
Role of the TGF‑β/Smad signaling pathway in the transition from acute kidney injury to chronic kidney disease (Review)
- Authors:
- Yuanxia Zou
- Jian Dai
- Jianchun Li
- Min Liu
- Run Li
- Guiping Li
- Junyu Lai
- Li Wang
-
Affiliations: Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China, Department of Neurology, The Third People's Hospital of Luzhou, Luzhou, Sichuan 646000, P.R. China, Department of Children's Diagnosis and Treatment Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China, Department of Urology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China - Published online on: July 30, 2025 https://doi.org/10.3892/ijmm.2025.5603
- Article Number: 162
-
Copyright: © Zou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
This article is mentioned in:
Abstract
![]() |
![]() |
![]() |
Sutherland SM, Kaddourah A, Gillespie SE, Soranno DE, Woroniecki RP, Basu RK and Zappitelli M; Assessment of the Worldwide Acute Kidney Injury, Renal Angina and Epidemiology (AWARE) Investigators: Cumulative application of creatinine and urine output staging optimizes the kidney disease: Improving global outcomes definition and identifies increased mortality risk in hospitalized patients with acute kidney injury. Crit Care Med. 49:1912–1922. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chawla LS, Bellomo R, Bihorac A, Goldstein SL, Siew ED, Bagshaw SM, Bittleman D, Cruz D, Endre Z, Fitzgerald RL, et al: Acute kidney disease and renal recovery: Consensus report of the acute disease quality initiative (ADQI) 16 workgroup. Nat Rev Nephrol. 13:241–257. 2017. View Article : Google Scholar : PubMed/NCBI | |
Fu Y and Dong Z: Immune response in COVID-19-associated acute kidney injury and maladaptive kidney repair. Integr Med Nephrol Androl. 10:e000222023. View Article : Google Scholar | |
Zhu Z, Hu J, Chen Z, Feng J, Yang X, Liang W and Ding G: Transition of acute kidney injury to chronic kidney disease: Role of metabolic reprogramming. Metabolism. 131:1551942022. View Article : Google Scholar : PubMed/NCBI | |
Niculae A, Gherghina ME, Peride I, Tiglis M, Nechita AM and Checherita IA: Pathway from acute kidney injury to chronic kidney disease: Molecules involved in renal fibrosis. Int J Mol Sci. 24:140192023. View Article : Google Scholar : PubMed/NCBI | |
Neyra JA and Chawla LS: Acute kidney disease to chronic kidney disease. Crit Care Clin. 37:453–474. 2021. View Article : Google Scholar : PubMed/NCBI | |
Nangaku M, Hirakawa Y, Mimura I, Inagi R and Tanaka T: Epigenetic changes in the acute kidney injury-to-chronic kidney disease transition. Nephron. 137:256–259. 2017. View Article : Google Scholar : PubMed/NCBI | |
Koh ES and Chung S: Recent update on acute kidney injury-to-chronic kidney disease transition. Yonsei Med J. 65:247–256. 2024. View Article : Google Scholar : PubMed/NCBI | |
Venkatachalam MA, Weinberg JM, Kriz W and Bidani AK: Failed tubule recovery, AKI-CKD transition, and kidney disease progression. J Am Soc Nephrol. 26:1765–1776. 2015. View Article : Google Scholar : PubMed/NCBI | |
Guo R, Duan J, Pan S, Cheng F, Qiao Y, Feng Q, Liu D and Liu Z: The road from AKI to CKD: Molecular mechanisms and therapeutic targets of ferroptosis. Cell Death Dis. 14:4262023. View Article : Google Scholar : PubMed/NCBI | |
André C, Bodeau S, Kamel S, Bennis Y and Caillard P: The AKI-to-CKD transition: The role of uremic toxins. Int J Mol Sci. 24:161522023. View Article : Google Scholar : PubMed/NCBI | |
Song Z and Gong X: Research progress on the potential mechanisms of acute kidney injury and chronic kidney disease induced by proton pump inhibitors. Integr Med Nephrol Androl. 10:e000272023. View Article : Google Scholar | |
Yu XY, Sun Q, Zhang YM, Zou L and Zhao YY: TGF-β/Smad signaling pathway in tubulointerstitial fibrosis. Front Pharmacol. 13:8605882022. View Article : Google Scholar | |
Ma TT and Meng XM: TGF-β/Smad and renal fibrosis. Adv Exp Med Biol. 1165:347–364. 2019. View Article : Google Scholar | |
Lan HY and Chung ACK: TGF-β/Smad signaling in kidney disease. Semin Nephrol. 32:236–243. 2012. View Article : Google Scholar : PubMed/NCBI | |
Lan HY: Diverse roles of TGF-β/Smads in renal fibrosis and inflammation. Int J Biol Sci. 7:1056–1067. 2011. View Article : Google Scholar : | |
Zou LL, Li JR, Li H, Tan JL, Wang MX, Liu NN, Gao RM, Yan HY, Wang XK, Dong B, et al: TGF-β isoforms inhibit hepatitis C virus propagation in transforming growth factor beta/SMAD protein signalling pathway dependent and independent manners. J Cell Mol Med. 25:3498–3510. 2021. View Article : Google Scholar : PubMed/NCBI | |
Sultana M, Tayyab M, Sunil, Parveen S, Hussain M, Saeed S, Riaz Z and Shabbir S: In silico molecular characterization of TGF-β gene family in Bufo bufo: Genome-wide analysis. J Biomol Struct Dyn. Feb 12–2024.Epub ahead of print. View Article : Google Scholar | |
Chen T, Zhu C, Wang X, Pan Y and Huang B: Asiatic acid encapsulated exosomes of hepatocellular carcinoma inhibit epithelial-mesenchymal transition through transforming growth factor beta/smad signaling pathway. J Biomed Nanotechnol. 17:2338–2350. 2021. View Article : Google Scholar | |
Chen Y, Di C, Zhang X, Wang J, Wang F, Yan JF, Xu C, Zhang J, Zhang Q, Li H, et al: Transforming growth factor β signaling pathway: A promising therapeutic target for cancer. J Cell Physiol. 235:1903–1914. 2020. View Article : Google Scholar | |
de Larco JE and Todaro GJ: Growth factors from murine sarcoma virus-transformed cells. Proc Natl Acad Sci USA. 75:4001–4005. 1978. View Article : Google Scholar : PubMed/NCBI | |
Wang HL, Wang L, Zhao CY and Lan HY: Role of TGF-beta signaling in beta cell proliferation and function in diabetes. Biomolecules. 12:3732022. View Article : Google Scholar : PubMed/NCBI | |
