Int J Stem Cells.  2023 May;16(2):191-201. 10.15283/ijsc23001.

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

Affiliations
  • 1Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon, Korea
  • 2Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Korea
  • 3Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea

Abstract

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.

Keyword

cP1P; MSC; ALI; HIPPO; STAT1

Figure

  • Fig. 1 An analogous compound of S1P, cP1P does not have cellular toxicity. (A) Structural formula of S1P and cP1P. (B) Cell viability was qua-ntified by MTT assay at various concentrations (Vehicle, 0.5, 1, 5 μM). All data are representative of multiple experiments. CTL: control group, ns: not significant.

  • Fig. 2 hdMSCcP1P was promoted homing by upregulated migration and invasion. (A) hdMSCs scratched by 1 ml tips and then treated to cP1P (0.5 μM, 1 μM, 5 μM) for 24 h. Scratch point captured by microscope (magnification ×40, Scale bars, 100 μm). (B) Wound healing degree of MSC was quantified by migration assay. After 24 h, various concentration was measured and presented as fold change. (C) Invasion assay was measured by trans well. Various concentrations. Original magnification, ×40. Scale bars, 100 μm. Cell numbers of various concentrations was counted and presented as numbers. (D) gene expression level of MMP2 and MMP9 was measured by real-time PCR. All data are representative of multiple experiments. CTL: control group, ns: not significant. *p<0.05, **p<0.01, and ***p<0.001 vs. control group.

  • Fig. 3 Changes in cytokines secreted by cP1P in hdMSC. (A) Heat map of hdMSCcP1P and non-primed hdMSC. Changes of cytokines were measured by cytokine assay kits. (B) Comparison of mean pixel density measurement between hdMSC and cP1P-primed hdMSC. (C) 1 μM cP1P-primed hdMSC and non-primed hdMSC measured mRNA expression level of hippo signaling pathway and migration associated factors by Real-time PCR. (D) Protein level of Lats1, YAP1 measured by Western blot. All data are representative of multiple experiments. CTL: control group, MSCcP1P: 1 μM cP1P-primed hdMSC, ns: not significant. *p<0.05, **p<0.01, and ***p<0.001 vs. MSC group.

  • Fig. 4 hdMSCcP1P ameliorates LPS-induced acute lung injury. (A) Histopathological examinations of lung tissue. Mouse lung sections were stained by H&E staining. Original magnification, ×100. Scale bars, 200 μm. (B) Bar graphs showing blinded analysis of lung injury score. (C) Mouse weight before and after treating LPS was measured in each group. (D) Mouse lung weight per body weight was measured in each group. (E) Total protein in BALF was quantified by BCA assay. All data are representative of multiple experiments. CTL: control group, MSCcP1P: 1 μM cP1P-primed hdMSC, ns: not significant. *p<0.05, **p<0.01, and ***p<0.001 vs. LPS groups; #p<0.05 vs. LPS treated MSCcP1P groups.

  • Fig. 5 hdMSCcP1P ameliorates pro-inflammatory cytokine expression and inflammatory signaling pathway of LPS-induced acute lung injury. (A) Macrophages, neutrophils, lymphocytes, and total cells in BALF was counted at each groups. BALF cell was stained by Giemsa staining and counted each group. (B) Pro-inflammatory cytokines in BALF of each group was measured by ELISA. (C) inflammatory signaling factors of each group were quantified by western blot and expression ratio was measured as fold change. All data are representative of multiple experiments. CTL: control group, MSCcP1P: 1 μM cP1P-primed hdMSC, ns: not significant. *p<0.05, **p<0.01, and ***p<0.001 vs. LPS groups; #p<0.05, ##p<0.01 vs. LPS treated MSCcP1P groups.

  • Fig. 6 cP1P acts as a ligand of the S1P receptor of hdMSCs, promoting Hippo signaling. Increased RhoA and reduced Lats1 cause nucleus-translo-cation of YAP/TAZ. YAP/TAZ acts as a transcription factor in TEAD gene site, increasing the expression of CTGF, AREG, and SLUG genes, enhancing the cell migration, invasion, and immune modulation functions of hdMSCs. Enhanced hdMSCs reduce p-STAT1 and iNOS, attenuating inflammation, and recovering damaged tissue LPS-induced ALI mice.


