DOI QR코드

DOI QR Code

Gene Profiles in a Smoke-Induced COPD Mouse Lung Model Following Treatment with Mesenchymal Stem Cells

  • Received : 2016.04.11
  • Accepted : 2016.09.07
  • Published : 2016.10.31

Abstract

Mesenchymal stem cells (MSCs) effectively reduce airway inflammation and regenerate the alveolus in cigarette- and elastase-induced chronic obstructive pulmonary disease (COPD) animal models. The effects of stem cells are thought to be paracrine and immune-modulatory because very few stem cells remain in the lung one day after their systemic injection, which has been demonstrated previously. In this report, we analyzed the gene expression profiles to compare mouse lungs with chronic exposure to cigarette smoke with non-exposed lungs. Gene expression profiling was also conducted in a mouse lung tissue with chronic exposure to cigarette smoke following the systemic injection of human cord blood-derived mesenchymal stem cells (hCB-MSCs). Globally, 834 genes were differentially expressed after systemic injection of hCB-MSCs. Seven and 21 genes, respectively, were up-and downregulated on days 1, 4, and 14 after HCB-MSC injection. The Hbb and Hba, genes with oxygen transport and antioxidant functions, were increased on days 1 and 14. A serine protease inhibitor was also increased at a similar time point after injection of hCB-MSCs. Gene Ontology analysis indicated that the levels of genes related to immune responses, metabolic processes, and blood vessel development were altered, indicating host responses after hCB-MSC injection. These gene expression changes suggest that MSCs induce a regeneration mechanism against COPD induced by cigarette smoke. These analyses provide basic data for understanding the regeneration mechanisms promoted by hCB-MSCs in cigarette smoke-induced COPD.