Kahata K, Dadras MS and Moustakas A: TGF-β family signaling in epithelial differentiation and epithelial-mesenchymal transition. Cold Spring Harb Perspect Biol. 10:a0221942018. View Article : Google Scholar | |
Song J and Shi W: The concomitant apoptosis and EMT underlie the fundamental functions of TGF-β. Acta Biochim Biophys Sin (Shanghai). 50:91–97. 2018. View Article : Google Scholar | |
Peng D, Fu M, Wang M, Wei Y and Wei X: Targeting TGF-β signal transduction for fibrosis and cancer therapy. Mol Cancer. 21:1042022. View Article : Google Scholar | |
Roberts AB, Anzano MA, Lamb LC, Smith JM and Sporn MB: New class of transforming growth factors potentiated by epidermal growth factor: Isolation from non-neoplastic tissues. Proc Natl Acad Sci USA. 78:5339–5343. 1981. View Article : Google Scholar : PubMed/NCBI | |
Moses HL, Branum EL, Proper JA and Robinson RA: Transforming growth factor production by chemically transformed cells. Cancer Res. 41:2842–2848. 1981.PubMed/NCBI | |
Roberts AB, Anzano MA, Meyers CA, Wideman J, Blacher R, Pan YC, Stein S, Lehrman SR, Smith JM, Lamb LC, et al: Purification and properties of a type beta transforming growth factor from bovine kidney. Biochemistry. 22:5692–5698. 1983. View Article : Google Scholar : PubMed/NCBI | |
Derynck R, Jarrett JA, Chen EY, Eaton DH, Bell JR, Assoian RK, Roberts AB, Sporn MB and Goeddel DV: Human transforming growth factor-beta complementary DNA sequence and expression in normal and transformed cells. Nature. 316:701–705. 1985. View Article : Google Scholar : PubMed/NCBI | |
Cheifetz S, Weatherbee JA, Tsang ML, Anderson JK, Mole JE, Lucas R and Massagué J: The transforming growth factor-beta system, a complex pattern of cross-reactive ligands and receptors. Cell. 48:409–415. 1987. View Article : Google Scholar : PubMed/NCBI | |
ten Dijke P, Hansen P, Iwata KK, Pieler C and Foulkes JG: Identification of another member of the transforming growth factor type beta gene family. Proc Natl Acad Sci USA. 85:4715–4719. 1988. View Article : Google Scholar : PubMed/NCBI | |
Derynck R, Lindquist PB, Lee A, Wen D, Tamm J, Graycar JL, Rhee L, Mason AJ, Miller DA, Coffey RJ, et al: A new type of transforming growth factor-beta, TGF-beta 3. EMBO J. 7:3737–3743. 1988. View Article : Google Scholar : PubMed/NCBI | |
Connor TB Jr, Roberts AB, Sporn MB, Danielpour D, Dart LL, Michels RG, de Bustros S, Enger C, Kato H, Lansing M, et al: Correlation of fibrosis and transforming growth factor-beta type 2 levels in the eye. J Clin Invest. 83:1661–1666. 1989. View Article : Google Scholar : PubMed/NCBI | |
Border WA, Okuda S, Languino LR, Sporn MB and Ruoslahti E: Suppression of experimental glomerulonephritis by antiserum against transforming growth factor beta 1. Nature. 346:371–374. 1990. View Article : Google Scholar : PubMed/NCBI | |
Pierce DF Jr, Gorska AE, Chytil A, Meise KS, Page DL, Coffey RJ Jr and Moses HL: Mammary tumor suppression by transforming growth factor beta 1 transgene expression. Proc Natl Acad Sci USA. 92:4254–4258. 1995. View Article : Google Scholar : PubMed/NCBI | |
Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, Fan RS, Zborowska E, Kinzler KW, Vogelstein B, et al: Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science. 268:1336–1338. 1995. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Feng X, We R and Derynck R: Receptor-associated Mad homologues synergize as effectors of the TGF-beta response. Nature. 383:168–172. 1996. View Article : Google Scholar : PubMed/NCBI | |
Eppert K, Scherer SW, Ozcelik H, Pirone R, Hoodless P, Kim H, Tsui LC, Bapat B, Gallinger S, Andrulis IL, et al: MADR2 maps to 18q21 and encodes a TGFbeta-regulated MAD-related protein that is functionally mutated in colorectal carcinoma. Cell. 86:543–552. 1996. View Article : Google Scholar : PubMed/NCBI | |
Fakhrai H, Mantil JC, Liu L, Nicholson GL, Murphy-Satter CS, Ruppert J and Shawler DL: Phase I clinical trial of a TGF-beta antisense-modified tumor cell vaccine in patients with advanced glioma. Cancer Gene Ther. 13:1052–1060. 2006. View Article : Google Scholar : PubMed/NCBI | |
Trachtman H, Fervenza FC, Gipson DS, Heering P, Jayne DR, Peters H, Rota S, Remuzzi G, Rump LC, Sellin LK, et al: A phase 1, single-dose study of fresolimumab, an anti-TGF-β antibody, in treatment-resistant primary focal segmental glomerulosclerosis. Kidney Int. 79:1236–1243. 2011. View Article : Google Scholar : PubMed/NCBI | |
King TE Jr, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, Gorina E, Hopkins PM, Kardatzke D, Lancaster L, et al: A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 370:2083–2092. 2014. View Article : Google Scholar : PubMed/NCBI | |
Rodon J, Carducci MA, Sepulveda-Sánchez JM, Azaro A, Calvo E, Seoane J, Braña I, Sicart E, Gueorguieva I, Cleverly AL, et al: First-in-human dose study of the novel transforming growth factor-β receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma. Clin Cancer Res. 21:553–560. 2015. View Article : Google Scholar | |
Yang S, Yang G, Wang X, Xiang J, Kang L and Liang Z: SIRT2 alleviated renal fibrosis by deacetylating SMAD2 and SMAD3 in renal tubular epithelial cells. Cell Death Dis. 14:6462023. View Article : Google Scholar : PubMed/NCBI | |
Miyazawa K, Itoh Y, Fu H and Miyazono K: Receptor-activated transcription factors and beyond: Multiple modes of Smad2/3-dependent transmission of TGF-β signaling. J Biol Chem. 300:1072562024. View Article : Google Scholar | |
Massagué J: TGF-beta signal transduction. Annu Rev Biochem. 67:753–791. 1998. View Article : Google Scholar : PubMed/NCBI | |
Itoh S and ten Dijke P: Negative regulation of TGF-beta receptor/Smad signal transduction. Curr Opin Cell Biol. 19:176–184. 2007. View Article : Google Scholar : PubMed/NCBI | |
Moustakas A and Heldin CH: The regulation of TGFbeta signal transduction. Development. 136:3699–3714. 2009. View Article : Google Scholar : PubMed/NCBI | |
