Acessibilidade / Reportar erro

Epithelial-Mesenchymal Transition and its Role in Renal Fibrogenesis

Abstract

Renal fibrosis in chronic renal failure poses a major challenge with regard to providing specific therapeutic strategies.

The current hypothesis of the epithelial-mesenchymal transition (EMT) in renal fibrosis proposes that matrix producing fibroblasts and myofibroblasts generated in the diseased kidney causes fibrosis. This paper explores the pathogenetic mechanisms, the role of EMT in renal fibrogenesis in chronic kidney disease, biomarkers, and EMT related anti-fibrotic therapy.

Keywords:
renal fibrosis; myofibroblast; epithelial-mesenchymal transition; chronic kidney disease; anti-fibrotic therapy.

HIGHLIGHTS

  • Exploring the role of TGFβ and its associate genes in the pathogenetic mechanism of EMT in renal fibrosis.

  • Possible immunohistochemical markers (Pancytokeratin, SMA and Vimentin) to identify the process of EMT.

  • Availability of various EMT related anti-fibrotic therapy with emphasis on stem cell therapy.

HIGHLIGHTS

  • Exploring the role of TGFβ and its associate genes in the pathogenetic mechanism of EMT in renal fibrosis.

  • Possible immunohistochemical markers (Pancytokeratin, SMA and Vimentin) to identify the process of EMT.

  • Availability of various EMT related anti-fibrotic therapy with emphasis on stem cell therapy.

INTRODUCTION

Epithelial-mesenchymal transition (EMT) was first described by Elizabeth Hay in 1995 as "epithelial-mesenchymal transformation", which was later modified to "transition" due to the phenomenon of phenotypic plasticity and distinction from neoplastic transformation [11 Hay ED. An overview of epithelio-mesenchymal transformation. Acta Anat (Basel). 1995;154:8-20.]. It is a biological process wherein epithelial cells lose the epithelial markers and gain mesenchymal markers, resulting in a mesenchymal phenotype. These transformed cells have the properties of increased migratory capacity, the potential to invade tissues, marked resistance to apoptosis and enhanced production of extracellular matrix components[22 Kalluri R WAR. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420-8.]. Numerous well-coordinated molecular mechanisms are known regarding the initiation of EMT and its outcome, which includes transcription factor activation, the pronouncement of cell surface receptors, reorganization and expression of cytoskeletal proteins, production of extracellular matrix (ECM) degrading enzymes and alterations in specific microRNAs. In vivo, some of these factors are used as biomarkers to demonstrate the process of EMT [22 Kalluri R WAR. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420-8.]

Classification of epithelial-mesenchymal transition

The exact mechanisms as to how terminally differentiated cells are derived from the germ cell layers i.e. ectoderm, mesoderm and endoderm during embryogenesis and organ development are debatable. However, it is now known that cells within certain tissue compartments appear to be plastic and thus can transform from epithelial to mesenchymal states and vice-versa [22 Kalluri R WAR. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420-8.].

EMTs are divided into three distinct biological subtypes based on the different functional outcomes:

Type 1 EMT

This type of EMT plays a role in implantation and embryonic development where primitive epithelial cells (e.g., cells of paraxial mesoderm) give rise to mesenchymal cells-mesoderm, endoderm, and neural crest cells during organogenesis. An important feature of this type of transformation is its ability to undergo a reverse mesenchymal-epithelial transition (MET) to generate different types of epithelia[33 Acloque H, Adams MS, Fishwick K, Bronner-Fraser M, Nieto MA. Epithelial-mesenchymal transitions: The importance of changing cell state in development and disease. J Clin Invest. 2009;119(6):1438-49.].

Type 2 EMT

The second type of EMT is associated with wound healing. This process commences following trauma and inflammatory injury which leads to the production of fibroblasts as a response to inflammation. This results in functional cells being completely replaced by a fibrotic scar secondary to an unabated inflammatory response [22 Kalluri R WAR. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420-8.].

Type 3 EMT

The third type of EMT plays an important role in invasion and metastasis in cancer progression here, neoplastic cells develop a mesenchymal invasive phenotype that has the propensity to invade and metastasize to various organs [44 Fragiadaki M, Mason RM. Epithelial-mesenchymal transition in renal fibrosis - evidence for and against. Int J Exp Pathol. 2011;92(3):143-50.].

