DOI QR코드

DOI QR Code

Cyclic Phytosphingosine-1-Phosphate Primed Mesenchymal Stem Cells Ameliorate LPS-Induced Acute Lung Injury in Mice

  • Youngheon Park (Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University) ;
  • Jimin Jang (Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University) ;
  • Jooyeon Lee (Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University) ;
  • Hyosin Baek (Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University) ;
  • Jaehyun Park (Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University) ;
  • Sang-Ryul Cha (Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University) ;
  • Se Bi Lee (Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University) ;
  • Sunghun Na (Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University) ;
  • Jae-Woo Kwon (Department of Internal Medicine, School of Medicine, Kangwon National University) ;
  • Seok-Ho Hong (Department of Internal Medicine, School of Medicine, Kangwon National University) ;
  • Se-Ran Yang (Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University)
  • 투고 : 2023.01.02
  • 심사 : 2023.02.06
  • 발행 : 2023.05.30

초록

Background and Objectives: O-cyclic phytosphingosine-1-phosphate (cP1P) is a synthetic chemical and has a structure like sphingosine-1-phosphate (S1P). S1P is known to promote cell migration, invasion, proliferation, and anti-apoptosis through hippocampal signals. However, S1P mediated cellular-, molecular mechanism is still remained in the lung. Acute lung injury (ALI) and its severe form acute respiratory distress syndrome (ARDS) are characterized by excessive immune response, increased vascular permeability, alveolar-peritoneal barrier collapse, and edema. In this study, we determined whether cP1P primed human dermal derived mesenchymal stem cells (hdMSCs) ameliorate lung injury and its therapeutic pathway in ALI mice. Methods and Results: cP1P treatment significantly stimulated MSC migration and invasion ability. In cytokine array, secretion of vascular-related factors was increased in cP1P primed hdMSCs (hdMSCcP1P), and cP1P treatment induced inhibition of Lats while increased phosphorylation of Yap. We next determined whether hdMSCcP1P reduce inflammatory response in LPS exposed mice. hdMSCcP1P further decreased infiltration of macrophage and neutrophil, and release of TNF-α, IL-1β, and IL-6 were reduced rather than naïve hdMSC treatment. In addition, phosphorylation of STAT1 and expression of iNOS were significantly decreased in the lungs of MSCcP1P treated mice. Conclusions: Taken together, these data suggest that cP1P treatment enhances hdMSC migration in regulation of Hippo signaling and MSCcP1P provide a therapeutic potential for ALI/ARDS treatment.

키워드

과제정보

This work was supported by the National Research Foundation (NRF) funded by the Korean government (MSIT) (2020R1A2C2010712, 2020R1A5A8019180), the Korean Fund for Regenerative Medicine (KFRM) (22A0304L1-01).