Meng XM, Nikolic-Paterson DJ and Lan HY: TGF-β: The master regulator of fibrosis. Nat Rev Nephrol. 12:325–338. 2016. View Article : Google Scholar : PubMed/NCBI | |
Derynck R and Budi EH: Specificity, versatility, and control of TGF-β family signaling. Sci Signal. 12:eaav51832019. View Article : Google Scholar | |
Gewin LS: Transforming growth factor-β in the acute kidney injury to chronic kidney disease transition. Nephron. 143:154–157. 2019. View Article : Google Scholar | |
Yang Q, Ren GL, Wei B, Jin J, Huang XR, Shao W, Li J, Meng XM and Lan HY: Conditional knockout of TGF-βRII/Smad2 signals protects against acute renal injury by alleviating cell necroptosis, apoptosis and inflammation. Theranostics. 9:8277–8293. 2019. View Article : Google Scholar : | |
Gewin L: The many talents of transforming growth factor-β in the kidney. Curr Opin Nephrol Hypertens. 28:203–210. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hoi S, Tsuchiya H, Itaba N, Suzuki K, Oka H, Morimoto M, Takata T, Isomoto H and Shiota G: WNT/β-catenin signal inhibitor IC-2-derived small-molecule compounds suppress TGF-β1-induced fibrogenic response of renal epithelial cells by inhibiting SMAD2/3 signalling. Clin Exp Pharmacol Physiol. 47:940–946. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li J, Zou Y, Kantapan J, Su H, Wang L and Dechsupa N: TGF-β/Smad signaling in chronic kidney disease: Exploring post-translational regulatory perspectives (review). Mol Med Rep. 30:1432024. View Article : Google Scholar | |
Chen DQ, Cao G, Zhao H, Chen L, Yang T, Wang M, Vaziri ND, Guo Y and Zhao YY: Combined melatonin and poricoic acid A inhibits renal fibrosis through modulating the interaction of Smad3 and β-catenin pathway in AKI-to-CKD continuum. Ther Adv Chronic Dis. 10:20406223198691162019. View Article : Google Scholar | |
Kim IY, Song SH, Seong EY, Lee DW, Bae SS and Lee SB: Akt1 is involved in renal fibrosis and tubular apoptosis in a murine model of acute kidney injury-to-chronic kidney disease transition. Exp Cell Res. 424:1135092023. View Article : Google Scholar : PubMed/NCBI | |
Liu W, Li F, Guo D, Du C, Zhao S, Li J, Yan Z and Hao J: Schisandrin B alleviates renal tubular cell epithelial-mesenchymal transition and mitochondrial dysfunction by kielin/chordin-like protein upregulation via Akt pathway inactivation and adenosine 5′-monophosphate, AMP)-activated protein kinase pathway activation in diabetic kidney disease. Molecules. 28:78512023. View Article : Google Scholar | |
Kim IY, Park YK, Song SH, Seong EY, Lee DW, Bae SS and Lee SB: Role of Akt1 in renal fibrosis and tubular dedifferentiation during the progression of acute kidney injury to chronic kidney disease. Korean J Intern Med. 36:962–974. 2021. View Article : Google Scholar : | |
Zhou L, Chen X, Lu M, Wu Q, Yuan Q, Hu C, Miao J, Zhang Y, Li H, Hou FF, et al: Wnt/β-catenin links oxidative stress to podocyte injury and proteinuria. Kidney Int. 95:830–845. 2019. View Article : Google Scholar : PubMed/NCBI | |
Feng Y, Liang Y, Ren J and Dai C: Canonical Wnt signaling promotes macrophage proliferation during kidney fibrosis. Kidney Dis (Basel). 4:95–103. 2018. View Article : Google Scholar : PubMed/NCBI | |
Djudjaj S and Boor P: Cellular and molecular mechanisms of kidney fibrosis. Mol Aspects Med. 65:16–36. 2019. View Article : Google Scholar | |
Finke M, Kümpers P and Rovas A: Epidemiology and causes of acute renal failure and transition to chronic kidney disease. Dtsch Med Wochenschr. 147:227–235. 2022.In German. PubMed/NCBI | |
Leng X, Li Q, Chen W, Feng H, Li L, Yu L, Huang P, Ma P and Xie F: C-176 inhibits macrophage polarization towards M1-subtype and ameliorates LPS induced acute kidney injury. Eur J Pharmacol. 984:1770282024. View Article : Google Scholar : PubMed/NCBI | |
Ibrahim H, Sharawy MH, Hamed MF and Abu-Elsaad N: Peficitinib halts acute kidney injury via JAK/STAT3 and growth factors immunomodulation. Eur J Pharmacol. 984:1770202024. View Article : Google Scholar : PubMed/NCBI | |
Guzzi F, Cirillo L, Roperto RM, Romagnani P and Lazzeri E: Molecular mechanisms of the acute kidney injury to chronic kidney disease transition: An updated view. Int J Mol Sci. 20:49412019. View Article : Google Scholar : PubMed/NCBI | |
Belavgeni A, Meyer C, Stumpf J, Hugo C and Linkermann A: Ferroptosis and necroptosis in the kidney. Cell Chem Biol. 27:448–462. 2020. View Article : Google Scholar : PubMed/NCBI | |
Luo K: Signaling cross talk between TGF-β/Smad and other signaling pathways. Cold Spring Harb Perspect Biol. 9:a0221372017. View Article : Google Scholar | |
Seoane J, Le HV, Shen L, Anderson SA and Massagué J: Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell. 117:211–223. 2004. View Article : Google Scholar : PubMed/NCBI | |
Conery AR, Cao Y, Thompson EA, Townsend CM Jr, Ko TC and Luo K: Akt interacts directly with Smad3 to regulate the sensitivity to TGF-beta induced apoptosis. Nat Cell Biol. 6:366–372. 2004. View Article : Google Scholar : PubMed/NCBI | |
Wang M, Chen DQ, Chen L, Liu D, Zhao H, Zhang ZH, Vaziri ND, Guo Y, Zhao YY and Cao G: Novel RAS inhibitors poricoic acid ZG and poricoic Acid ZH attenuate renal fibrosis via a Wnt/β-catenin pathway and targeted phosphorylation of smad3 signaling. J Agric Food Chem. 66:1828–1842. 2018. View Article : Google Scholar : PubMed/NCBI | |
Takekawa M, Tatebayashi K, Itoh F, Adachi M, Imai K and Saito H: Smad-dependent GADD45beta expression mediates delayed activation of p38 MAP kinase by TGF-beta. EMBO J. 21:6473–6482. 2002. View Article : Google Scholar : PubMed/NCBI | |
Yeh YY, Chiao CC, Kuo WY, Hsiao YC, Chen YJ, Wei YY, Lai TH, Fong YC and Tang CH: TGF-beta1 increases motility and alphavbeta3 integrin up-regulation via PI3K, Akt and NF-kappaB-dependent pathway in human chondrosarcoma cells. Biochem Pharmacol. 75:1292–1301. 2008. View Article : Google Scholar : PubMed/NCBI | |
