Exosomes of Whartons’ jelly mesenchymal stem cell reduce the NOX genes in TGF-β-induced hepatic fibrosis

Document Type : Original Article

Authors

1 Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran

2 Department of Clinical Biochemistry, Faculty of Medicine, Jundishapour University of Medical Sciences, Ahvaz, Iran

Abstract

Objective(s): Activated cells which are called star-shaped cells, are some of the key factors in the development of liver fibrosis. Activation of NADPH oxidase (NOX) is associated with increased HSCs activity and progression of hepatic fibrosis. In this study, the effects of human exosomes derived from WJ-MSCs on NOX1, NOX2, and NOX4 gene expression in TGF-β-induced hepatic fibrosis were investigated.
Materials and Methods: LX2 cell line was treated with 2 ng/ml TGF-β for 24 hr, in order to induce liver fibrosis after starvation. In the next step, the cells were treated with several concentrations of the exosomes derived from WJ-MSCs (10, 20, 30, 40, and 50 μg/ml). Finally, Smad3C phosphorylated protein expression level and NOX1, NOX2, and NOX4 gene expression levels were measured.
Results: The results demonstrated that the level of NOX1, NOX2, and NOX4 mRNA expressions decreased significantly during 24 hrs at concentrations of 40 and 50 μg/ml of WJ-MSCs exosomes in TGF-β-induced-HSCs. The p-Smad3C proteins were significantly decreased (fold change: 1.83, P-value<0.05) after exposure to WJ-MSC-derived exosomes. 
Conclusion: Treatment with exosomes prevents further activation of HSCs by inhibiting the level of Smad3C phosphorylation. The experimental data of our study suggested that in liver fibrosis, the protection of HSCs activation against TGF-β by inhibiting the NOX pathway via human exosomes of WJ-MSCs is extremely important. It needs further research as a treatment method.