참고문헌

  1. Bakowitz M, Bruns B, McCunn M. Acute lung injury and the acute respiratory distress syndrome in the injured patient. Scand J Trauma Resusc Emerg Med 2012;20:54 
  2. Esper AM, Martin GS. Evolution of treatments for patients with acute lung injury. Expert Opin Investig Drugs 2005;14:633-645 
  3. Sauer A, Peukert K, Putensen C, Bode C. Antibiotics as immunomodulators: a potential pharmacologic approach for ARDS treatment. Eur Respir Rev 2021;30:210093 
  4. Parekkadan B, Milwid JM. Mesenchymal stem cells as therapeutics. Annu Rev Biomed Eng 2010;12:87-117 
  5. Lin H, Xu R, Zhang Z, Chen L, Shi M, Wang FS. Implications of the immunoregulatory functions of mesenchymal stem cells in the treatment of human liver diseases. Cell Mol Immunol 2011;8:19-22 
  6. De Becker A, Riet IV. Homing and migration of mesenchymal stromal cells: how to improve the efficacy of cell therapy? World J Stem Cells 2016;8:73-87 
  7. Noronha NC, Mizukami A, Caliari-Oliveira C, Cominal JG, Rocha JLM, Covas DT, Swiech K, Malmegrim KCR. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther 2019;10:131 Erratum in: Stem Cell Res Ther 2019;10:132 
  8. English D, Welch Z, Kovala AT, Harvey K, Volpert OV, Brindley DN, Garcia JG. Sphingosine 1-phosphate released from platelets during clotting accounts for the potent endothelial cell chemotactic activity of blood serum and provides a novel link between hemostasis and angiogenesis. FASEB J 2000;14:2255-2265 
  9. Mendelson K, Evans T, Hla T. Sphingosine 1-phosphate signalling. Development 2014;141:5-9 
  10. Lee HJ, Jung YH, Choi GE, Kim JS, Chae CW, Lim JR, Kim SY, Lee JE, Park MC, Yoon JH, Choi MJ, Kim KS, Han HJ. O-cyclic phytosphingosine-1-phosphate stimulates HIF1α-dependent glycolytic reprogramming to enhance the therapeutic potential of mesenchymal stem cells. Cell Death Dis 2019;10:590 
  11. Wick KD, Leligdowicz A, Zhuo H, Ware LB, Matthay MA. Mesenchymal stromal cells reduce evidence of lung injury in patients with ARDS. JCI Insight 2021;6:e148983 
  12. Park JR, Kim E, Yang J, Lee H, Hong SH, Woo HM, Park SM, Na S, Yang SR. Isolation of human dermis derived mesenchymal stem cells using explants culture method: expansion and phenotypical characterization. Cell Tissue Bank 2015;16:209-218 
  13. Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay MA, Slutsky AS, Kuebler WM. An official American Thoracic Society workshop report: features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol 2011;44:725-738 
  14. Xie Z, Liu H, Geng M. Targeting sphingosine-1-phosphate signaling for cancer therapy. Sci China Life Sci 2017;60:585-600 
  15. Kennedy S, Kane KA, Pyne NJ, Pyne S. Targeting sphingosine-1-phosphate signalling for cardioprotection. Curr Opin Pharmacol 2009;9:194-201 
  16. Mahajan-Thakur S, Bien-Moller S, Marx S, Schroeder H, Rauch BH. Sphingosine 1-phosphate (S1P) signaling in glioblastoma multiforme-a systematic review. Int J Mol Sci 2017;18:2448 
  17. Johnson R, Halder G. The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov 2014;13:63-79 
  18. Li L, Dong L, Wang Y, Zhang X, Yan J. Lats1/2-mediated alteration of Hippo signaling pathway regulates the fate of bone marrow-derived mesenchymal stem cells. Biomed Res Int 2018;2018:4387932 
  19. Li L, Dong L, Zhang J, Gao F, Hui J, Yan J. Mesenchymal stem cells with downregulated Hippo signaling attenuate lung injury in mice with lipopolysaccharide‑induced acute respiratory distress syndrome. Int J Mol Med 2019;43:1241-1252 
  20. Yu FX, Zhao B, Guan KL. Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell 2015;163:811-828 
  21. Kalhori V, Tornquist K. MMP2 and MMP9 participate in S1P-induced invasion of follicular ML-1 thyroid cancer cells. Mol Cell Endocrinol 2015;404:113-122 
  22. Burnham EL, Janssen WJ, Riches DW, Moss M, Downey GP. The fibroproliferative response in acute respiratory distress syndrome: mechanisms and clinical significance. Eur Respir J 2014;43:276-285 
  23. Willis GR, Fernandez-Gonzalez A, Anastas J, Vitali SH, Liu X, Ericsson M, Kwong A, Mitsialis SA, Kourembanas S. Mesenchymal stromal cell exosomes ameliorate experimental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. Am J Respir Crit Care Med 2018;197:104-116 
  24. Gho YS, Kleinman HK, Sosne G. Angiogenic activity of human soluble intercellular adhesion molecule-1. Cancer Res 1999;59:5128-5132 
  25. Tang B, Li X, Liu Y, Chen X, Li X, Chu Y, Zhu H, Liu W, Xu F, Zhou F, Zhang Y. The therapeutic effect of ICAM-1-overexpressing mesenchymal stem cells on acute graft-versus-host disease. Cell Physiol Biochem 2018;46:2624-2635 
  26. Sun Z, Gong X, Zhu H, Wang C, Xu X, Cui D, Qian W, Han X. Inhibition of Wnt/β-catenin signaling promotes engraftment of mesenchymal stem cells to repair lung injury. J Cell Physiol 2014;229:213-224 
  27. De Paepe ME, Patel C, Tsai A, Gundavarapu S, Mao Q. Endoglin (CD105) up-regulation in pulmonary microvasculature of ventilated preterm infants. Am J Respir Crit Care Med 2008;178:180-187 
  28. Horie S, Gonzalez HE, Laffey JG, Masterson CH. Cell therapy in acute respiratory distress syndrome. J Thorac Dis 2018;10:5607-5620 
  29. Huh JW, Kim WY, Park YY, Lim CM, Koh Y, Kim MJ, Hong SB. Anti-inflammatory role of mesenchymal stem cells in an acute lung injury mouse model. Acute Crit Care 2018;33:154-161 
  30. Gonzales JN, Lucas R, Verin AD. The acute respiratory distress syndrome: mechanisms and perspective therapeutic approaches. Austin J Vasc Med 2015;2:1009 
  31. Herrero R, Sanchez G, Lorente JA. New insights into the mechanisms of pulmonary edema in acute lung injury. Ann Transl Med 2018;6:32 
  32. Muller JM, Ziegler-Heitbrock HW, Baeuerle PA. Nuclear factor kappa B, a mediator of lipopolysaccharide effects. Immunobiology 1993;187:233-256 
  33. Jayasingam SD, Citartan M, Thang TH, Mat Zin AA, Ang KC, Ch'ng ES. Evaluating the polarization of tumor-associated macrophages into M1 and M2 phenotypes in human cancer tissue: technicalities and challenges in routine clinical practice. Front Oncol 2020;9:1512 
  34. Shin SA, Joo BJ, Lee JS, Ryu G, Han M, Kim WY, Park HH, Lee JH, Lee CS. Phytochemicals as anti-inflammatory agents in animal models of prevalent inflammatory diseases. Molecules 2020;25:5932