DOI QR코드

DOI QR Code

Therapeutic potential of BMSC-conditioned medium in an in vitro model of renal fibrosis using the RPTEC/TERT1 cell line

  • Yunji Kim (Department of Medical Sciences, General Graduate School, Soonchunhyang University) ;
  • Dayeon Kang (Department of Medical Sciences, General Graduate School, Soonchunhyang University) ;
  • Ga-eun Choi (Department of Medical Sciences, General Graduate School, Soonchunhyang University) ;
  • Sang Dae Kim (Department of Medical Sciences, General Graduate School, Soonchunhyang University) ;
  • Sun-ja Yang (Pharmicell Co., Ltd.) ;
  • Hyosang Kim (Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine) ;
  • Dalsan You (Department of Urology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine) ;
  • Choung Soo Kim (Urology Institute, Ewha Womans University Mokdong Hospital) ;
  • Nayoung Suh (Department of Medical Sciences, General Graduate School, Soonchunhyang University)
  • Received : 2023.12.14
  • Accepted : 2024.01.15
  • Published : 2024.02.29

Abstract

We investigated the therapeutic potential of bone marrow-derived mesenchymal stem cell-conditioned medium (BMSC-CM) on immortalized renal proximal tubule epithelial cells (RPTEC/TERT1) in a fibrotic environment. To replicate the increased stiffness characteristic of kidneys in chronic kidney disease, we utilized polyacrylamide gel platforms. A stiff matrix was shown to increase α-smooth muscle actin (α-SMA) levels, indicating fibrogenic activation in RPTEC/TERT1 cells. Interestingly, treatment with BMSC-CM resulted in significant reductions in the levels of fibrotic markers (α-SMA and vimentin) and increases in the levels of the epithelial marker E-cadherin and aquaporin 7, particularly under stiff conditions. Furthermore, BMSC-CM modified microRNA (miRNA) expression and reduced oxidative stress levels in these cells. Our findings suggest that BMSC-CM can modulate cellular morphology, miRNA expression, and oxidative stress in RPTEC/TERT1 cells, highlighting its therapeutic potential in fibrotic kidney disease.

Keywords

Acknowledgement

This work was supported by the 2022 Sabbatical Year of Soonchunhyang University, the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2021R1I1A3048341), and the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI18C0283).