Ogawa K, Chen F, Kuang C and Chen Y: Suppression of matrix metalloproteinase-9 transcription by transforming growth factor-beta is mediated by a nuclear factor-kappaB site. Biochem J. 381:413–422. 2004. View Article : Google Scholar : PubMed/NCBI | |
Liu HJ, Miao H, Yang JZ, Liu F, Cao G and Zhao YY: Deciphering the role of lipoproteins and lipid metabolic alterations in ageing and ageing-associated renal fibrosis. Ageing Res Rev. 85:1018612023. View Article : Google Scholar : PubMed/NCBI | |
Liu Y: kidney fibrosis: Fundamental questions, challenges, and perspectives. Integr Med Nephrol Androl. 11:e24–00027. 2024. View Article : Google Scholar | |
Wang Y, Guo J, Shao B, Chen H and Lan H: The Role of TGF-β1/SMAD in diabetic nephropathy: Mechanisms and research development. Sichuan Da Xue Xue Bao Yi Xue Ban. 54:1065–1073. 2023.In Chinese. | |
Zhang J, Cao L, Wang X, Li Q, Zhang M, Cheng C, Yu L, Xue F, Sui W, Sun S, et al: The E3 ubiquitin ligase TRIM31 plays a critical role in hypertensive nephropathy by promoting proteasomal degradation of MAP3K7 in the TGF-β1 signaling pathway. Cell Death Differ. 29:556–567. 2022. View Article : Google Scholar | |
Dan Hu Q, Wang HL, Liu J, He T, Tan RZ, Zhang Q, Su HW, Kantawong F, Lan HY and Wang L: Btg2 promotes focal segmental glomerulosclerosis via smad3-dependent podocyte-mesenchymal transition. Adv Sci (Weinh). 10:e23043602023. View Article : Google Scholar : PubMed/NCBI | |
Chalkia A, Gakiopoulou H, Theohari I, Foukas PG, Vassilopoulos D and Petras D: Transforming growth factor-β1/Smad signaling in glomerulonephritis and its association with progression to chronic kidney disease. Am J Nephrol. 52:653–665. 2021. View Article : Google Scholar | |
Chen L, Yang T, Lu DW, Zhao H, Feng YL, Chen H, Chen DQ, Vaziri ND and Zhao YY: Central role of dysregulation of TGF-β/Smad in CKD progression and potential targets of its treatment. Biomed Pharmacother. 101:670–681. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang W, Li X, Tang Y, Chen C, Jing R and Liu T: miR-155-5p implicates in the pathogenesis of renal fibrosis via targeting SOCS1 and SOCS6. Oxid Med Cell Longev. 2020:62639212020.PubMed/NCBI | |
Wang R, Wu G, Dai T, Lang Y, Chi Z, Yang S and Dong D: Naringin attenuates renal interstitial fibrosis by regulating the TGF-β/Smad signaling pathway and inflammation. Exp Ther Med. 21:662021. View Article : Google Scholar | |
Mai X, Shang J, Chen Q, Gu S, Hong Y, Zhou J and Zhang M: Endophilin A2 protects against renal fibrosis by targeting TGF-β/Smad signaling. FASEB J. 36:e226032022. View Article : Google Scholar | |
El-Waseif EG, Sharawy MH and Suddek GM: The modulatory effect of sodium molybdate against cisplatin-induced CKD: Role of TGF-β/Smad signaling pathway. Life Sci. 306:1208452022. View Article : Google Scholar | |
Zou X, Wu M, Tu M, Tan X, Long Y, Xu Y and Li M: 4-Octyl itaconate inhibits high glucose induced renal tubular epithelial cell fibrosis through TGF-β-ROS pathway. J Recept Signal Transduct Res. 44:27–34. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Zha H, Huang J and Shi L: Flavin containing monooxygenase 2 regulates renal tubular cell fibrosis and paracrine secretion via SMURF2 in AKI-CKD transformation. Int J Mol Med. 52:1102023. View Article : Google Scholar : | |
Kurzhagen JT, Dellepiane S, Cantaluppi V and Rabb H: AKI: An increasingly recognized risk factor for CKD development and progression. J Nephrol. 33:1171–1187. 2020. View Article : Google Scholar : PubMed/NCBI | |
Rayego-Mateos S, Marquez-Expósito L, Rodrigues-Diez R, Sanz AB, Guiteras R, Doladé N, Rubio-Soto I, Manonelles A, Codina S, Ortiz A, et al: Molecular mechanisms of kidney injury and repair. Int J Mol Sci. 23:15422022. View Article : Google Scholar : PubMed/NCBI | |
Zhao M, Wang Y, Li L, Liu S, Wang C, Yuan Y, Yang G, Chen Y, Cheng J, Lu Y and Liu J: Mitochondrial ROS promote mitochondrial dysfunction and inflammation in ischemic acute kidney injury by disrupting TFAM-mediated mtDNA maintenance. Theranostics. 11:1845–1863. 2021. View Article : Google Scholar : PubMed/NCBI | |
Canaud G and Bonventre JV: Cell cycle arrest and the evolution of chronic kidney disease from acute kidney injury. Nephrol Dial Transplant. 30:575–583. 2015. View Article : Google Scholar : | |
Wang Z and Zhang C: From AKI to CKD: Maladaptive repair and the underlying mechanisms. Int J Mol Sci. 23:108802022. View Article : Google Scholar : PubMed/NCBI | |
Wu M, Chen W, Miao M, Jin Q, Zhang S, Bai M, Fan J, Zhang Y, Zhang A, Jia Z and Huang S: Anti-anemia drug FG4592 retards the AKI-to-CKD transition by improving vascular regeneration and antioxidative capability. Clin Sci (Lond). 135:1707–1726. 2021. View Article : Google Scholar : PubMed/NCBI | |
Huang R, Fu P and Ma L: Kidney fibrosis: From mechanisms to therapeutic medicines. Signal Transduct Target Ther. 8:1292023. View Article : Google Scholar : PubMed/NCBI | |
Li L, Fu H and Liu Y: The fibrogenic niche in kidney fibrosis: Components and mechanisms. Nat Rev Nephrol. 18:545–557. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zheng D, Liu J, Piao H, Zhu Z, Wei R and Liu K: ROS-triggered endothelial cell death mechanisms: Focus on pyroptosis, parthanatos, and ferroptosis. Front Immunol. 13:10392412022. View Article : Google Scholar : PubMed/NCBI | |
Molema G, Zijlstra JG, van Meurs M and Kamps JAAM: Renal microvascular endothelial cell responses in sepsis-induced acute kidney injury. Nat Rev Nephrol. 18:95–112. 2022. View Article : Google Scholar | |
Jourde-Chiche N, Fakhouri F, Dou L, Bellien J, Burtey S, Frimat M, Jarrot PA, Kaplanski G, Le Quintrec M, Pernin V, et al: Endothelium structure and function in kidney health and disease. Nat Rev Nephrol. 15:87–108. 2019. View Article : Google Scholar : PubMed/NCBI | |