Keywords

References

  1. Aaron, S.D., Vandemheen, K.L., Fergusson, D., Maltais, F., Bourbeau, J., Goldstein, R., Balter, M., O'Donnell, D., McIvor, A., Sharma, S., et al. (2007). Tiotropium in combination with placebo, salmeterol, or fluticasone-salmeterol for treatment of chronic obstructive pulmonary disease: a randomized trial. Ann. Intern. Med. 146, 545-555. https://doi.org/10.7326/0003-4819-146-8-200704170-00152
  2. Ahmad, M.K., and Mahmood, R. (2016). Protective effect of taurine against potassium bromate-induced hemoglobin oxidation, oxidative stress, and impairment of antioxidant defense system in blood. Environ. Toxicol. 31, 304-313. https://doi.org/10.1002/tox.22045
  3. Banks, M.F., Gerasimovskaya, E.V., Tucker, D.A., Frid, M.G., Carpenter, T.C., and Stenmark, K.R. (2005). Egr-1 antisense oligonucleotides inhibit hypoxia-induced proliferation of pulmonary artery adventitial fibroblasts. J. Appl. Physiol. (1985) 98, 732-738. https://doi.org/10.1152/japplphysiol.00821.2004
  4. Benarafa, C., Priebe, G.P., and Remold-O'Donnell, E. (2007). The neutrophil serine protease inhibitor serpinb1 preserves lung defense functions in Pseudomonas aeruginosa infection. J. Exp. Med. 204, 1901-1909. https://doi.org/10.1084/jem.20070494
  5. Calverley, P.M., Anderson, J.A., Celli, B., Ferguson, G.T., Jenkins, C., Jones, P.W., Yates, J.C., and Vestbo, J. (2007). Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N. Engl. J. Med. 356, 775-789. https://doi.org/10.1056/NEJMoa063070
  6. Chen, Z.H., Kim, H.P., Ryter, S.W., and Choi, A.M. (2008). Identifying targets for COPD treatment through gene expression analyses. Int. J. Chron. Obstruct. Pulmon. Dis. 3, 359-370. https://doi.org/10.2147/COPD.S1758
  7. Grek, C.L., Newton, D.A., Spyropoulos, D.D., and Baatz, J.E. (2011). Hypoxia up-regulates expression of hemoglobin in alveolar epithelial cells. Am. J. Respir. Cell Mol. Biol. 44, 439-447. https://doi.org/10.1165/rcmb.2009-0307OC
  8. Halappanavar, S., Russell, M., Stampfli, M.R., Williams, A., and Yauk, C.L. (2009). Induction of the interleukin 6/ signal transducer and activator of transcription pathway in the lungs of mice sub-chronically exposed to mainstream tobacco smoke. BMC Med. Genomics 2, 56. https://doi.org/10.1186/1755-8794-2-56
  9. Hautamaki, R.D., Kobayashi, D.K., Senior, R.M., and Shapiro, S.D. (1997). Requirement for macrophage elastase for cigarette smoke-induced emphysema in mice. Science 277, 2002-2004. https://doi.org/10.1126/science.277.5334.2002
  10. Henning, R.J., Sanberg, P., and Jimenez, E. (2014). Human cord blood stem cell paracrine factors activate the survival protein kinase Akt and inhibit death protein kinases JNK and p38 in injured cardiomyocytes. Cytotherapy 16, 1158-1168. https://doi.org/10.1016/j.jcyt.2014.01.415
  11. Huffman, L.J., Miles, P.R., Shi, X., and Bowman, L. (2000). Hemoglobin potentiates the production of reactive oxygen species by alveolar macrophages. Exp. Lung Res. 26, 203-217. https://doi.org/10.1080/019021400269871
  12. Huh, J.W., Kim, S.Y., Lee, J.H., Lee, J.S., Van Ta, Q., Kim, M., Oh, Y.M., Lee, Y.S., and Lee, S.D. (2011). Bone marrow cells repair cigarette smoke-induced emphysema in rats. Am. J. Physiol. Lung Cell Mol. Physiol. 301, L255-266. https://doi.org/10.1152/ajplung.00253.2010
  13. Katsha, A.M., Ohkouchi, S., Xin, H., Kanehira, M., Sun, R., Nukiwa, T., and Saijo, Y. (2011). Paracrine factors of multipotent stromal cells ameliorate lung injury in an elastase-induced emphysema model. Mol. Ther. 19, 196-203. https://doi.org/10.1038/mt.2010.192
  14. Kim, Y.S., Kim, J.Y., Shin, D.M., Huh, J.W., Lee, S.W., and Oh, Y.M. (2014). Tracking intravenous adipose-derived mesenchymal stem cells in a model of elastase-induced emphysema. Tuberc. Respir. Dis. (Seoul) 77, 116-123. https://doi.org/10.4046/trd.2014.77.3.116
  15. Lee, R.H., Pulin, A.A., Seo, M.J., Kota, D.J., Ylostalo, J., Larson, B.L., Semprun-Prieto, L., Delafontaine, P., and Prockop, D.J. (2009). Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell 5, 54-63. https://doi.org/10.1016/j.stem.2009.05.003
  16. Liang, X., Ding, Y., Zhang, Y., Tse, H.F., and Lian, Q. (2014). Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives. Cell Transplant. 23, 1045-1059. https://doi.org/10.3727/096368913X667709