Other cells capable of EMT include podocytes and endothelial cells in the kidney

Podocyte Epithelial-Mesenchymal Transition

Podocytes, one of the components of the filtration barrier in the kidney, are glomerular visceral epithelial cells that allow selective permeability owing to their unique structure and specialized function. Injury to the podocyte is an important cause of several glomerular diseases. One of the responses of podocytes to various injuries is EMT which results in depletion of these cells leading to heavy proteinuria [55 Pasupulati AK, Nishad, R, Nakuluri, K, Motrapu, M. Epithelial-mesenchymal Transition of Glomerular Podocytes: Implications in Proteinuria. MGM J Med Sci. 2017;4(1):26-34.]. The phenotypic switch seen in the podocytes closely resembles type II EMT [66 May CJ, Saleem M, Welsh GI. Podocyte dedifferentiation : a specialized process for a specialized cell. 2014;5(October):1-8.].

In diabetic nephropathy, high glucose medium upregulates the expression of MMP9, alpha-SMA and fibronectin and downregulates podocalyxin in podocytes resulting in podocyte EMT [77 Ling LI, Chen L, Zhang C, Gui S, Zhao H, Li Z. High glucose induces podocyte epithelial ‑ to ‑ mesenchymal transition by demethylation ‑ mediated enhancement of MMP9 expression. 2018;5642-51.]

Endothelial Mesenchymal Transition.

Endothelial cells lining the vasculature of the renal parenchyma has been shown to have the propensity for the phenotypic switch through a process called endothelial-mesenchymal transition [88 Dejana E, Hirschi KK, Simons M. The molecular basis of endothelial cell plasticity. Nat Commun [Internet]. 2017;8:1-11. Available from: http://dx.doi.org/10.1038/ncomms14361
http://dx.doi.org/10.1038/ncomms14361...
]. These cells lose their inherent endothelial characteristics and acquire the properties of fibroblast-like phenotype and gene expression [99 Lovisa S, Fletcher-sananikone E, Sugimoto H, Hensel J, Hertig A, Taduri G, et al. Endothelial-to-mesenchymal transition compromises vascular integrity to induce Myc-mediated metabolic reprogramming in kidney fibrosis. 2021;13(635):1-35.]. This endothelial-mesenchymal transition has been demonstrated as an important pathological process in cardiac fibrosis [1010 Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007;13(8):952-61.], renal fibrosis [1111 Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol. 2008;19(12):2282-7.], carcinoma-associated fibrosis [1212 Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007;67(21):10123-8.] and atherosclerosis [1313 Chen PY, Qin L, Baeyens N, Li G, Afolabi T, Budatha M, et al. Endothelial-to-mesenchymal transition drives atherosclerosis progression. J Clin Invest. 2015;125(12):4514-28.]. Any noxious stimuli to the renal parenchyma cause the endothelial cells to switch to mesenchymal cells as evidenced by the co-expression of CD31 (endothelial) and alpha-SMA and FSP-1 (mesenchymal) immunohistochemical markers in these lesions [99 Lovisa S, Fletcher-sananikone E, Sugimoto H, Hensel J, Hertig A, Taduri G, et al. Endothelial-to-mesenchymal transition compromises vascular integrity to induce Myc-mediated metabolic reprogramming in kidney fibrosis. 2021;13(635):1-35.].

EMT and Biomarkers in Renal Fibrosis

In the normal kidney, renal tubules are lined by low cuboidal epithelial cells and are surrounded by the interstitium, which contains an occasional fibroblast in the quiescent state. These fibroblasts are formed during development with the help of type 1 EMT [1414 Neilson EG. Mechanisms of disease: Fibroblasts - A new look at an old problem. Nat Clin Pract Nephrol. 2006;2(2):101-8.] and are responsible for maintaining the integrity of the interstitium [1515 Qi W, Chen X, Poronnik P, Pollock CA. The renal cortical fibroblast in renal tubulointerstitial fibrosis. 2006;38:1-5.].

Upon exposure to toxins, the tubular epithelial cells (TEC) undergo acute injury leading to death which activates surviving TEC to revert to its near normal structure and function. However, repeated TEC injury or chronic disease leads to prolonged release of stimulatory cytokines from the injured TECs as well as the inflammatory cells in the vicinity leading to the production of activated myofibroblasts in the interstitium which contributes to fibrosis [44 Fragiadaki M, Mason RM. Epithelial-mesenchymal transition in renal fibrosis - evidence for and against. Int J Exp Pathol. 2011;92(3):143-50.]. This process leads to the deposition of fibrotic tissue leading to progressive loss of renal function [1616 Gregorio J Di, Robuffo I, Spalletta S, Giambuzzi G, Iuliis V De, Toniato E, et al. The Epithelial-to-Mesenchymal Transition as a Possible Therapeutic Target in Fibrotic Disorders. 2020;8(December):1-32.]. A wide range of pro-fibrotic cytokines plays an important role in chronic inflammation, which regulates the infiltration of inflammatory cells- neutrophils, macrophages and lymphocytes [1717 Pan B, Liu G, Jiang Z, Zheng D. Regulation of renal fibrosis by macrophage polarization. Cell Physiol Biochem. 2015;35(3):1062-9.].

In chronic kidney disease, the three major signaling pathways which operate in the tubular and podocyte mesenchymal transition includes, TGFβ/Smad, integrin/ ILK, and Wnt/β-catenin signaling pathways [1818 Liu Y. New insights into epithelial-mesenchymal transition in kidney fibrosis. J Am Soc Nephrol. 2010;21(2):212-22.]. Among these pathways, the TGFβ/Smad pathway is considered to be the prototype inducer of the tubular and podocyte mesenchymal transition [1919 Yang J, Liu Y. Dissection of key events in tubular epithelial to myofibroblast transition and its implications in renal interstitial fibrosis. Am J Pathol. 2001;159(4):1465-75. Available from: http://dx.doi.org/10.1016/S0002-9440(10)62533-3
http://dx.doi.org/10.1016/S0002-9440(10)...
]. Renal fibrosis is the consequence of the failed tissue repair following injury by noxious stimuli. This results in the secretion and gradual increase in various cytokines and proinflammatory cells in the tubular and capillary compartment of the renal parenchyma. This adversarial microenvironment makes the tubular epithelial cells to undergo phenotypic switching in order to escape apoptosis [1818 Liu Y. New insights into epithelial-mesenchymal transition in kidney fibrosis. J Am Soc Nephrol. 2010;21(2):212-22.].

There are many factors in the tubular and capillary microenvironment which plays a vital role in promoting and suppressing EMT. The factors that promote EMT are cytokines (IL-1, Oncostatin M), growth factors (TGF-B1, FGF-2, connective tissue growth factor, angiotensin II), proteases (MMP-2, tissue-type plasminogen activator, Plasmin) and various environmental stresses. Suppressors of EMT on the other hand works in conjunction with injurious insults to balance the effect of EMT and they include growth factors (hepatocyte growth factor, bone morphogenic protein-7), nuclear receptor activator (vitamin D) and angiotensin II receptor blocker [1818 Liu Y. New insights into epithelial-mesenchymal transition in kidney fibrosis. J Am Soc Nephrol. 2010;21(2):212-22.].