Aranda-Rivera AK, Cruz-Gregorio A, Aparicio-Trejo OE and Pedraza-Chaverri J: Mitochondrial redox signaling and oxidative stress in kidney diseases. Biomolecules. 11:11442021. View Article : Google Scholar : PubMed/NCBI | |
Kishi S, Nagasu H, Kidokoro K and Kashihara N: Oxidative stress and the role of redox signalling in chronic kidney disease. Nat Rev Nephrol. 20:101–119. 2024. View Article : Google Scholar | |
Fontecha-Barriuso M, Martin-Sanchez D, Martinez-Moreno JM, Monsalve M, Ramos AM, Sanchez-Niño MD, Ruiz-Ortega M, Ortiz A and Sanz AB: The role of PGC-1α and mitochondrial biogenesis in kidney diseases. Biomolecules. 10:3472020. View Article : Google Scholar | |
Zhang X, Agborbesong E and Li X: The role of mitochondria in acute kidney injury and chronic kidney disease and its therapeutic potential. Int J Mol Sci. 22:112532021. View Article : Google Scholar : PubMed/NCBI | |
Grgic I, Duffield JS and Humphreys BD: The origin of interstitial myofibroblasts in chronic kidney disease. Pediatr Nephrol. 27:183–193. 2012. View Article : Google Scholar | |
Yeh TH, Tu KC, Wang HY and Chen JY: From acute to chronic: Unraveling the pathophysiological mechanisms of the progression from acute kidney injury to acute kidney disease to chronic kidney Disease. Int J Mol Sci. 25:17552024. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Kumar S, Dolzhenko E, Alvarado GF, Guo J, Lu C, Chen Y, Li M, Dessing MC, Parvez RK, et al: Molecular characterization of the transition from acute to chronic kidney injury following ischemia/reperfusion. JCI Insight. 2:e947162017. View Article : Google Scholar : PubMed/NCBI | |
Li H, Hu L, Zheng C, Kong Y, Liang M and Li Q: Ankrd1 as a potential biomarker for the transition from acute kidney injury to chronic kidney disease. Sci Rep. 15:46592025. View Article : Google Scholar : PubMed/NCBI | |
Wen Y, Xu L, Melchinger I, Thiessen-Philbrook H, Moledina DG, Coca SG, Hsu CY, Go AS, Liu KD, Siew ED, et al: Longitudinal biomarkers and kidney disease progression after acute kidney injury. JCI Insight. 8:e1677312023. View Article : Google Scholar : PubMed/NCBI | |
Muto Y, Dixon EE, Yoshimura Y, Wu H, Omachi K, Ledru N, Wilson PC, King AJ, Eric Olson N, Gunawan MG, et al: Defining cellular complexity in human autosomal dominant polycystic kidney disease by multimodal single cell analysis. Nat Commun. 13:64972022. View Article : Google Scholar : PubMed/NCBI | |
Kirita Y, Wu H, Uchimura K, Wilson PC and Humphreys BD: Cell profiling of mouse acute kidney injury reveals conserved cellular responses to injury. Proc Natl Acad Sci USA. 117:15874–15883. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ma T, Li H, Liu H, Peng Y, Lin T, Deng Z, Jia N, Chen Z and Wang P: Neat1 promotes acute kidney injury to chronic kidney disease by facilitating tubular epithelial cells apoptosis via sequestering miR-129-5p. Mol Ther. 30:3313–3332. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zheng Z, Xu K, Li C, Qi C, Fang Y, Zhu N, Bao J, Zhao Z, Yu Q, Wu H and Liu J: NLRP3 associated with chronic kidney disease progression after ischemia/reperfusion-induced acute kidney injury. Cell Death Discov. 7:3242021. View Article : Google Scholar : PubMed/NCBI | |
Fu Y, Xiang Y, Wang Y, Liu Z, Yang D, Zha J, Tang C, Cai J, Chen G and Dong Z: The STAT1/HMGB1/NF-κB pathway in chronic inflammation and kidney injury after cisplatin exposure. Theranostics. 13:2757–2773. 2023. View Article : Google Scholar : | |
Cui N, Liu C, Tang X, Song L, Xiao Z, Wang C, Wu Y, Zhou Y, Peng C, Liu Y, et al: ISG15 accelerates acute kidney injury and the subsequent AKI-to-CKD transition by promoting TGFβR1 ISGylation. Theranostics. 14:4536–4553. 2024. View Article : Google Scholar : | |
Doke T, Mukherjee S, Mukhi D, Dhillon P, Abedini A, Davis JG, Chellappa K, Chen B, Baur JA and Susztak K: NAD+ precursor supplementation prevents mtRNA/RIG-I-dependent inflammation during kidney injury. Nat Metab. 5:414–430. 2023. View Article : Google Scholar : PubMed/NCBI | |
Cai Y, Chen J, Liu J, Zhu K, Xu Z, Shen J, Wang D and Chu L: Identification of six hub genes and two key pathways in two rat renal fibrosis models based on bioinformatics and RNA-seq transcriptome analyses. Front Mol Biosci. 9:10357722022. View Article : Google Scholar : PubMed/NCBI | |
Berezin AE, Berezina TA, Hoppe UC, Lichtenauer M and Berezin AA: An overview of circulating and urinary biomarkers capable of predicting the transition of acute kidney injury to chronic kidney disease. Expert Rev Mol Diagn. 24:627–647. 2024. View Article : Google Scholar : PubMed/NCBI | |
Puri B, Majumder S and Gaikwad AB: Novel dysregulated long non-coding RNAs in the acute kidney injury-to-chronic kidney diseases transition unraveled by transcriptomic analysis. Pharmacol Res Perspect. 12:e700362024. View Article : Google Scholar : PubMed/NCBI | |
Badurdeen Z, Alli-Shaik A, Ratnatunga NVI, Abeysekera TDJ, Wijetunge S, Hemage RKD, Fernando BNTW, Hettiarachchi TW, Gunaratne J and Nanayakkara N: Serum transforming growth factor-beta 1 and creatinine for early diagnosis of CKD of unknown or uncertain etiology phenotypes. Kidney Int Rep. 8:368–372. 2022. View Article : Google Scholar | |
Wu W, Wang X, Yu X and Lan HY: Smad3 signatures in renal inflammation and fibrosis. Int J Biol Sci. 18:2795–2806. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kuang Q, Wu S, Xue N, Wang X, Ding X and Fang Y: Selective Wnt/β-Catenin pathway activation concomitant with sustained overexpression of miR-21 is responsible for aristolochic acid-induced AKI-to-CKD transition. Front Pharmacol. 12:6672822021. View Article : Google Scholar | |
Nath KA, Croatt AJ, Warner GM and Grande JP: Genetic deficiency of Smad3 protects against murine ischemic acute kidney injury. Am J Physiol Renal Physiol. 301:F436–F442. 2011. View Article : Google Scholar : PubMed/NCBI | |
Yan Z, Wang G and Shi X: Advances in the progression and prognosis biomarkers of chronic kidney disease. Front Pharmacol. 12:7853752021. View Article : Google Scholar | |
González-Nicolás MÁ, González-Guerrero C, Goicoechea M, Boscá L, Valiño-Rivas L and Lázaro A: Biomarkers in contrast-induced acute kidney injury: Towards A new perspective. Int J Mol Sci. 25:34382024. View Article : Google Scholar : PubMed/NCBI | |
Wang JY, Gao YB, Zhang N, Zou DW, Wang P, Zhu ZY, Li JY, Zhou SN, Wang SC, Wang YY and Yang JK: miR-21 overexpression enhances TGF-β1-induced epithelial-to-mesenchymal transition by target smad7 and aggravates renal damage in diabetic nephropathy. Mol Cell Endocrinol. 392:163–172. 2014. View Article : Google Scholar : PubMed/NCBI | |
Tsuji K, Nakanoh H, Fukushima K, Kitamura S and Wada J: MicroRNAs as biomarkers and therapeutic targets for acute kidney injury. Diagnostics (Basel). 13:28932023. View Article : Google Scholar : PubMed/NCBI | |
Chen C, Lu C, Qian Y, Li H, Tan Y, Cai L and Weng H: Urinary miR-21 as a potential biomarker of hypertensive kidney injury and fibrosis. Sci Rep. 7:177372017. View Article : Google Scholar : PubMed/NCBI | |
Lv LL, Cao YH, Ni HF, Xu M, Liu D, Liu H, Chen PS and Liu BC: MicroRNA-29c in urinary exosome/microvesicle as a biomarker of renal fibrosis. Am J Physiol Renal Physiol. 305:F1220–F1227. 2013. View Article : Google Scholar : PubMed/NCBI | |
He L, Wei Q, Liu J, Yi M, Liu Y, Liu H, Sun L, Peng Y, Liu F, Venkatachalam MA and Dong Z: AKI on CKD: Heightened injury, suppressed repair, and the underlying mechanisms. Kidney Int. 92:1071–1083. 2017. View Article : Google Scholar : PubMed/NCBI | |
Lu Y, Xu S, Tang R, Han C and Zheng C: A potential link between fibroblast growth factor-23 and the progression of AKI to CKD. BMC Nephrol. 24:872023. View Article : Google Scholar : PubMed/NCBI | |
Gifford CC, Lian F, Tang J, Costello A, Goldschmeding R, Samarakoon R and Higgins PJ: PAI-1 induction during kidney injury promotes fibrotic epithelial dysfunction via deregulation of klotho, p53, and TGF-β1-receptor signaling. FASEB J. 35:e217252021. View Article : Google Scholar | |
Paniagua-Sancho M, Quiros Y, Casanova AG, Blanco-Gozalo V, Agüeros-Blanco C, Benito-Hernández A, Ramos-Barron MA, Gómez-Alamillo C, Arias M, Sancho-Martínez SM and López-Hernández FJ: Urinary plasminogen activator inhibitor-1: A biomarker of acute tubular injury. Am J Nephrol. 52:714–724. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yang L, Si P, Kuerban T, Guo L, Zhan S, Zuhaer Y, Zuo Y, Lu P, Bai X and Liu T: UHRF1 promotes epithelial-mesenchymal transition mediating renal fibrosis by activating the TGF-β/SMAD signaling pathway. Sci Rep. 15:33462025. View Article : Google Scholar | |
Livingston MJ, Shu S, Fan Y, Li Z, Jiao Q, Yin XM, Venkatachalam MA and Dong Z: Tubular cells produce FGF2 via autophagy after acute kidney injury leading to fibroblast activation and renal fibrosis. Autophagy. 19:256–277. 2023. View Article : Google Scholar : | |
Ren LL, Miao H, Wang YN, Liu F, Li P and Zhao YY: TGF-β as A master regulator of aging-associated tissue fibrosis. Aging Dis. 14:1633–1650. 2023. View Article : Google Scholar : PubMed/NCBI | |
Fei S, Ma Y, Zhou B, Chen X, Zhang Y, Yue K, Li Q, Gui Y, Xiang T, Liu J, et al: Platelet membrane biomimetic nanoparticle-targeted delivery of TGF-β1 siRNA attenuates renal inflammation and fibrosis. Int J Pharm. 659:1242612024. View Article : Google Scholar | |
Wang B, Ding X, Ding C, Tesch G, Zheng J, Tian P, Ricardo S, Shen HH and Xue W: WNT1-inducible-signaling pathway protein 1 regulates the development of kidney fibrosis through the TGF-β1 pathway. FASEB J. 34:14507–14520. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Wang B, Zhang A, Hassounah F, Seow Y, Wood M, Ma F, Klein JD, Price SR and Wang XH: Exosome-mediated miR-29 transfer reduces muscle atrophy and kidney fibrosis in mice. Mol Ther. 27:571–583. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang B, Jha JC, Hagiwara S, McClelland AD, Jandeleit-Dahm K, Thomas MC, Cooper ME and Kantharidis P: Transforming growth factor-β1-mediated renal fibrosis is dependent on the regulation of transforming growth factor receptor 1 expression by let-7b. Kidney Int. 85:352–361. 2014. View Article : Google Scholar | |
Qin W, Chung AC, Huang XR, Meng XM, Hui DS, Yu CM, Sung JJ and Lan HY: TGF-β/Smad3 signaling promotes renal fibrosis by inhibiting miR-29. J Am Soc Nephrol. 22:1462–1474. 2011. View Article : Google Scholar : PubMed/NCBI | |
Kato M, Arce L, Wang M, Putta S, Lanting L and Natarajan R: A microRNA circuit mediates transforming growth factor-β1 autoregulation in renal glomerular mesangial cells. Kidney Int. 80:358–368. 2011. View Article : Google Scholar : PubMed/NCBI | |
Singh SP, Tao S, Fields TA, Webb S, Harris RC and Rao R: Glycogen synthase kinase-3 inhibition attenuates fibroblast activation and development of fibrosis following renal ischemia-reperfusion in mice. Dis Model Mech. 8:931–940. 2015.PubMed/NCBI | |
Wang X, Feng S, Fan J, Li X, Wen Q and Luo N: New strategy for renal fibrosis: Targeting Smad3 proteins for ubiquitination and degradation. Biochem Pharmacol. 116:200–209. 2016. View Article : Google Scholar : PubMed/NCBI | |
Jinnin M, Ihn H and Tamaki K: Characterization of SIS3, a novel specific inhibitor of Smad3, and its effect on transforming growth factor-beta1-induced extracellular matrix expression. Mol Pharmacol. 69:597–607. 2006. View Article : Google Scholar | |
Lan HY: Smad7 as a therapeutic agent for chronic kidney diseases. Front Biosci. 13:4984–4992. 2008. View Article : Google Scholar : PubMed/NCBI | |
Meng XM, Chung AC and Lan HY: Role of the TGF-β/BMP-7/Smad pathways in renal diseases. Clin Sci (Lond). 124:243–254. 2013. View Article : Google Scholar | |