  17. Macari, E.R., and Lowrey, C.H. (2011). Induction of human fetal hemoglobin via the NRF2 antioxidant response signaling pathway. Blood 117, 5987-5997. https://doi.org/10.1182/blood-2010-10-314096
  18. MacNee, W., and Tuder, R.M. (2009). New paradigms in the pathogenesis of chronic obstructive pulmonary disease I. Proc. Am. Thorac. Soc. 6, 527-531. https://doi.org/10.1513/pats.200905-027DS
  19. Murphy, M.B., Moncivais, K., and Caplan, A.I. (2013). Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp. Mol. Med. 45, e54. https://doi.org/10.1038/emm.2013.94
  20. Newton, D.A., Rao, K.M., Dluhy, R.A., and Baatz, J.E. (2006). Hemoglobin is expressed by alveolar epithelial cells. J. Biol. Chem. 281, 5668-5676. https://doi.org/10.1074/jbc.M509314200
  21. Rabe, K.F., Hurd, S., Anzueto, A., Barnes, P.J., Buist, S.A., Calverley, P., Fukuchi, Y., Jenkins, C., Rodriguez-Roisin, R., van Weel, C., 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. https://doi.org/10.1164/rccm.200703-456SO
  22. Rangasamy, T., Misra, V., Zhen, L., Tankersley, C.G., Tuder, R.M., and Biswal, S. (2009). Cigarette smoke-induced emphysema in A/J mice is associated with pulmonary oxidative stress, apoptosis of lung cells, and global alterations in gene expression. Am. J. Physiol. Lung Cell Mol. Physiol. 296, L888-900. https://doi.org/10.1152/ajplung.90369.2008
  23. Schweitzer, K.S., Johnstone, B.H., Garrison, J., Rush, N.I., Cooper, S., Traktuev, D.O., Feng, D., Adamowicz, J.J., Van Demark, M., Fisher, A.J., et al. (2011). Adipose stem cell treatment in mice attenuates lung and systemic injury induced by cigarette smoking. Am. J. Respir. Crit. Care Med. 183, 215-225. https://doi.org/10.1164/rccm.201001-0126OC
  24. Shapiro, S.D. (1995). The pathogenesis of emphysema: the elastase:antielastase hypothesis 30 years later. Proc. Assoc. Am. Phys. 107, 346-352.
  25. Shapiro, S.D., Goldstein, N.M., Houghton, A.M., Kobayashi, D.K., Kelley, D., and Belaaouaj, A. (2003). Neutrophil elastase contributes to cigarette smoke-induced emphysema in mice. Am. J. Pathol. 163, 2329-2335. https://doi.org/10.1016/S0002-9440(10)63589-4
  26. Stone, P.J. (1983). The elastase-antielastase hypothesis of the pathogenesis of emphysema. Clin. Chest Med. 4, 405-412.
  27. Suga, H., Glotzbach, J.P., Sorkin, M., Longaker, M.T., and Gurtner, G.C. (2014). Paracrine mechanism of angiogenesis in adipose-derived stem cell transplantation. Ann. Plast. Surg. 72, 234-241. https://doi.org/10.1097/SAP.0b013e318264fd6a
  28. Yang, J.Y., Jin, J., Zhang, Z., Zhang, L., and Shen, C. (2013). Integration microarray and regulation datasets for chronic obstructive pulmonary disease. Eur. Rev. Med. Pharmacol. Sci. 17, 1923-1931.
  29. Yoshida, T., and Tuder, R.M. (2007). Pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease. Physiol. Rev. 87, 1047-1082. https://doi.org/10.1152/physrev.00048.2006
  30. Zandi-Nejad, K., Takakura, A., Jurewicz, M., Chandraker, A.K., Offermanns, S., Mount, D., and Abdi, R. (2013). The role of HCA2 (GPR109A) in regulating macrophage function. FASEB J. 27, 4366-4374. https://doi.org/10.1096/fj.12-223933

Cited by

  1. Mesenchymal stromal cell therapy in COPD: from bench to bedside vol.12, pp.None, 2016, https://doi.org/10.2147/copd.s146671
  2. Cell therapy for lung disease vol.26, pp.144, 2016, https://doi.org/10.1183/16000617.0044-2017
  3. Adipose Tissue-Derived Stem Cells Have the Ability to Differentiate into Alveolar Epithelial Cells and Ameliorate Lung Injury Caused by Elastase-Induced Emphysema in Mice vol.2019, pp.None, 2019, https://doi.org/10.1155/2019/5179172
  4. Mesenchymal Stem Cells Suppress Severe Asthma by Directly Regulating Th2 Cells and Type 2 Innate Lymphoid Cells vol.44, pp.8, 2021, https://doi.org/10.14348/molcells.2021.0101
  5. Current therapeutic strategies for respiratory diseases using mesenchymal stem cells vol.2, pp.3, 2016, https://doi.org/10.1002/mco2.74
  6. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles ameliorate airway inflammation in a rat model of chronic obstructive pulmonary disease (COPD) vol.12, pp.1, 2016, https://doi.org/10.1186/s13287-020-02088-6