Cited by  1 articles

Corrigendum to “Cyclic Phytosphingosine-1-Phosphate Primed Mesenchymal Stem Cells Ameliorate LPS-Induced Acute Lung Injury in Mice”
Youngheon Park, Jimin Jang, Jooyeon Lee, Hyosin Baek, Jaehyun Park, Sang-Ryul Cha, Se Bi Lee, Sunghun Na, Jae-Woo Kwon, Young Jun Park, Myeong Jun Choi, Kye-Seong Kim, Seok-Ho Hong, Se-Ran Yang
Int J Stem Cells. 2023;16(4):448-449.    doi: 10.15283/23001C.


Reference

References

1. Bakowitz M, Bruns B, McCunn M. 2012; Acute lung injury and the acute respiratory distress syndrome in the injured patient. Scand J Trauma Resusc Emerg Med. 20:54. DOI: 10.1186/1757-7241-20-54. PMID: 22883052. PMCID: PMC3518173. PMID: 5cbd3e3a1fa741d39d72c9bdb03b20c1.
Article
2. Esper AM, Martin GS. 2005; Evolution of treatments for patients with acute lung injury. Expert Opin Investig Drugs. 14:633–645. DOI: 10.1517/13543784.14.5.633. PMID: 15926869.
Article
3. Sauer A, Peukert K, Putensen C, Bode C. 2021; Antibiotics as immunomodulators: a potential pharmacologic approach for ARDS treatment. Eur Respir Rev. 30:210093. DOI: 10.1183/16000617.0093-2021. PMID: 34615700. PMCID: PMC9489085. PMID: cc74f6659fe04d4e915ac389ca612377.
Article
4. Parekkadan B, Milwid JM. 2010; Mesenchymal stem cells as the-rapeutics. Annu Rev Biomed Eng. 12:87–117. DOI: 10.1146/annurev-bioeng-070909-105309. PMID: 20415588. PMCID: PMC3759519.
Article
5. Lin H, Xu R, Zhang Z, Chen L, Shi M, Wang FS. 2011; Implica-tions of the immunoregulatory functions of mesenchymal stem cells in the treatment of human liver diseases. Cell Mol Immunol. 8:19–22. DOI: 10.1038/cmi.2010.57. PMID: 21200380. PMCID: PMC4002992.
Article
6. De Becker A, Riet IV. 2016; Homing and migration of mesenchymal stromal cells: how to improve the efficacy of cell therapy? World J Stem Cells. 8:73–87. DOI: 10.4252/wjsc.v8.i3.73. PMID: 27022438. PMCID: PMC4807311.
Article
7. Noronha NC, Mizukami A, Caliári-Oliveira C, Cominal JG, Rocha JLM, Covas DT, Swiech K, Malmegrim KCR. 2019; Pri-ming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther. 10:131. Erratum in: Stem Cell Res Ther 2019;10:132. DOI: 10.1186/s13287-019-1224-y. PMID: 31046833. PMCID: PMC6498654. PMID: 3b2ec96227cd4be081df40d2750b7ce1.
Article
8. English D, Welch Z, Kovala AT, Harvey K, Volpert OV, Brindley DN, Garcia JG. 2000; 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. 14:2255–2265. DOI: 10.1096/fj.00-0134com. PMID: 11053247.
Article
9. Mendelson K, Evans T, Hla T. 2014; Sphingosine 1-phosphate signalling. Development. 141:5–9. DOI: 10.1242/dev.094805. PMID: 24346695. PMCID: PMC3865745.
Article
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. 2019; O-cyclic phytosphingosine-1-phosphate stimulates HIF1α-dependent glycolytic reprogramming to enhance the therapeutic potential of mesenchymal stem cells. Cell Death Dis. 10:590. DOI: 10.1038/s41419-019-1823-7. PMID: 31383843. PMCID: PMC6683124.
Article
11. Wick KD, Leligdowicz A, Zhuo H, Ware LB, Matthay MA. 2021; Mesenchymal stromal cells reduce evidence of lung injury in patients with ARDS. JCI Insight. 6:e148983. DOI: 10.1172/jci.insight.148983. PMID: 33974564. PMCID: PMC8262503.
