343:269–80. doi: 10.1056/NEJM200007273430407.
    1. Brusselle GG, Joos GF, Bracke KR. New insights into the immunology of chronic obstructive pulmonary disease. Lancet. 2011;378:1015–26. doi: 10.1016/S0140-6736(11)60988-4.
    1. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2007;176:532–55. doi: 10.1164/rccm.200703-456SO.
    1. Barnes PJ. New anti-inflammatory targets for chronic obstructive pulmonary disease. Nat Rev Drug Discov. 2013;12:543–59. doi: 10.1038/nrd4025.
    1. Dominici M, Le BK, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7. doi: 10.1080/14653240600855905.
    1. Meirelles LS, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev. 2009;20:419–27. doi: 10.1016/j.cytogfr.2009.10.002.
    1. Nauta AJ, Fibbe WE. Immunomodulatory properties of mesenchymal stromal cells. Blood. 2007;110:3499–506. doi: 10.1182/blood-2007-02-069716.
    1. Guan XJ, Song L, Han FF, Cui ZL, Chen X, Guo XJ, et al. Mesenchymal stem cells protect cigarette smoke-damaged lung and pulmonary function partly via VEGF-VEGF receptors. J Cell Biochem. 2013;114:323–35. doi: 10.1002/jcb.24377.
    1. Huh JW, Kim SY, Lee JH, Lee JS, Van TQ, Kim M, et al. Bone marrow cells repair cigarette smoke-induced emphysema in rats. Am J Physiol Lung Cell Mol Physiol. 2011;301:L255–66. doi: 10.1152/ajplung.00253.2010.
    1. Katsha AM, Ohkouchi S, Xin H, Kanehira M, Sun R, Nukiwa T, et al. Paracrine factors of multipotent stromal cells ameliorate lung injury in an elastase-induced emphysema model. Mol Ther. 2011;19:196–203. doi: 10.1038/mt.2010.192.
    1. Kim JS, McKinnis VS, Nawrocki A, White SR. Stimulation of migration and wound repair of guinea-pig airway epithelial cells in response to epidermal growth factor. Am J Respir Cell Mol Biol. 1998;18:66–74. doi: 10.1165/ajrcmb.18.1.2740.
    1. Panos RJ, Patel R, Bak PM. Intratracheal administration of hepatocyte growth factor/scatter factor stimulates rat alveolar type II cell proliferation in vivo. Am J Respir Cell Mol Biol. 1996;15:574–81. doi: 10.1165/ajrcmb.15.5.8918364.
    1. Crosby LM, Waters CM. Epithelial repair mechanisms in the lung. Am J Physiol Lung Cell Mol Physiol. 2010;298:L715–31. doi: 10.1152/ajplung.00361.2009.
    1. Ryan RM, Mineo-Kuhn MM, Kramer CM, Finkelstein JN. Growth factors alter neonatal type II alveolar epithelial cell proliferation. Am J Physiol. 1994;266:L17–22.
    1. Leslie CC, McCormick-Shannon K, Shannon JM, Garrick B, Damm D, Abraham JA, et al. Heparin-binding EGF-like growth factor is a mitogen for rat alveolar type II cells. Am J Respir Cell Mol Biol. 1997;16:379–87. doi: 10.1165/ajrcmb.16.4.9115748.
    1. Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature. 2001;410:37–40. doi: 10.1038/35065000.
    1. Akram KM, Samad S, Spiteri MA, Forsyth NR. Mesenchymal stem cells promote alveolar epithelial cell wound repair in vitro through distinct migratory and paracrine mechanisms. Respir Res. 2013;14:9. doi: 10.1186/1465-9921-14-9.
    1. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25:2739–49. doi: 10.1634/stemcells.2007-0197.
    1. Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell. 2009;4:206–16. doi: 10.1016/j.stem.2009.02.001.
    1. Crisostomo PR, Wang Y, Markel TA, Wang M, Lahm T, Meldrum DR. Human mesenchymal stem cells stimulated by TNF-alpha, LPS, or hypoxia produce growth factors by an NF kappa B- but not JNK-dependent mechanism. Am J Physiol Cell Physiol. 2008;294:C675–82. doi: 10.1152/ajpcell.00437.2007.
    1. Miettinen JA, Pietila M, Salonen RJ, Ohlmeier S, Ylitalo K, Huikuri HV, et al. Tumor necrosis factor alpha promotes the expression of immunosuppressive proteins and enhances the cell growth in a human bone marrow-derived stem cell culture. Exp Cell Res. 2011;317:791–801. doi: 10.1016/j.yexcr.2010.12.010.
    1. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2:141–50. doi: 10.1016/j.stem.2007.11.014.