Keywords


1. Schuppan D, Kim YO. Evolving therapies for liver fibrosis. J Clin Invest 2013;123:1887-1901.
2. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest  2005;115:209-218.
3. Kawaratani H, Tsujimoto T, Douhara A, Takaya H, Moriya K, Namisaki T, et al. The effect of inflammatory cytokines in alcoholic liver disease. Mediators Inflamm 2013;2013:495156.
4. Pinzani M, Marra F. Cytokine receptors and signaling in hepatic stellate cells. Semin Liver Dis 2001;21:397-416.
5. Koyama Y, Xu J, Liu X, Brenner DA. New developments on the treatment of liver fibrosis. Dig Dis 2016;34:589-596.
6. Chang Y, Li H. Hepatic antifibrotic pharmacotherapy: Are we approaching success? J Clin Transl Hepatol 2020;8:222-229.
7. Mortezaee K. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) and liver fibrosis: A review. Cell Biochem Funct 2018;36:292-302.
8. Cui W, Matsuno K, Iwata K, Ibi M, Matsumoto M, Zhang J, et al. NOX1/nicotinamide adenine dinucleotide phosphate, reduced form (NADPH) oxidase promotes proliferation of stellate cells and aggravates liver fibrosis induced by bile duct ligation. Hepatology 2011; 54: 949-958.
9. Yoshida K, Murata M, Yamaguchi T, Matsuzaki K. TGF-β/Smad signaling during hepatic fibro-carcinogenesis. Int J Oncol 2014; 45: 1363-1371.
10. Liang S, Kisseleva T, Brenner DA. The role of NADPH oxidases (NOXs) in liver fibrosis and the activation of myofibroblasts. Front Physiol 2016; 7: 17.
11. Datto MB, Frederick JP, Pan L, Borton AJ, Zhuang Y, Wang X-F. Targeted disruption of Smad3 reveals an essential role in transforming growth factor β-mediated signal transduction. Mol Cell Biol 1999; 19: 2495-2504.
12. Huang F, Chen Y-G. Regulation of TGF-β receptor activity. Cell Biosci 2012;2:1-10.
13. Parsons CJ, Takashima M, Rippe RA. Molecular mechanisms of hepatic fibrogenesis. J Gastroenterol Hepatol 2007; 22: S79-S84.
14. Gressner A, Weiskirchen R. Modern pathogenetic concepts of liver fibrosis suggest stellate cells and TGF‐β as major players and therapeutic targets. J Cell Mol Med 2006; 10: 76-99.
15. Liu X, Hu H, Yin JQ. Therapeutic strategies against TGF‐β signaling pathway in hepatic fibrosis. Liver Int 2006; 26: 8-22.
16. Balta C, Herman H, Boldura OM, Gasca I, Rosu M, Ardelean A, et al. Chrysin attenuates liver fibrosis and hepatic stellate cell activation through TGF-β/Smad signaling pathway. Chem Biol Interact 2015; 240: 94-101.
17. Zhang Z, Wang F-S. Stem cell therapies for liver failure and cirrhosis. J Hepatol 2013; 59: 183-185.
18. Lou G, Chen Z, Zheng M, Liu Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp Mol Med 2017; 49: e346.
19. Asgarpour K, Shojaei Z, Amiri F, Ai J, Mahjoubin-Tehran M, Ghasemi F, et al. Exosomal microRNAs derived from mesenchymal stem cells: Cell-to-cell messages. Cell Commun Signal 2020; 18: 1-16.
20. Puig-Pijuan T, de Godoy MA, Pinheiro Carvalho LR, Bodart-Santos V, Lindoso RS, Pimentel-Coelho PM, et al. Human Wharton’s jelly mesenchymal stem cells protect neural cells from oxidative stress through paracrine mechanisms. Future Sci OA 2020; 6: FSO627.
21. Yu B, Zhang X, Li X. Exosomes derived from mesenchymal stem cells. Int J Mol Sci 2014; 15: 4142-4157.
22. Nojehdehi S, Hashemi SM, Hesampour A. Isolation and characterization of exosomes separated from stem cells by ultra-centrifuge method. Res Med 2017; 73: 244-250.
23. Gowen A, Shahjin F, Chand S, Odegaard KE, Yelamanchili SV. Mesenchymal stem cell-derived extracellular vesicles: Challenges in clinical applications. Front Cell Dev Biol 2020; 8: 149.
24. Shibata N, Watanabe T, Okitsu T, Sakaguchi M, Takesue M, Kunieda T, et al. Establishment of an immortalized human hepatic stellate cell line to develop antifibrotic therapies. Cell transplant 2003; 12: 499-507.
25. Shi Y-F, Zhang Q, Cheung P-Y, Shi L, Fong C-C, Zhang Y, et al. Effects of rhDecorin on TGF-β1 induced human hepatic stellate cells LX-2 activation. Biochim Biophys Acta 2006; 1760: 1587-1595.
26. Sokolova V, Ludwig A-K, Hornung S, Rotan O, Horn PA, Epple M, et al. Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy. Colloids Surf B Biointerfaces 2011; 87: 146-150.
27. Van der Pol E, Coumans F, Varga Z, Krumrey M, Nieuwland R. Innovation in detection of microparticles and exosomes. J Thromb Haemost 2013; 11: 36-45.
28. Mohammadzadeh G, Afarin R, Bavarsad SS, Aslani F, Zadeh SA, Shakerian E. Comparison of the effects of cholesterol, palmitic acid, and glucose on activation of human hepatic stellate cells to induce liver fibrosis. J Diabetes Metab Disord 2022: 1-8.
29. Terai S, Ishikawa T, Omori K, Aoyama K, Marumoto Y, Urata Y, et al. Improved liver function in patients with liver cirrhosis after autologous bone marrow cell infusion therapy. Stem cells 2006; 24: 2292-2298.
30. Abbaszadeh H, Ghorbani F, Derakhshani M, Movassaghpour AA, Yousefi M, Talebi M, et al. Regenerative potential of Wharton’s jelly‐derived mesenchymal stem cells: A new horizon of stem cell therapy. J Cell Physiol 2020;235:9230-9240.
31. Tsai PC, Fu TW, Chen YMA, Ko TL, Chen TH, Shih YH, et al. The therapeutic potential of human umbilical mesenchymal stem cells from Wharton’s jelly in the treatment of rat liver fibrosis. Liver transpl 2009; 15: 484-495.
32. Parekkadan B, van Poll D, Megeed Z, Kobayashi N, Tilles AW, Berthiaume F, et al. Immunomodulation of activated hepatic stellate cells by mesenchymal stem cells. Biochem biophys Res Commun 2007; 363: 247-252.
33. Qiao H, Zhou Y, Qin X, Cheng J, He Y, Jiang Y. NADPH oxidase signaling pathway mediates mesenchymal stem cell-induced inhibition of hepatic stellate cell activation. Stem cells int 2018; 2018: 1239143.
34. Weiskirchen R. Hepatoprotective and anti-fibrotic agents: It’s time to take the next step. Frontiers Pharmacol 2016;6:303.
35. Paik Y-H, Kim J, Aoyama T, De Minicis S, Bataller R, Brenner DA. Role of NADPH oxidases in liver fibrosis. Anti-oxid Redox Signal 2014; 20: 2854-2572.
36. Lan T, Kisseleva T, Brenner DA. Deficiency of NOX1 or NOX4 prevents liver inflammation and fibrosis in mice through inhibition of hepatic stellate cell activation. PloS One 2015; 10: e0129743.
37. Paik YH, Iwaisako K, Seki E, Inokuchi S, Schnabl B, Österreicher CH, et al. The nicotinamide adenine dinucleotide phosphate oxidase (NOX) homologues NOX1 and NOX2/gp91phox mediate hepatic fibrosis in mice. Hepatology 2011; 53: 1730-1741.
38. Zhang W, Liu R, Chen Y, Wang M, Du J. Crosstalk between Oxidative Stress and Exosomes. Oxid Med Cell Longev 2022; 2022: 3553617.
39. Liu M, Huang Q, Zhu Y, Chen L, Li Y, Gong Z, et al. Harnessing reactive oxygen/nitrogen species and inflammation: Nanodrugs for liver injury. Mater Today Bio 2022; 13:100215.
40. Bataller R, Schwabe RF, Choi YH, Yang L, Paik YH, Lindquist J, et al. NADPH oxidase signal transduces angiotensin II in hepatic stellate cells and is critical in hepatic fibrosis. J Clin Invest 2003; 112:1383-1394.
41. Zhang G, Zou X, Miao S, Chen J, Du T, Zhong L, et al. The anti-oxidative role of micro-vesicles derived from human Wharton-Jelly mesenchymal stromal cells through NOX2/gp91 (phox) suppression in alleviating renal ischemia-reperfusion injury in rats. PloS One 2014; 9: e92129.
42. Lin K-C, Yip H-K, Shao P-L, Wu S-C, Chen K-H, Chen Y-T, et al. Combination of adipose-derived mesenchymal stem cells (ADMSC) and ADMSC-derived exosomes for protecting kidney from acute ischemia–reperfusion injury. Int J cardiol 2016; 216: 173-185.
43. Chen L, Wang Y, Pan Y, Zhang L, Shen C, Qin G, et al. Cardiac progenitor-derived exosomes protect ischemic myocardium from acute ischemia/reperfusion injury. Biochem Biophys Res Commun 2013; 431: 566-571.
44. Sart S, Song L, Li Y. Controlling redox status for stem cell survival, expansion, and differentiation. Oxid Med Cell Longev 2015; 2015: 105135.