Article
12. Park JR, Kim E, Yang J, Lee H, Hong SH, Woo HM, Park SM, Na S, Yang SR. 2015; Isolation of human dermis derived me-senchymal stem cells using explants culture method: expansion and phenotypical characterization. Cell Tissue Bank. 16:209–218. DOI: 10.1007/s10561-014-9471-8. PMID: 25163610.
Article
13. Matute-Bello G, Downey G, Moore BB, Groshong SD, Mat-thay MA, Slutsky AS, Kuebler WM. 2011; An official American Thoracic Society workshop report: features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol. 44:725–738. DOI: 10.1165/rcmb.2009-0210ST. PMID: 21531958. PMCID: PMC7328339.
Article
14. Xie Z, Liu H, Geng M. 2017; Targeting sphingosine-1-phosphate signaling for cancer therapy. Sci China Life Sci. 60:585–600. DOI: 10.1007/s11427-017-9046-6. PMID: 28623546.
Article
15. Kennedy S, Kane KA, Pyne NJ, Pyne S. 2009; Targeting sphingosine-1-phosphate signalling for cardioprotection. Curr Opin Pharmacol. 9:194–201. DOI: 10.1016/j.coph.2008.11.002. PMID: 19070545.
Article
16. Mahajan-Thakur S, Bien-Möller S, Marx S, Schroeder H, Rauch BH. 2017; Sphingosine 1-phosphate (S1P) signaling in glioblastoma multiforme-a systematic review. Int J Mol Sci. 18:2448. DOI: 10.3390/ijms18112448. PMID: 29149079. PMCID: PMC5713415. PMID: 7b106d275cce41279c64680eb6cae219.
Article
17. Johnson R, Halder G. 2014; The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov. 13:63–79. DOI: 10.1038/nrd4161. PMID: 24336504. PMCID: PMC4167640.
Article
18. Li L, Dong L, Wang Y, Zhang X, Yan J. 2018; Lats1/2-mediated alteration of Hippo signaling pathway regulates the fate of bone marrow-derived mesenchymal stem cells. Biomed Res Int. 2018:4387932. DOI: 10.1155/2018/4387932. PMID: 30671453. PMCID: PMC6323436.
19. Li L, Dong L, Zhang J, Gao F, Hui J, Yan J. 2019; Mesenchymal stem cells with downregulated Hippo signaling attenuate lung injury in mice with lipopolysaccharide‑induced acute respiratory distress syndrome. Int J Mol Med. 43:1241–1252. DOI: 10.3892/ijmm.2018.4047. PMID: 30628652. PMCID: PMC6365074.
Article
20. Yu FX, Zhao B, Guan KL. 2015; Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell. 163:811–828. DOI: 10.1016/j.cell.2015.10.044. PMID: 26544935. PMCID: PMC4638384.
Article
21. Kalhori V, Törnquist K. 2015; MMP2 and MMP9 participate in S1P-induced invasion of follicular ML-1 thyroid cancer cells. Mol Cell Endocrinol. 404:113–122. DOI: 10.1016/j.mce.2015.01.037. PMID: 25643979.
Article
22. Burnham EL, Janssen WJ, Riches DW, Moss M, Downey GP. 2014; The fibroproliferative response in acute respiratory distress syndrome: mechanisms and clinical significance. Eur Respir J. 43:276–285. DOI: 10.1183/09031936.00196412. PMID: 23520315. PMCID: PMC4015132.
Article
23. Willis GR, Fernandez-Gonzalez A, Anastas J, Vitali SH, Liu X, Ericsson M, Kwong A, Mitsialis SA, Kourembanas S. 2018; Mesenchymal stromal cell exosomes ameliorate experi-mental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. Am J Respir Crit Care Med. 197:104–116. DOI: 10.1164/rccm.201705-0925OC. PMID: 28853608. PMCID: PMC5765387.