Kim S, Jeong CH, Song SH, Um JE, Kim HS, Yun JS, Han D, Cho ES, Nam BY, Yook JI, et al: Micellized protein transduction domain-bone morphogenetic protein-7 efficiently blocks renal fibrosis via inhibition of transforming growth factor-beta-mediated epithelial-mesenchymal transition. Front Pharmacol. 11:5912752020. View Article : Google Scholar : PubMed/NCBI | |
Liu R, Das B, Xiao W, Li Z, Li H, Lee K and He JC: A novel inhibitor of homeodomain interacting protein kinase 2 mitigates kidney fibrosis through inhibition of the TGF-β1/Smad3 pathway. J Am Soc Nephrol. 28:2133–2143. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chang X, Zhen X, Liu J, Ren X, Hu Z, Zhou Z, Zhu F, Ding K and Nie J: The antihelmenthic phosphate niclosamide impedes renal fibrosis by inhibiting homeodomain-interacting protein kinase 2 expression. Kidney Int. 92:612–624. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Li Z, Cao K, Fang D, Wang F, Bi G, Yang J, He Y, Wu J, Wei Y and Song X: Adjunctive therapy with statins reduces residual albuminuria/proteinuria and provides further renoprotection by downregulating the angiotensin II-AT1 pathway in hypertensive nephropathy. J Hypertens. 35:1442–1456. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tian T, Zhang J, Zhu X, Wen S, Shi D and Zhou H: FTY720 ameliorates renal fibrosis by simultaneously affecting leucocyte recruitment and TGF-β signalling in fibroblasts. Clin Exp Immunol. 190:68–78. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhou X, Zhang J, Xu C and Wang W: Curcumin ameliorates renal fibrosis by inhibiting local fibroblast proliferation and extracellular matrix deposition. J Pharmacol Sci. 126:344–350. 2014. View Article : Google Scholar : PubMed/NCBI | |
Qin T, Yin S, Yang J, Zhang Q, Liu Y, Huang F and Cao W: Sinomenine attenuates renal fibrosis through Nrf2-mediated inhibition of oxidative stress and TGFβ signaling. Toxicol Appl Pharmacol. 304:1–8. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wang HW, Shi L, Xu YP, Qin XY and Wang QZ: Oxymatrine inhibits renal fibrosis of obstructive nephropathy by downregulating the TGF-β1-Smad3 pathway. Ren Fail. 38:945–951. 2016. View Article : Google Scholar : PubMed/NCBI | |
Liu L, Wang Y, Yan R, Li S, Shi M, Xiao Y and Guo B: Oxymatrine inhibits renal tubular EMT induced by high glucose via upregulation of SnoN and inhibition of TGF-β1/Smad signaling pathway. PLoS One. 11:e01519862016. View Article : Google Scholar | |
Wang DT, Huang RH, Cheng X, Zhang ZH, Yang YJ and Lin X: Tanshinone IIA attenuates renal fibrosis and inflammation via altering expression of TGF-β/Smad and NF-κB signaling pathway in 5/6 nephrectomized rats. Int Immunopharmacol. 26:4–12. 2015. View Article : Google Scholar : PubMed/NCBI | |
Cheng H, Bo Y, Shen W, Tan J, Jia Z, Xu C and Li F: Leonurine ameliorates kidney fibrosis via suppressing TGF-β and NF-κB signaling pathway in UUO mice. Int Immunopharmacol. 25:406–415. 2015. View Article : Google Scholar : PubMed/NCBI | |
Luo Q, Tian L, Di L, Yan YM, Wei XY, Wang XF and Cheng YX: (±)-Sinensilactam A, a pair of rare hybrid metabolites with Smad3 phosphorylation inhibition from Ganoderma sinensis. Org Lett. 17:1565–1568. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Liu N, Su X, Zhou G, Sun G, Du F, Bian X and Wang B: Epigallocatechin-3-gallate attenuates transforming growth factor-β1 induced epithelial-mesenchymal transition via Nrf2 regulation in renal tubular epithelial cells. Biomed Pharmacother. 70:260–267. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Li Z, He W, Xu L, Wang J, Shi J and Sheng M: Effects of astragaloside IV against the TGF-β1-induced epithelial-to-mesenchymal transition in peritoneal mesothelial cells by promoting smad 7 expression. Cell Physiol Biochem. 37:43–54. 2015. View Article : Google Scholar | |
Wang L, Chi YF, Yuan ZT, Zhou WC, Yin PH, Zhang XM, Peng W and Cai H: Astragaloside IV inhibits renal tubulointerstitial fibrosis by blocking TGF-β/Smad signaling pathway in vivo and in vitro. Exp Biol Med (Maywood). 239:1310–1324. 2014. View Article : Google Scholar : PubMed/NCBI | |
Dou F, Ding Y, Wang C, Duan J, Wang W, Xu H, Zhao X, Wang J and Wen A: Chrysophanol ameliorates renal interstitial fibrosis by inhibiting the TGF-β/Smad signaling pathway. Biochem Pharmacol. 180:1140792020. View Article : Google Scholar | |
Chen SJ, Wu P, Sun LJ, Zhou B, Niu W, Liu S, Lin FJ and Jiang GR: miR-204 regulates epithelial-mesenchymal transition by targeting SP1 in the tubular epithelial cells after acute kidney injury induced by ischemia-reperfusion. Oncol Rep. 37:1148–1158. 2017. View Article : Google Scholar | |
Song J, Yu W, Chen S, Huang J, Zhou C and Liang H: Remimazolam attenuates inflammation and kidney fibrosis following folic acid injury. Eur J Pharmacol. 966:1763422024. View Article : Google Scholar : PubMed/NCBI | |
Douvris A, Viñas JL, Gutsol A, Zimpelmann J, Burger D and Burns KD: miR-486-5p protects against rat ischemic kidney injury and prevents the transition to chronic kidney disease and vascular dysfunction. Clin Sci (Lond). 138:599–614. 2024. View Article : Google Scholar : PubMed/NCBI | |
Játiva S, Torrico S, Calle P, Muñoz Á, García M, Larque AB, Poch E and Hotter G: NGAL release from peripheral blood mononuclear cells protects against acute kidney injury and prevents AKI induced fibrosis. Biomed Pharmacother. 153:1134152022. View Article : Google Scholar : PubMed/NCBI | |
Torrico S, Hotter G, Muñoz Á, Calle P, García M, Poch E and Játiva S: PBMC therapy reduces cell death and tissue fibrosis after acute kidney injury by modulating the pattern of monocyte/macrophage survival in tissue. Biomed Pharmacother. 178:1171862024. View Article : Google Scholar : PubMed/NCBI | |