References

  1. Romagnani P, Remuzzi G, Glassock R et al (2017) Chronic kidney disease. Nat Rev Dis Primers 3, 17088
  2. Ju SH and Yi HS (2023) Clinical features and molecular mechanism of muscle wasting in end stage renal disease. BMB Rep 56, 426-438 https://doi.org/10.5483/BMBRep.2023-0097
  3. Kim KP, Williams CE and Lemmon CA (2022) Cell-matrix interactions in renal fibrosis. Kidney Dial 2, 607-624 https://doi.org/10.3390/kidneydial2040055
  4. Wang C, Li SW, Zhong X, Liu BC and Lv LL (2023) An update on renal fibrosis: from mechanisms to therapeutic strategies with a focus on extracellular vesicles. Kidney Res Clin Pract 42, 174-187 https://doi.org/10.23876/j.krcp.22.159
  5. Nogueira A, Pires MJ and Oliveira PA (2017) Pathophysiological mechanisms of renal fibrosis: a review of animal models and therapeutic strategies. In Vivo 31, 1-22 https://doi.org/10.21873/invivo.11019
  6. Schnaper HW (2017) The tubulointerstitial pathophysiology of progressive kidney disease. Adv Chronic Kidney Dis 24, 107-116 https://doi.org/10.1053/j.ackd.2016.11.011
  7. Zhang JQ, Li YY, Zhang XY et al (2023) Cellular senescence of renal tubular epithelial cells in renal fibrosis. Front Endocrinol 14, 1085605
  8. Huang Y, Wu Q and Tam PKH (2022) Immunomodulatory mechanisms of mesenchymal stem cells and their potential clinical applications. Int J Mol Sci 23, 10023
  9. Wang Y, Chen X, Cao W and Shi Y (2014) Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol 15, 1009-1016 https://doi.org/10.1038/ni.3002
  10. Zhuang Q, Ma RY, Yin YS, Lan TH, Yu M and Ming YZ (2019) Mesenchymal stem cells in renal fibrosis: the flame of cytotherapy. Stem Cells International 2019, 8387350
  11. Liu B, Ding F, Hu D et al (2018) Human umbilical cord mesenchymal stem cell conditioned medium attenuates renal fibrosis by reducing inflammation and epithelial-tomesenchymal transition via the TLR4/NF-kappaB signaling pathway in vivo and in vitro. Stem Cell Res Ther 9, 7
  12. Semedo P, Correa-Costa M, Antonio Cenedeze M et al (2009) Mesenchymal stem cells attenuate renal fibrosis through immune modulation and remodeling properties in a rat remnant kidney model. Stem Cells 27, 3063-3073 https://doi.org/10.1002/stem.214
  13. Qin L, Liu N, Bao CLM et al (2023) Mesenchymal stem cells in fibrotic diseases-the two sides of the same coin. Acta Pharmacol Sin 44, 268-287 https://doi.org/10.1038/s41401-022-00952-0
  14. Wu HJ, Yiu WH, Li RX et al (2014) Mesenchymal stem cells modulate albumin-induced renal tubular inflammation and fibrosis. PLoS One 9, e90883
  15. Zhou Y, Xu H, Xu W et al (2013) Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res Ther 4, 34
  16. Zhang J, Duan CR, Duan XX, Hu Y, Liu JQ and Chen WJ (2022) Quantitative evaluation of real-time shear-wave elastography under deep learning in children with chronic kidney disease. Sci Program 2022, 6051695
  17. Guimaraes CF, Gasperini L, Marques AP and Reis RL (2020) The stiffness of living tissues and its implications for tissue engineering. Nat Rev Mater 5, 351-370 https://doi.org/10.1038/s41578-019-0169-1
  18. Samir AE, Allegretti AS, Zhu QL et al (2015) Shear wave elastography in chronic kidney disease: a pilot experience in native kidneys. BMC Nephrology 16, 119
  19. Ishihara S, Kurosawa H and Haga H (2023) Stiffnessmodulation of collagen gels by genipin-crosslinking for cell culture. Gels 9, 148
  20. Ishihara S, Yasuda M, Harada I, Mizutani T, Kawabata K and Haga H (2013) Substrate stiffness regulates temporary NF-κB activation via actomyosin contractions. Exp Cell Res 319, 2916-2927 https://doi.org/10.1016/j.yexcr.2013.09.018
  21. Desai SS, Tung JC, Zhou VX et al (2016) Physiological ranges of matrix rigidity modulate primary mouse hepatocyte function in part through hepatocyte nuclear factor 4 alpha. Hepatology 64, 261-275 https://doi.org/10.1002/hep.28450
  22. Goffin JM, Pittet P, Csucs G, Lussi JW, Meister JJ and Hinz B (2006) Focal adhesion size controls tension-dependent recruitment of α-smooth muscle actin to stress fibers. J Cell Biol 172, 259-268 https://doi.org/10.1083/jcb.200506179
  23. Sacchi M, Bansal R and Rouwkema J (2020) Bioengineered 3D models to recapitulate tissue fibrosis. Trends Biotechnol 38, 623-636 https://doi.org/10.1016/j.tibtech.2019.12.010
  24. Davidson MD, Burdick JA and Wells RG (2020) Engineered biomaterial platforms to study fibrosis. Adv Healthc Mater 9, 1901682
  25. Secker PF, Schlichenmaier N, Beilmann M, Deschl U and Dietrich DR (2019) Functional transepithelial transport measurements to detect nephrotoxicity in vitro using the RPTEC/TERT1 cell line. Arch Toxicol 93, 1965-1978 https://doi.org/10.1007/s00204-019-02469-8
  26. Wang XJ, Wilkinson R, Kildey K et al (2021) Molecular and functional profiling of apical versus basolateral small extracellular vesicles derived from primary human proximal tubular epithelial cells under inflammatory conditions. J Extracell Vesicles 10, e12064
  27. Kim DA, Lee MR, Oh HJ, Kim M and Kong KH (2023) Effects of long-term tubular HIF-2alpha overexpression on progressive renal fibrosis in a chronic kidney disease model. BMB Rep 56, 196-201 https://doi.org/10.5483/BMBRep.2022-0145
  28. Fintha A, Gasparics A, Rosivall L and Sebe A (2019) Therapeutic targeting of fibrotic epithelial-mesenchymal transition-an outstanding challenge. Front Pharmacol 10, 388
  29. Yan H, Xu JX, Xu ZF, Yang B, Luo PH and He QJ (2021) Defining therapeutic targets for renal fibrosis: exploiting the biology of pathogenesis. Biomed Pharmacother 143, 112115
  30. Gluba-Sagr A, Franczyk B, Rysz-Gorzynska M, Lawinski J and Rysz J (2023) The role of miRNA in renal fibrosis leading to chronic kidney disease. Biomedicines 11, 2358
  31. Ichii O and Horino T (2018) MicroRNAs associated with the development of kidney diseases in humans and animals. J Toxicol Pathol 31, 23-34 https://doi.org/10.1293/tox.2017-0051
  32. Zhao H, Ma SX, Shang YQ, Zhang HQ and Su W (2019) microRNAs in chronic kidney disease. Clinica Chimica Acta 491, 59-65 https://doi.org/10.1016/j.cca.2019.01.008
  33. Glowacki F, Savary G, Gnemmi V et al (2013) Increased circulating miR-21 levels are associated with kidney fibrosis. PLoS One 8, e58014
  34. McClelland AD, Herman-Edelstein M, Komers R et al (2015) miR-21 promotes renal fibrosis in diabetic nephropathy by targeting PTEN and SMAD7. Clin Sci (Lond) 129, 1237-1249 https://doi.org/10.1042/CS20150427
  35. Bai L, Lin Y, Xie J, Zhang Y, Wang H and Zheng D (2021) MiR-27b-3p inhibits the progression of renal fibrosis via suppressing STAT1. Hum Cell 34, 383-393 https://doi.org/10.1007/s13577-020-00474-z
  36. Gregory PA, Bert AG, Paterson EL et al (2008) The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 10, 593-601  https://doi.org/10.1038/ncb1722