    1. Ryan JM, Barry F, Murphy JM, Mahon BP. Interferon-gamma does not break, but promotes the immunosuppressive capacity of adult human mesenchymal stem cells. Clin Exp Immunol. 2007;149:353–63. doi: 10.1111/j.1365-2249.2007.03422.x.
    1. Ren G, Zhao X, Zhang L, Zhang J, L'Huillier A, Ling W, et al. Inflammatory cytokine-induced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in mesenchymal stem cells are critical for immunosuppression. J Immunol. 2010;184:2321–8. doi: 10.4049/jimmunol.0902023.
    1. Heo SC, Jeon ES, Lee IH, Kim HS, Kim MB, Kim JH. Tumor necrosis factor-alpha-activated human adipose tissue-derived mesenchymal stem cells accelerate cutaneous wound healing through paracrine mechanisms. J Invest Dermatol. 2011;131:1559–67. doi: 10.1038/jid.2011.64.
    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;153:530–4. doi: 10.1164/ajrccm.153.2.8564092.
    1. Lappalainen U, Whitsett JA, Wert SE, Tichelaar JW, Bry K. Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am J Respir Cell Mol Biol. 2005;32:311–8. doi: 10.1165/rcmb.2004-0309OC.
    1. Lucey EC, Keane J, Kuang PP, Snider GL, Goldstein RH. Severity of elastase-induced emphysema is decreased in tumor necrosis factor-alpha and interleukin-1beta receptor-deficient mice. Lab Invest. 2002;82:79–85. doi: 10.1038/labinvest.3780397.
    1. Pauwels NS, Bracke KR, Dupont LL, Van Pottelberge GR, Provoost S, Vanden Berghe T, et al. Role of IL-1alpha and the Nlrp3/caspase-1/IL-1beta axis in cigarette smoke-induced pulmonary inflammation and COPD. Eur Respir J. 2011;38:1019–28. doi: 10.1183/09031936.00158110.
    1. Amatngalim GD, Van WY, de Mooij-Eijk Y, Verhoosel RM, Harder J, Lekkerkerker AN, et al. Basal cells contribute to innate immunity of the airway epithelium through production of the antimicrobial protein RNase 7. J Immunol. 2015;194:3340–50. doi: 10.4049/jimmunol.1402169.
    1. Duijvestein M, Vos AC, Roelofs H, Wildenberg ME, Wendrich BB, Verspaget HW, et al. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn's disease: results of a phase I study. Gut. 2010;59:1662–9. doi: 10.1136/gut.2010.215152.
    1. Aarbiou J, Verhoosel RM, Van WS, de Boer WI, van Krieken JH, Litvinov SV, et al. Neutrophil defensins enhance lung epithelial wound closure and mucin gene expression in vitro. Am J Respir Cell Mol Biol. 2004;30:193–201. doi: 10.1165/rcmb.2002-0267OC.
    1. Aarbiou J, Ertmann M, Van WS, Van NP, Rook D, Rabe KF, et al. Human neutrophil defensins induce lung epithelial cell proliferation in vitro. J Leukoc Biol. 2002;72:167–74.
    1. Burgel PR, Nadel JA. Epidermal growth factor receptor-mediated innate immune responses and their roles in airway diseases. Eur Respir J. 2008;32:1068–81. doi: 10.1183/09031936.00172007.
    1. Treinies I, Paterson HF, Hooper S, Wilson R, Marshall CJ. Activated MEK stimulates expression of AP-1 components independently of phosphatidylinositol 3-kinase (PI3-kinase) but requires a PI3-kinase signal To stimulate DNA synthesis. Mol Cell Biol. 1999;19:321–9. doi: 10.1128/MCB.19.1.321.
    1. Yew TL, Hung YT, Li HY, Chen HW, Chen LL, Tsai KS, et al. Enhancement of wound healing by human multipotent stromal cell conditioned medium: the paracrine factors and p38 MAPK activation. Cell Transplant. 2011;20:693–706. doi: 10.3727/096368910X550198.
    1. Ries C, Egea V, Karow M, Kolb H, Jochum M, Neth P. MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood. 2007;109:4055–63. doi: 10.1182/blood-2006-10-051060.
    1. Bernardo ME, Fibbe WE. Mesenchymal stromal cells: sensors and switchers of inflammation. Cell Stem Cell. 2013;13:392–402. doi: 10.1016/j.stem.2013.09.006.
    1. Wang M, Crisostomo PR, Herring C, Meldrum KK, Meldrum DR. Human progenitor cells from bone marrow or adipose tissue produce VEGF, HGF, and IGF-I in response to TNF by a p38 MAPK-dependent mechanism. Am J Physiol Regul Integr Comp Physiol. 2006;291:R880–4. doi: 10.1152/ajpregu.00280.2006.
    1. Zhang A, Wang Y, Ye Z, Xie H, Zhou L, Zheng S. Mechanism of TNF-alpha-induced migration and hepatocyte growth factor production in human mesenchymal stem cells. J Cell Biochem. 2010;111:469–75. doi: 10.1002/jcb.22729.
    1. Koff JL, Shao MX, Kim S, Ueki IF, Nadel JA. Pseudomonas lipopolysaccharide accelerates wound repair via activation of a novel epithelial cell signaling cascade. J Immunol. 2006;177:8693–700. doi: 10.4049/jimmunol.177.12.8693.
    1. Puddicombe SM, Polosa R, Richter A, Krishna MT, Howarth PH, Holgate ST, et al. Involvement of the epidermal growth factor receptor in epithelial repair in asthma. FASEB J. 2000;14:1362–74. doi: 10.1096/fj.14.10.1362.
    1. Ohmichi H, Matsumoto K, Nakamura T. In vivo mitogenic action of HGF on lung epithelial cells: pulmotrophic role in lung regeneration. Am J Physiol. 1996;270:L1031–9.
    1. Zahm JM, Debordeaux C, Raby B, Klossek JM, Bonnet N, Puchelle E. Motogenic effect of recombinant HGF on airway epithelial cells during the in vitro wound repair of the respiratory epithelium. J Cell Physiol. 2000;185:447–53. doi: 10.1002/1097-4652(200012)185:3<447::AID-JCP16>;2-D.
    1. Curley GF, Hayes M, Ansari B, Shaw G, Ryan A, Barry F, et al. Mesenchymal stem cells enhance recovery and repair following ventilator-induced lung injury in the rat. Thorax. 2012;67:496–501. doi: 10.1136/thoraxjnl-2011-201059.
    1. Wang WC, Kuo CY, Tzang BS, Chen HM, Kao SH. IL-6 augmented motility of airway epithelial cell BEAS-2B via Akt/GSK-3beta signaling pathway. J Cell Biochem. 2012;113:3567–75. doi: 10.1002/jcb.24235.
    1. Newland N, Richter A. Agents associated with lung inflammation induce similar responses in NCI-H292 lung epithelial cells. Toxicol In Vitro. 2008;22:1782–8. doi: 10.1016/j.tiv.2008.07.009.
    1. Takeyama K, Dabbagh K, Lee HM, Agusti C, Lausier JA, Ueki IF, et al. Epidermal growth factor system regulates mucin production in airways. Proc Natl Acad Sci U S A. 1999;96:3081–6. doi: 10.1073/pnas.96.6.3081.
    1. Zhang Y, Zhu M, Yang Z, Pan X, Jiang Y, Sun C, et al. The human Cathelicidin LL-37 induces MUC5AC mucin production by airway epithelial cells via TACE-TGF-alpha-EGFR pathway. Exp Lung Res. 2014;40:333–42. doi: 10.3109/01902148.2014.926434.
    1. Shao MX, Nakanaga T, Nadel JA. Cigarette smoke induces MUC5AC mucin overproduction via tumor necrosis factor-alpha-converting enzyme in human airway epithelial (NCI-H292) cells. Am J Physiol Lung Cell Mol Physiol. 2004;287:L420–7. doi: 10.1152/ajplung.00019.2004.
    1. Luppi F, Aarbiou J, Van WS, Rahman I, de Boer WI, Rabe KF, et al. Effects of cigarette smoke condensate on proliferation and wound closure of bronchial epithelial cells in vitro: role of glutathione. Respir Res. 2005;6:140. doi: 10.1186/1465-9921-6-140.
    1. Prockop DJ, Brenner M, Fibbe WE, Horwitz E, Le BK, Phinney DG, et al. Defining the risks of mesenchymal stromal cell therapy. Cytotherapy. 2010;12:576–8. doi: 10.3109/14653249.2010.507330.
    1. Sage EK, Kolluri KK, McNulty K, Lourenco SS, Kalber TL, Ordidge KL, et al. Systemic but not topical TRAIL-expressing mesenchymal stem cells reduce tumour growth in malignant mesothelioma. Thorax. 2014;69:638–47. doi: 10.1136/thoraxjnl-2013-204110.
    1. Mohr A, Albarenque SM, Deedigan L, Yu R, Reidy M, Fulda S, et al. Targeting of XIAP combined with systemic mesenchymal stem cell-mediated delivery of sTRAIL ligand inhibits metastatic growth of pancreatic carcinoma cells. Stem Cells. 2010;28:2109–20. doi: 10.1002/stem.533.
    1. Weiss DJ, Casaburi R, Flannery R, LeRoux-Williams M, Tashkin DP. A placebo-controlled, randomized trial of mesenchymal stem cells in COPD. Chest. 2013;143:1590–8. doi: 10.1378/chest.12-2094.

Source: PubMed

3
Suscribir