Chiang CH, Lan TY, Hsieh JH, Lin SC, Chen JW and Chang TT: Diosgenin reduces acute kidney injury and ameliorates the progression to chronic kidney disease by modifying the NOX4/p65 signaling pathways. J Agric Food Chem. 72:17444–17454. 2024. View Article : Google Scholar : PubMed/NCBI | |
Xin W, Gong S, Chen Y, Yao M, Qin S, Chen J, Zhang A, Yu W, Zhou S, Zhang B, et al: Self-assembling P38 peptide inhibitor nanoparticles ameliorate the transition from acute to chronic kidney disease by suppressing ferroptosis. Adv Healthc Mater. 13:e24004412024. View Article : Google Scholar : PubMed/NCBI | |
Perez-Moreno E, Toledo T, Campusano P, Zuñiga S, Azócar L, Feuerhake T, Méndez GP, Labarca M, Pérez-Molina F, de la Peña A, et al: Galectin-8 counteracts folic acid-induced acute kidney injury and prevents its transition to fibrosis. Biomed Pharmacother. 177:1169232024. View Article : Google Scholar : PubMed/NCBI | |
Barrera-Chimal J, Rocha L, Amador-Martínez I, Pérez-Villalva R, González R, Cortés-González C, Uribe N, Ramírez V, Berman N, Gamba G and Bobadilla NA: Delayed spironolactone administration prevents the transition from acute kidney injury to chronic kidney disease through improving renal inflammation. Nephrol Dial Transplant. 34:794–801. 2019. View Article : Google Scholar | |
Li ZL, Wang B, Lv LL, Tang TT, Wen Y, Cao JY, Zhu XX, Feng ST, Crowley SD and Liu BC: FIH-1-modulated HIF-1α C-TAD promotes acute kidney injury to chronic kidney disease progression via regulating KLF5 signaling. Acta Pharmacol Sin. 42:2106–2119. 2021. View Article : Google Scholar : PubMed/NCBI | |
Guo X, Xu L, Velazquez H, Chen TM, Williams RM, Heller DA, Burtness B, Safirstein R and Desir GV: Kidney-targeted renalase agonist prevents cisplatin-induced chronic kidney disease by inhibiting regulated necrosis and inflammation. J Am Soc Nephrol. 33:342–356. 2022. View Article : Google Scholar : | |
Czopek A, Moorhouse R, Gallacher PJ, Pugh D, Ivy JR, Farrah TE, Godden E, Hunter RW, Webb DJ, Tharaux PL, et al: Endothelin blockade prevents the long-term cardiovascular and renal sequelae of acute kidney injury in mice. Sci Transl Med. 14:eabf50742022. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Bai X, Chen J, Huang M, Hong Q, Ouyang Q, Sun X, Zhang Y, Liu J, Wang X, et al: Pyruvate kinase M2 regulates kidney fibrosis through pericyte glycolysis during the progression from acute kidney injury to chronic kidney disease. Cell Prolif. 57:e135482024. View Article : Google Scholar : | |
Hu Z, Zhan J, Pei G and Zeng R: Depletion of macrophages with clodronate liposomes partially attenuates renal fibrosis on AKI-CKD transition. Ren Fail. 45:21494122023. View Article : Google Scholar : PubMed/NCBI | |
Gu L, Gao Q, Ni L, Wang M and Shen F: Fasudil inhibits epithelial-myofibroblast transdifferentiation of human renal tubular epithelial HK-2 cells induced by high glucose. Chem Pharm Bull (Tokyo). 61:688–694. 2013. View Article : Google Scholar : PubMed/NCBI | |
Wang Z, Perez M, Lee ES, Kojima S and Griffin M: The functional relationship between transglutaminase 2 and transforming growth factor β1 in the regulation of angiogenesis and endothelial-mesenchymal transition. Cell Death Dis. 8:e30322017. View Article : Google Scholar | |
Cao Y, Su H, Zeng J, Xie Y, Liu Z, Liu F, Qiu Y, Yi F, Lin J, Hammes HP and Zhang C: Integrin β8 prevents pericyte-myofibroblast transition and renal fibrosis through inhibiting the TGF-β1/TGFBR1/Smad3 pathway in diabetic kidney disease. Transl Res. 265:36–50. 2024. View Article : Google Scholar | |
Li J, Qu X, Yao J, Caruana G, Ricardo SD, Yamamoto Y, Yamamoto H and Bertram JF: Blockade of endothelial-mesenchymal transition by a Smad3 inhibitor delays the early development of streptozotocin-induced diabetic nephropathy. Diabetes. 59:2612–2624. 2010. View Article : Google Scholar : PubMed/NCBI | |
Akhurst RJ and Hata A: Targeting the TGFβ signalling pathway in disease. Nat Rev Drug Discov. 11:790–811. 2012. View Article : Google Scholar : PubMed/NCBI | |
Peñalva MA, Zhang J, Xiang X and Pantazopoulou A: Transport of fungal RAB11 secretory vesicles involves myosin-5, dynein/dynactin/p25, and kinesin-1 and is independent of kinesin-3. Mol Biol Cell. 28:947–961. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tzavlaki K and Moustakas A: TGF-β signaling. Biomolecules. 10:4872020. View Article : Google Scholar | |
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C and He J: TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther. 9:612024. View Article : Google Scholar | |
Rodón J, Carducci M, Sepulveda-Sánchez JM, Azaro A, Calvo E, Seoane J, Braña I, Sicart E, Gueorguieva I, Cleverly A, et al: Pharmacokinetic, pharmacodynamic and biomarker evaluation of transforming growth factor-β receptor I kinase inhibitor, galunisertib, in phase 1 study in patients with advanced cancer. Invest New Drugs. 33:357–370. 2015. View Article : Google Scholar | |
Kawamura M, Sato S, Matsumoto G, Fukuda T, Shiba-Fukushima K, Noda S, Takanashi M, Mori N and Hattori N: Loss of nuclear REST/NRSF in aged-dopaminergic neurons in Parkinson's disease patients. Neurosci Lett. 699:59–63. 2019. View Article : Google Scholar : PubMed/NCBI | |
Levey AS, Inker LA, Matsushita K, Greene T, Willis K, Lewis E, de Zeeuw D, Cheung AK and Coresh J: GFR decline as an end point for clinical trials in CKD: A scientific workshop sponsored by the National Kidney Foundation and the US food and drug administration. Am J Kidney Dis. 64:821–835. 2014. View Article : Google Scholar : PubMed/NCBI | |
Levey AS, Gansevoort RT, Coresh J, Inker LA, Heerspink HL, Grams ME, Greene T, Tighiouart H, Matsushita K, Ballew SH, et al: Change in albuminuria and GFR as end points for clinical trials in early stages of CKD: A scientific workshop sponsored by the national kidney foundation in collaboration with the US food and drug administration and european medicines agency. Am J Kidney Dis. 75:84–104. 2020. View Article : Google Scholar |