DOI QR코드

DOI QR Code

Mesenchymal Stem Cells Ameliorate Fibrosis by Enhancing Autophagy via Inhibiting Galectin-3/Akt/mTOR Pathway and by Alleviating the EMT via Inhibiting Galectin-3/Akt/GSK3β/Snail Pathway in NRK-52E Fibrosis

  • Yu Zhao (Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University) ;
  • Chuan Guo (Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University) ;
  • Lianlin Zeng (Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University) ;
  • Jialing Li (Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University) ;
  • Xia Liu (Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University) ;
  • Yiwei Wang (Department of Chemistry, School of Basic Medical Sciences, Southwest Medical University) ;
  • Kun Zhao (Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University) ;
  • Bo Chen (Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University)
  • Received : 2022.01.17
  • Accepted : 2022.02.24
  • Published : 2023.02.28

Abstract

Background and Objectives: Epithelial-Mesenchymal transition (EMT) is one of the origins of myofibroblasts in renal interstitial fibrosis. Mesenchymal stem cells (MSCs) alleviating EMT has been proved, but the concrete mechanism is unclear. To explore the mechanism, serum-free MSCs conditioned medium (SF-MSCs-CM) was used to treat rat renal tubular epithelial cells (NRK-52E) fibrosis induced by transforming growth factor-β1 (TGF-β1) which ameliorated EMT. Methods and Results: Galectin-3 knockdown (Gal-3 KD) and overexpression (Gal-3 OE) lentiviral vectors were established and transfected into NRK-52E. NRK-52E fibrosis model was induced by TGF-β1 and treated with the SF-MSCs-CM for 24 h after modelling. Fibrosis and autophagy related indexes were detected by western blot and immunocytochemistry. In model group, the expressions of α-smooth muscle actin (α-SMA), fibronectin (FN), Galectin-3, Snail, Kim-1, and the ratios of P-Akt/Akt, P-GSK3β/GSK3β, P-PI3K/PI3K, P-mTOR/mTOR, TIMP1/MMP9, and LC3B-II/I were obviously increased, and E-Cadherin (E-cad) and P62 decreased significantly compared with control group. SF-MSCs-CM showed an opposite trend after treatment compared with model group. Whether in Gal-3 KD or Gal-3 OE NRK-52E cells, SF-MSCs-CM also showed similar trends. However, the effects of anti-fibrosis and enhanced autophagy in Gal-3 KD cells were more obvious than those in Gal-3 OE cells. Conclusions: SF-MSCs-CM probably alleviated the EMT via inhibiting Galectin-3/Akt/GSK3β/Snail pathway. Meanwhile, Gal-3 KD possibly enhanced autophagy via inhibiting Galectin-3/Akt/mTOR pathway, which synergistically ameliorated renal fibrosis. Targeting galectin-3 may be a potential target for the treatment of renal fibrosis.

Keywords

Acknowledgement

We thank the Beijing Syngentech Co., LTD, for the support for lentivirus transfection technology. This work was supported by the Science and Technology Department of Sichuan Province (No.2018JY0490), A Project Supported by Scientific Research Fund of Sichuan Provincial Education Department (No. 18ZA0524), Research project of Sichuan Traditional Chinese Medicine Administration (2021MS553), the Health and Family Planning Commission of Sichuan province (No. 16PJ540), Luzhou Science and Technology Bureau (No.2016-S-65(1/9), 2016LZXNYD-J18), and National Innovation and Entrepreneurship Program for College Students (S202110632020; S202110632031).