Article
24. Gho YS, Kleinman HK, Sosne G. 1999; Angiogenic activity of human soluble intercellular adhesion molecule-1. Cancer Res. 59:5128–5132. DOI: 10.32388/v6c2fi. PMID: 10537287.
25. Tang B, Li X, Liu Y, Chen X, Li X, Chu Y, Zhu H, Liu W, Xu F, Zhou F, Zhang Y. 2018; The therapeutic effect of ICAM-1-overexpressing mesenchymal stem cells on acute graft-versus-host disease. Cell Physiol Biochem. 46:2624–2635. DOI: 10.1159/000489689. PMID: 29763906. PMID: d1cf72a8a0a34510a05989fc38b6e288.
Article
26. Sun Z, Gong X, Zhu H, Wang C, Xu X, Cui D, Qian W, Han X. 2014; Inhibition of Wnt/β-catenin signaling promotes engraftment of mesenchymal stem cells to repair lung injury. J Cell Physiol. 229:213–224. DOI: 10.1002/jcp.24436. PMID: 23881674.
Article
27. De Paepe ME, Patel C, Tsai A, Gundavarapu S, Mao Q. 2008; Endoglin (CD105) up-regulation in pulmonary microvascu-lature of ventilated preterm infants. Am J Respir Crit Care Med. 178:180–187. DOI: 10.1164/rccm.200608-1240OC. PMID: 18420967. PMCID: PMC2453512.
Article
28. Horie S, Gonzalez HE, Laffey JG, Masterson CH. 2018; Cell therapy in acute respiratory distress syndrome. J Thorac Dis. 10:5607–5620. DOI: 10.21037/jtd.2018.08.28. PMID: 30416812. PMCID: PMC6196176.
Article
29. Huh JW, Kim WY, Park YY, Lim CM, Koh Y, Kim MJ, Hong SB. 2018; Anti-inflammatory role of mesenchymal stem cells in an acute lung injury mouse model. Acute Crit Care. 33:154–161. DOI: 10.4266/acc.2018.00619. PMID: 31723879. PMCID: PMC6786701. PMID: 283c3f2cef0344f79dcc5d9a2a615d48.
30. Gonzales JN, Lucas R, Verin AD. 2015; The acute respiratory distress syndrome: mechanisms and perspective therapeutic approaches. Austin J Vasc Med. 2:1009. PMID: 26973981. PMCID: PMC4786180.
31. Herrero R, Sanchez G, Lorente JA. 2018; New insights into the mechanisms of pulmonary edema in acute lung injury. Ann Transl Med. 6:32. DOI: 10.21037/atm.2017.12.18. PMID: 29430449. PMCID: PMC5799138.
Article
32. Müller JM, Ziegler-Heitbrock HW, Baeuerle PA. 1993; Nuclear factor kappa B, a mediator of lipopolysaccharide effects. Immunobiology. 187:233–256. DOI: 10.1016/S0171-2985(11)80342-6. PMID: 8330898.
Article
33. Jayasingam SD, Citartan M, Thang TH, Mat Zin AA, Ang KC, Ch'ng ES. 2020; Evaluating the polarization of tumor-asso-ciated macrophages into M1 and M2 phenotypes in human cancer tissue: technicalities and challenges in routine clinical practice. Front Oncol. 9:1512. DOI: 10.3389/fonc.2019.01512. PMID: 32039007. PMCID: PMC6992653. PMID: d4b393b7b9b34fa9a508c0cffaf681e8.
Article
34. Shin SA, Joo BJ, Lee JS, Ryu G, Han M, Kim WY, Park HH, Lee JH, Lee CS. 2020; Phytochemicals as anti-inflammatory agents in animal models of prevalent inflammatory diseases. Molecules. 25:5932. DOI: 10.3390/molecules25245932. PMID: 33333788. PMCID: PMC7765227. PMID: 4cd42ca7898b41dabfe586c2b9f7c9dd.
Article
Full Text Links
  • IJSC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr