Research Progress of Each Cell Signaling Pathway in Renal Interstitial Fibrosis and Anti-Fibrotic Intervention Countermeasures

  • Yue Zhou First Affiliated Hospital of Dalian Medical University
  • Hongjie Qian Integrated TCM and Western Medicine Collage of Dalian Medical University
  • Yangjianing Zhao Integrated TCM and Western Medicine Collage of Dalian Medical University
  • Lu Liu Integrated TCM and Western Medicine Collage of Dalian Medical University
  • Qing Guan Integrated TCM and Western Medicine Collage of Dalian Medical University
  • Hongyan Zhou Integrated TCM and Western Medicine Collage of Dalian Medical University
  • Jingchun Pan Integrated TCM and Western Medicine Collage of Dalian Medical University
  • Yu Xue Integrated TCM and Western Medicine Collage of Dalian Medical University&Dalian Hospital of Traditional Chinese Medicine
  • Chao Yan Institute of traditional Chinese medicine
  • Changchuan Bai Dalian Hospital of Traditional Chinese Medicine&Institute of traditional Chinese medicine
  • Dapeng Wang Integrated TCM and Western Medicine Collage of Dalian Medical University & Nephrology Department of First Affiliated Hospital of Dalian Medical University
Keywords: Interstitial Kidney, Fibrosis, Fibroblasts, Pericytes, Anti-Fibrosis, Mirna

Abstract

Interstitial fibrosis is a common pathological feature of various progressive renal diseases, andthis result is mainly caused with the activation of renal interstitial innate cells (fibroblasts, pericytes, immune cells, mesenchymal stem cells, etc.) and the massive expression and deposition of extracellular matrix(ECM). According to statistics,chronic kidney disease and interstitial renal fibrosis affect half of the world's adults over the age of 70 and 10% of the population.Although there are currently no drugs or other means to halt this process, as more and more key players affecting fibrosis are identified, this provides new research directions for anti-fibrotic therapy. In this review, we highlight the relationship between renal interstitial lamina propria and the progression of interstitial fibrosis and describe new advances in anti-fibrotic strategies.Finally,we hope to provide new ideas for the treatment of interstitial renal fibrosis.

References

[1] Zeisberg M, Kalluri R. Physiology of the Renal Interstitium [J]. Clin J Am Soc Nephrol, 2015, 10(10): 1831-40.

[2] Lemley KV, Kriz W. Anatomy of the renal interstitium [J]. Kidney Int, 1991, 39(3): 370-81.

[3] Zhu F, Bai X, Chen XB, lymphocytes in renal interstitial fibrosis [J]. J Cell Commun Signal, 2017, 11(3): 213-8.

[4] Takahashi-Iwanaga H. The three-dimensional cytoarchitecture of the interstitial tissue in the rat kidney [J]. Cell and tissue research, 1991, 264(2): 269-81.

[5] Humphreys BD. Mechanisms of Renal Fibrosis [J]. Annu Rev Physiol, 2018, 80: 309-26.

[6] Kuppe C, Ibrahim M M, Kranz J, et al. Decoding myofibroblast origins in human kidney fibrosis [J]. Nature, 2021, 589(7841): 281-6.

[7] Zeisberg M, Neilson EG. Mechanisms of tubulointerstitial fibrosis [J]. J Am Soc Nephrol, 2010, 21(11): 1819-34.

[8] Pardali E, Sanchez-Duffhues G, Gomez-Puerto MC, et al. TGF-β-Induced Endothelial-Mesenchymal Transition in Fibrotic Diseases [J]. Int J Mol Sci, 2017, 18(10).

[9] Derynck, R, Zhang, YE. Smad-dependent and Smad-independent pathways in TGF-β family signalling [J]. Nature, 2003, 425(6958): 577-84.

[10] Hu HH, Chen DQ, Wang YN, et al. New insights into TGF-β/Smad signaling in tissue fibrosis [J]. Chem Biol Interact, 2018, 292: 76-83.

[11] Alexis D, Antoine G, Franfoise G, et al. Transforming Growth Factor-ill Induces u-Smooth Muscle Actin Expression in Granulation Tissue Myofibroblasts and in Quiescent and Growing Cultured Fibroblasts [J]. The Journal of Cell Biology, 1993, 122(1): 103-11.

[12] Chen YT, Jhao PY, Hung CT, et al. Endoplasmic reticulum protein TXNDC5 promotes renal fibrosis by enforcing TGF-beta signaling in kidney fibroblasts [J]. J Clin Invest, 2021, 131(5).

[13] Wang Y, Lu M, Xiong L, et al. Drp1-mediated mitochondrial fission promotes renal fibroblast activation and fibrogenesis [J]. Cell Death Dis, 2020, 11(1): 29.

[14] Samain R, Sanz-Moreno V. Cancer-associated fibroblasts: activin A adds another string to their bow [J]. EMBO Mol Med, 2020, 12(4): e12102.

[15] Lipphardt M, Song J W, Matsumoto K, et al. The third path of tubulointerstitial fibrosis: aberrant endothelial secretome [J]. Kidney Int, 2017, 92(3): 558-68.

[16] Zhou D, Fu H, Zhang L, et al. Tubule-Derived Wnts Are Required for Fibroblast Activation and Kidney Fibrosis [J]. J Am Soc Nephrol, 2017, 28(8): 2322-36.

[17] Feng Y, Ren J, Gui Y, et al. Wnt/β-Catenin-Promoted Macrophage Alternative Activation Contributes to Kidney Fibrosis [J]. J Am Soc Nephrol, 2018, 29(1): 182-93.

[18] Burgy O, Konigshoff M. The WNT signaling pathways in wound healing and fibrosis [J]. Matrix Biol, 2018, 68-69: 67-80.

[19] Beyer C, Schramm A, Akhmetshina A, et al. β-catenin is a central mediator of pro-fibrotic Wnt signaling in systemic sclerosis [J]. Ann Rheum Dis, 2012, 71(5): 761-7.

[20] Lam A P, Flozak A S, Russell S, et al. Nuclear β-catenin is increased in systemic sclerosis pulmonary fibrosis and promotes lung fibroblast migration and proliferation [J]. Am J Respir Cell Mol Biol, 2011, 45(5): 915-22.

[21] Kramann R, Humphreys B D. Kidney pericytes: roles in regeneration and fibrosis [J]. Semin Nephrol, 2014, 34(4): 374-83.

[22] Khairoun M, Van Der Pol P, De Vries DK, et al. Renal ischemia-reperfusion induces a dysbalance of angiopoietins, accompanied by proliferation of pericytes and fibrosis [J]. Am J Physiol Renal Physiol, 2013, 305(6): F901-10.

[23] Shaw I, Rider S, Mullins J, et al. Pericytes in the renal vasculature: roles in health and disease [J]. Nat Rev Nephrol, 2018, 14(8): 521-34.

[24] Koszegi S, Molnar A, Lenart L, et al. RAAS inhibitors directly reduce diabetes-induced renal fibrosis via growth factor inhibition [J]. The Journal of physiology, 2019, 597(1): 193-209.

[25] Lin YC, Chang YH, Yang SY, et al. Update of pathophysiology and management of diabetic kidney disease [J]. J Formos Med Assoc, 2018, 117(8): 662-75.

[26] Patel S, Rauf A, Khan H, et al. Renin-angiotensin-aldosterone (RAAS): The ubiquitous system for homeostasis and pathologies [J]. Biomed Pharmacother, 2017, 94: 317-25.

[27] Nomura H, Kuruppu S, Rajapakse NW. Stimulation of Angiotensin Converting Enzyme 2: A Novel Treatment Strategy for Diabetic Nephropathy [J]. Front Physiol, 2021, 12: 813012.

[28] Chou YH, Pan SY, Shao YH, et al. Methylation in pericytes after acute injury promotes chronic kidney disease [J]. J Clin Invest, 2020, 130(9): 4845-57.

[29] Meng XM, Nikolic-Paterson DJ, Lan HY. Inflammatory processes in renal fibrosis [J]. Nat Rev Nephrol, 2014, 10(9): 493-503.

[30] Chi HH, Hua KF, Lin YC, et al. IL-36 Signaling Facilitates Activation of the NLRP3 Inflammasome and IL-23/IL-17 Axis in Renal Inflammation and Fibrosis [J]. J Am Soc Nephrol, 2017, 28(7): 2022-37.

[31] Zheng N, Xie K, Ye H, et al. TLR7 in B cells promotes renal inflammation and Gd-IgA1 synthesis in IgA nephropathy [J]. JCI Insight, 2020, 5(14).

[32] Lu X, Rudemiller NP, Privratsky JR, et al. Classical Dendritic Cells Mediate Hypertension by Promoting Renal Oxidative Stress and Fluid Retention [J]. Hypertension, 2020, 75(1): 131-8.

[33] Wang YY, Jiang H, Pan J, et al. Macrophage-to-Myofibroblast Transition Contributes to Interstitial Fibrosis in Chronic Renal Allograft Injury [J]. J Am Soc Nephrol, 2017, 28(7): 2053-67.

[34] Tang PM, Nikolic-Paterson DJ, Lan HY. Macrophages: versatile players in renal inflammation and fibrosis [J]. Nat Rev Nephrol, 2019, 15(3): 144-58.

[35] Kramann R, Schneider R K, Dirocco D P, et al. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis [J]. Cell Stem Cell, 2015, 16(1): 51-66.

[36] Rong X, Liu J, Yao X, et al. Human bone marrow mesenchymal stem cells-derived exosomes alleviate liver fibrosis through the Wnt/beta-catenin pathway [J]. Stem Cell Res Ther, 2019, 10(1): 98.

[37] Shojaati G, Khandaker I, Funderburgh ML, et al. Mesenchymal Stem Cells Reduce Corneal Fibrosis and Inflammation via Extracellular Vesicle-Mediated Delivery of miRNA [J]. Stem Cells Transl Med, 2019, 8(11): 1192-201.

[38] Watanabe Y, Tsuchiya A, Seino S, et al. Mesenchymal Stem Cells and Induced Bone Marrow-Derived Macrophages Synergistically Improve Liver Fibrosis in Mice [J]. Stem Cells Transl Med, 2019, 8(3): 271-84.

[39] Zanoni M, Cortesi M, Zamagni A, et al. The Role of Mesenchymal Stem Cells in Radiation-Induced Lung Fibrosis [J]. Int J Mol Sci, 2019, 20(16).

[40] Ishiuchi N, Nakashima A, Doi S, et al. Hypoxia-preconditioned mesenchymal stem cells prevent renal fibrosis and inflammation in ischemia-reperfusion rats [J]. Stem Cell Res Ther, 2020, 11(1): 130.

[41] Nastase MV, Zeng-Brouwers J, Wygrecka M, et al. Targeting renal fibrosis: Mechanisms and drug delivery systems [J]. Adv Drug Deliv Rev, 2018, 129: 295-307.

[42] Bai Y, Wang W, Yin P, et al. Ruxolitinib Alleviates Renal Interstitial Fibrosis in UUO Mice [J]. Int J Biol Sci, 2020, 16(2): 194-203.

[43] Borza C M, Bolas G, Bock F, et al. DDR1 contributes to kidney inflammation and fibrosis by promoting the phosphorylation of BCR and STAT3 [J]. JCI Insight, 2022, 7(3).

[44] Li R, Guo Y, Zhang Y, et al. Salidroside Ameliorates Renal Interstitial Fibrosis by Inhibiting the TLR4/NF-kappaB and MAPK Signaling Pathways [J]. Int J Mol Sci, 2019, 20(5).

[45] Chen Y, Mu L, Xing L, et al. Rhein alleviates renal interstitial fibrosis by inhibiting tubular cell apoptosis in rats [J]. Biol Res, 2019, 52(1): 50.

[46] Yan H, Xu J, Xu Z, et al. Defining therapeutic targets for renal fibrosis: Exploiting the biology of pathogenesis [J]. Biomed Pharmacother, 2021, 143: 112115.

[47] Ajay A K, Zhao L, Vig S, et al. Deletion of STAT3 from Foxd1 cell population protects mice from kidney fibrosis by inhibiting pericytes trans-differentiation and migration [J]. Cell Rep, 2022, 38(10): 110473.

[48] Aggarwal D, Singh G. Effects of single and dual RAAS blockade therapy on progressive kidney disease transition to CKD in rats [J]. Naunyn Schmiedebergs Arch Pharmacol, 2020, 393(4): 615-27.

[49] Cha J J, Mandal C, Ghee J Y, et al. Inhibition of Renal Stellate Cell Activation Reduces Renal Fibrosis [J]. Biomedicines, 2020, 8(10).

[50] Martínez-Arias L, Panizo S, Alonso-Montes C, et al. Effects of calcitriol and paricalcitol on renal fibrosis in CKD [J]. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association, 2021, 36(5): 793-803.

[51] Li C, Xie N, Li Y, et al. N-acetylcysteine ameliorates cisplatin-induced renal senescence and renal interstitial fibrosis through sirtuin1 activation and p53 deacetylation [J]. Free Radic Biol Med, 2019, 130: 512-27.

[52] Yang H, Hua C, Yang X, et al. Pterostilbene prevents LPS-induced early pulmonary fibrosis by suppressing oxidative stress, inflammation and apoptosis in vivo [J]. Food Funct, 2020, 11(5): 4471-84.

[53] Wu KK. Control of Tissue Fibrosis by 5-Methoxytryptophan, an Innate Anti-Inflammatory Metabolite [J]. Front Pharmacol, 2021, 12: 759199.

[54] Conserva F, Barozzino M, Pesce F, et al. Urinary miRNA-27b-3p and miRNA-1228-3p correlate with the progression of Kidney Fibrosis in Diabetic Nephropathy [J]. Sci Rep, 2019, 9(1): 11357.

[55] Zhao Z, Lin CY, Cheng K. siRNA- and miRNA-based therapeutics for liver fibrosis [J]. Transl Res, 2019, 214: 17-29.

[56] Nosalski R, Siedlinski M, Denby L, et al. T-Cell-Derived miRNA-214 Mediates Perivascular Fibrosis in Hypertension [J]. Circ Res, 2020, 126(8): 988-1003.

[57] Pang XF, Lin X, Du JJ, et al. LTBP2 knockdown by siRNA reverses myocardial oxidative stress injury, fibrosis and remodelling during dilated cardiomyopathy [J]. Acta Physiol (Oxf), 2020, 228(3): e13377.

[58] Pozzi A, Zent R. Integrins in kidney disease [J]. J Am Soc Nephrol, 2013, 24(7): 1034-9.

[59] Henderson NC, Sheppard D. Integrin-mediated regulation of TGFbeta in fibrosis [J]. Biochim Biophys Acta, 2013, 1832(7): 891-6.

[60] Smith SW, Croft AP, Morris HL, et al. Genetic Deletion of the Stromal Cell Marker CD248 (Endosialin) Protects against the Development of Renal Fibrosis [J]. Nephron, 2015, 131(4): 265-77.

[61] Macfadyen J R, Haworth O, Roberston D, et al. Endosialin (TEM1, CD248) is a marker of stromal fibroblasts and is not selectively expressed on tumour endothelium [J]. FEBS Lett, 2005, 579(12): 2569-75.

[62] Xu C, Liu S, Yang F, et al. Antibody-drug conjugates targeting CD248(+) myofibroblasts effectively alleviate renal fibrosis in mice [J]. FASEB J, 2022, 36(2): e22102.

[63] Jansen J, Reimer KC, Nagai JS, et al. SARS-CoV-2 infects the human kidney and drives fibrosis in kidney organoids [J]. Cell Stem Cell, 2022, 29(2): 217-31.e8.
Published
2022-11-01
Section
Original Research Article