References

  1. Yan H, Xu J, Xu Z, Yang B, Luo P, He Q. Defining therapeutic targets for renal fibrosis: exploiting the biology of pathogenesis. Biomed Pharmacother 2021;143:112115
  2. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol 2016;12:325-338 https://doi.org/10.1038/nrneph.2016.48
  3. Mack M, Yanagita M. Origin of myofibroblasts and cellular events triggering fibrosis. Kidney Int 2015;87:297-307 https://doi.org/10.1038/ki.2014.287
  4. Liu F, Zhuang S. New therapies for the treatment of renal fibrosis. Adv Exp Med Biol 2019;1165:625-659 https://doi.org/10.1007/978-981-13-8871-2_31
  5. Zhuang Q, Ma R, Yin Y, Lan T, Yu M, Ming Y. Mesenchymal stem cells in renal fibrosis: the flame of cytotherapy. Stem Cells Int 2019;2019:8387350
  6. Prakoura N, Hadchouel J, Chatziantoniou C. Novel targets for therapy of renal fibrosis. J Histochem Cytochem 2019; 67:701-715 https://doi.org/10.1369/0022155419849386
  7. Dikic I, Elazar Z. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol 2018;19:349-364 https://doi.org/10.1038/s41580-018-0003-4
  8. Tang C, Livingston MJ, Liu Z, Dong Z. Autophagy in kidney homeostasis and disease. Nat Rev Nephrol 2020;16:489-508 https://doi.org/10.1038/s41581-020-0309-2
  9. Kim YA, Kim HJ, Gwon MG, Gu H, An HJ, Bae S, Leem J, Jung HJ, Park KK. Inhibitory effects of STAT3 transcription factor by synthetic decoy ODNs on autophagy in renal fibrosis. Biomedicines 2021;9:331
  10. Xue X, Ren J, Sun X, Gui Y, Feng Y, Shu B, Wei W, Lu Q, Liang Y, He W, Yang J, Dai C. Protein kinase Cα drives fibroblast activation and kidney fibrosis by stimulating autophagic flux. J Biol Chem 2018;293:11119-11130 https://doi.org/10.1074/jbc.RA118.002191
  11. Gong W, Luo C, Peng F, Xiao J, Zeng Y, Yin B, Chen X, Li S, He X, Liu Y, Cao H, Xu J, Long H. Brahma-related gene-1 promotes tubular senescence and renal fibrosis through Wnt/β-catenin/autophagy axis. Clin Sci (Lond) 2021;135:1873-1895 https://doi.org/10.1042/CS20210447
  12. Wang YJ, Chen YY, Hsiao CM, Pan MH, Wang BJ, Chen YC, Ho CT, Huang KC, Chen RJ. Induction of autophagy by pterostilbene contributes to the prevention of renal fibrosis via attenuating NLRP3 inflammasome activation and epithelial-mesenchymal transition. Front Cell Dev Biol 2020;8:436
  13. Zhao XC, Livingston MJ, Liang XL, Dong Z. Cell apoptosis and autophagy in renal fibrosis. Adv Exp Med Biol 2019;1165:557-584 https://doi.org/10.1007/978-981-13-8871-2_28
  14. Gubert F, da Silva JS, Vasques JF, de Jesus Goncalves RG, Martins RS, de Sa MPL, Mendez-Otero R, Zapata-Sudo G. Mesenchymal stem cells therapies on fibrotic heart diseases. Int J Mol Sci 2021;22:7447
  15. Ntolios P, Steiropoulos P, Karpathiou G, Anevlavis S, Karampitsakos T, Bouros E, Froudarakis ME, Bouros D, Tzouvelekis A. Cell therapy for idiopathic pulmonary fibrosis: rationale and progress to date. BioDrugs 2020;34:543-556 https://doi.org/10.1007/s40259-020-00437-8
  16. Tsuchiya A, Takeuchi S, Watanabe T, Yoshida T, Nojiri S, Ogawa M, Terai S. Mesenchymal stem cell therapies for liver cirrhosis: MSCs as "conducting cells" for improvement of liver fibrosis and regeneration. Inflamm Regen 2019;39:18
  17. Yun CW, Lee SH. Potential and therapeutic efficacy of cell-based therapy using mesenchymal stem cells for acute/chronic kidney disease. Int J Mol Sci 2019;20:1619
  18. Makhlough A, Shekarchian S, Moghadasali R, Einollahi B, Dastgheib M, Janbabaee G, Hosseini SE, Falah N, Abbasi F, Baharvand H, Aghdami N. Bone marrow-mesenchymal stromal cell infusion in patients with chronic kidney disease: a safety study with 18 months of follow-up. Cytotherapy 2018;20:660-669 https://doi.org/10.1016/j.jcyt.2018.02.368
  19. Quimby JM, Webb TL, Habenicht LM, Dow SW. Safety and efficacy of intravenous infusion of allogeneic cryopreserved mesenchymal stem cells for treatment of chronic kidney disease in cats: results of three sequential pilot studies. Stem Cell Res Ther 2013;4:48
  20. Perico N, Casiraghi F, Remuzzi G. Clinical translation of mesenchymal stromal cell therapies in nephrology. J Am Soc Nephrol 2018;29:362-375 https://doi.org/10.1681/ASN.2017070781
  21. Li H, Rong P, Ma X, Nie W, Chen Y, Zhang J, Dong Q, Yang M, Wang W. Mouse umbilical cord mesenchymal stem cell paracrine alleviates renal fibrosis in diabetic nephropathy by reducing myofibroblast transdifferentiation and cell proliferation and upregulating MMPs in mesangial cells. J Diabetes Res 2020;2020:3847171
  22. Liu B, Ding FX, Liu Y, Xiong G, Lin T, He DW, Zhang YY, Zhang DY, Wei GH. Human umbilical cord-derived mesenchymal stem cells conditioned medium attenuate interstitial fibrosis and stimulate the repair of tubular epithelial cells in an irreversible model of unilateral ureteral obstruction. Nephrology (Carlton) 2018;23:728-736 https://doi.org/10.1111/nep.13099
  23. Chen L, Wang Y, Li S, Zuo B, Zhang X, Wang F, Sun D. Exosomes derived from GDNF-modified human adipose mesenchymal stem cells ameliorate peritubular capillary loss in tubulointerstitial fibrosis by activating the SIRT1/ eNOS signaling pathway. Theranostics 2020;10:9425-9442 https://doi.org/10.7150/thno.43315
  24. Bernard M, Yang B, Migneault F, Turgeon J, Dieude M, Olivier MA, Cardin GB, El-Diwany M, Underwood K, Rodier F, Hebert MJ. Autophagy drives fibroblast senescence through MTORC2 regulation. Autophagy 2020;16:2004-2016 https://doi.org/10.1080/15548627.2020.1713640
  25. Shu S, Wang H, Zhu J, Liu Z, Yang D, Wu W, Cai J, Chen A, Tang C, Dong Z. Reciprocal regulation between ER stress and autophagy in renal tubular fibrosis and apoptosis. Cell Death Dis 2021;12:1016
  26. Li S, Lin Q, Shao X, Zhu X, Wu J, Wu B, Zhang M, Zhou W, Zhou Y, Jin H, Zhang Z, Qi C, Shen J, Mou S, Gu L, Ni Z. Drp1-regulated PARK2-dependent mitophagy protects against renal fibrosis in unilateral ureteral obstruction. Free Radic Biol Med 2020;152:632-649 https://doi.org/10.1016/j.freeradbiomed.2019.12.005
  27. Kimura T, Takahashi A, Takabatake Y, Namba T, Yamamoto T, Kaimori JY, Matsui I, Kitamura H, Niimura F, Matsusaka T, Soga T, Rakugi H, Isaka Y. Autophagy protects kidney proximal tubule epithelial cells from mitochondrial metabolic stress. Autophagy 2013;9:1876-1886 https://doi.org/10.4161/auto.25418
  28. Chen K, Yu B, Liao J. LncRNA SOX2OT alleviates mesangial cell proliferation and fibrosis in diabetic nephropathy via Akt/mTOR-mediated autophagy. Mol Med 2021;27:71
  29. Cong LH, Li T, Wang H, Wu YN, Wang SP, Zhao YY, Zhang GQ, Duan J. IL-17A-producing T cells exacerbate fine particulate matter-induced lung inflammation and fibrosis by inhibiting PI3K/Akt/mTOR-mediated autophagy. J Cell Mol Med 2020;24:8532-8544 https://doi.org/10.1111/jcmm.15475
  30. Dai J, Sun Y, Chen D, Zhang Y, Yan L, Li X, Wang J. Negative regulation of PI3K/AKT/mTOR axis regulates fibroblast proliferation, apoptosis and autophagy play a vital role in triptolide-induced epidural fibrosis reduction. Eur J Pharmacol 2019;864:172724
  31. Nam SA, Kim WY, Kim JW, Park SH, Kim HL, Lee MS, Komatsu M, Ha H, Lim JH, Park CW, Yang CW, Kim J, Kim YK. Autophagy attenuates tubulointerstital fibrosis through regulating transforming growth factor-β and NLRP3 inflammasome signaling pathway. Cell Death Dis 2019;10:78
  32. Zhao XC, Livingston MJ, Liang XL, Dong Z. Cell apoptosis and autophagy in renal fibrosis. Adv Exp Med Biol 2019;1165:557-584 https://doi.org/10.1007/978-981-13-8871-2_28
  33. Saccon F, Gatto M, Ghirardello A, Iaccarino L, Punzi L, Doria A. Role of galectin-3 in autoimmune and non-autoimmune nephropathies. Autoimmun Rev 2017;16:34-47 https://doi.org/10.1016/j.autrev.2016.09.023
  34. Hara A, Niwa M, Noguchi K, Kanayama T, Niwa A, Matsuo M, Hatano Y, Tomita H. Galectin-3 as a next-generation biomarker for detecting early stage of various diseases. Biomolecules 2020;10:389
  35. Martinez-Martinez E, Ibarrola J, Fernandez-Celis A, Calvier L, Leroy C, Cachofeiro V, Rossignol P, Lopez- Andres N. Galectin-3 pharmacological inhibition attenuates early renal damage in spontaneously hypertensive rats. J Hypertens 2018;36:368-376 https://doi.org/10.1097/HJH.0000000000001545
  36. Oikonomou T, Goulis I, Ntogramatzi F, Athanasiadou Z, Vagdatli E, Akriviadis E, Cholongitas E. Galectin-3 is associated with glomerular filtration rate and outcome in patients with stable decompensated cirrhosis. Dig Liver Dis 2019;51:1692-1697 https://doi.org/10.1016/j.dld.2019.05.030
  37. Shen H, Wang J, Min J, Xi W, Gao Y, Yin L, Yu Y, Liu K, Xiao J, Zhang YF, Wang ZN. Activation of TGF-β1/α- SMA/Col I profibrotic pathway in fibroblasts by galectin-3 contributes to atrial fibrosis in experimental models and patients. Cell Physiol Biochem 2018;47:851-863 https://doi.org/10.1159/000490077
  38. Dong R, Zhang M, Hu Q, Zheng S, Soh A, Zheng Y, Yuan H. Galectin-3 as a novel biomarker for disease diagnosis and a target for therapy (Review). Int J Mol Med 2018;41:599-614
  39. Wang Y, Chen X, Cao W, Shi Y. Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol 2014;15:1009-1016 https://doi.org/10.1038/ni.3002
  40. Yang D, Wang W, Li L, Peng Y, Chen P, Huang H, Guo Y, Xia X, Wang Y, Wang H, Wang WE, Zeng C. The relative contribution of paracine effect versus direct differentiation on adipose-derived stem cell transplantation mediated cardiac repair. PLoS One 2013;8:e59020
  41. d'Angelo M, Cimini A, Castelli V. Insights into the effects of mesenchymal stem cell-derived secretome in Parkinson's disease. Int J Mol Sci 2020;21:5241
  42. Gunawardena TNA, Rahman MT, Abdullah BJJ, Abu Kasim NH. Conditioned media derived from mesenchymal stem cell cultures: the next generation for regenerative medicine. J Tissue Eng Regen Med 2019;13:569-586 https://doi.org/10.1002/term.2806
  43. Kholia S, Herrera Sanchez MB, Cedrino M, Papadimitriou E, Tapparo M, Deregibus MC, Bruno S, Antico F, Brizzi MF, Quesenberry PJ, Camussi G. Mesenchymal stem cell derived extracellular vesicles ameliorate kidney injury in aristolochic acid nephropathy. Front Cell Dev Biol 2020;8:188
  44. Reis LA, Borges FT, Simoes MJ, Borges AA, Sinigaglia- Coimbra R, Schor N. Bone marrow-derived mesenchymal stem cells repaired but did not prevent gentamicin-induced acute kidney injury through paracrine effects in rats. PLoS One 2012;7:e44092
  45. Yu B, Zhang X, Li X. Exosomes derived from mesenchymal stem cells. Int J Mol Sci 2014;15:4142-4157 https://doi.org/10.3390/ijms15034142
  46. Yin S, Zhou S, Ren D, Zhang J, Xin H, He X, Gao H, Hou J, Zeng F, Lu Y, Zhang X, Fan M. Mesenchymal stem cell-derived exosomes attenuate epithelial-mesenchymal transition of HK-2 cells. Tissue Eng Part A 2022 doi: 10.1089/ten.TEA.2021.0190 [Epub ahead of print]
  47. Wang B, Yao K, Huuskes BM, Shen HH, Zhuang J, Godson C, Brennan EP, Wilkinson-Berka JL, Wise AF, Ricardo SD. Mesenchymal stem cells deliver exogenous microRNA-let7c via exosomes to attenuate renal fibrosis. Mol Ther 2016;24:1290-1301 https://doi.org/10.1038/mt.2016.90
  48. Shi Z, Wang Q, Zhang Y, Jiang D. Extracellular vesicles produced by bone marrow mesenchymal stem cells attenuate renal fibrosis, in part by inhibiting the RhoA/ROCK pathway, in a UUO rat model. Stem Cell Res Ther 2020;11:253