Studies have shown that experimental obstruction of one ureter followed by extraction of TECs and addition of transforming growth factor-beta-1 (TGF-β1) results in EMT [2020 Liu Y. Epithelial to Mesenchymal Transition in Renal Fibrogenesis: Pathologic Significance, Molecular Mechanism, and Therapeutic Intervention. J Am Soc Nephrol. 2004;15(1):1-12.]. The stages of this transformation result in the loss of epithelial cell adhesion, de-novo synthesis or cytoskeletal re-organization of alpha-smooth muscle actin (αSMA), disruption of the tubular basement membrane and migration of transformed cells into the interstitium [2020 Liu Y. Epithelial to Mesenchymal Transition in Renal Fibrogenesis: Pathologic Significance, Molecular Mechanism, and Therapeutic Intervention. J Am Soc Nephrol. 2004;15(1):1-12.]. TGFβ1 induces the transcriptional repressor gene Snail-1, which further downregulates the expression of E-cadherin in the renal tubular cells which is a cell adhesion molecule in renal tubular cells [1919 Yang J, Liu Y. Dissection of key events in tubular epithelial to myofibroblast transition and its implications in renal interstitial fibrosis. Am J Pathol. 2001;159(4):1465-75. Available from: http://dx.doi.org/10.1016/S0002-9440(10)62533-3
http://dx.doi.org/10.1016/S0002-9440(10)...
]. Other transcriptional factors that play a role in the downregulation of E-cadherin include Twist, Zeb 1, Zeb 2, and Slug [22 Kalluri R WAR. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420-8.]. (Figure 1) Additionally, this causes the tubular epithelial cells to acquire the morphological and functional features of myofibroblasts by de-novo expression of αSMA and re-organization of actin microfilaments [2121 Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, et al. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med. 2016;241(1):1-13.]. This provides plasticity and mobility which are necessary properties in wound healing and fibrosis. These transformed cells express matrix metalloproteinase 9 (MMP 9) which has a major role in the dissociation of epithelial cells in EMT. Additionally, they have the capacity to degrade the basement membrane of the tubule and invade the interstitium, an inherent functional property of the myofibroblasts [2222 Zhao Y, Qiao X, Tan TK, Zhao H, Zhang Y, Liu L, et al. Matrix metalloproteinase 9-dependent Notch signaling contributes to kidney fibrosis through peritubular endothelial-mesenchymal transition. Nephrol Dial Transplant. 2017;32(5):781-91.].

Figure 1
A simplified diagram showing the process of EMT where the transition of polarized epithelia cells transdifferentiates into mobile mesenchymal cells. TGF β induces the SMAD 2/3 which further upregulates the transcriptional repressor gene such as SNAIL/TWIST/SLUG to downregulate E-cadherin via activation of Zeb 1, 2 and also express vimentin and fibronectin in renal tubular cells

In murine models, it has been demonstrated that in the normal kidney, interleukin-15 (IL-15), maintains the homeostasis of the renal epithelial cells through an autocrine loop that protects the tubular epithelial cells from apoptosis and inflammation during nephritis [2323 Shinozaki M, Hirahashi J, Lebedeva T, Liew FY, Salant DJ, Maron R, et al. IL-15, a survival factor for kidney epithelial cells, counteracts apoptosis and inflammation during nephritis. J Clin Invest. 2002;109(7):951-60.]. Devocelle and his colleagues demonstrated a sharp depression of intra-renal IL-15 in different human inflammatory nephropathies resulting in fibrosis [2424 Devocelle A, Lecru L, François H, Desterke C, Gallerne C, Eid P, et al. Inhibition of TGF- β 1 Signaling by IL-15: A Novel Role for IL-15 in the Control of Renal Epithelial-Mesenchymal Transition: IL-15 Counteracts TGF- β 1-Induced EMT in Renal Fibrosis. Int J Cell Biol. 2019.].

In an initial study by Strutz and coauthors, the authors used FSP1/S100A4 to demonstrate EMT. This protein is present on the fibroblast and not in the epithelial cells. In chronic kidney diseases (CKD) with renal fibrosis, FSP1/S100A4 was demonstrated in the interstitium and the tubular epithelial cells [2525 Strutz F, Okada H, Lo CW, Danoff T, Carone RL, Tomaszewski JE, et al. Identification and characterization of a fibroblast marker: FSP1. J Cell Biol. 1995;130(2):393-405.]. Subsequent studies showed that FSP1/S100A4 is not expressed in normal myofibroblasts. However, in diseased states, the expression of this marker is seen in fibroblasts and immune cells. Thus, making it a non-specific marker for myofibroblasts (2626 Inoue T, Plieth D, Venkov CD, Xu C, Neilson EG. Antibodies against macrophages that overlap in specificity with fibroblasts. Kidney Int. 2005;67(6):2488-93.,2727 Le Hir M, Hegyi I, Cueni-Loffing D, Loffing J, Kaissling B. Characterization of renal interstitial fibroblast-specific protein 1/S100A4-positive cells in healthy and inflamed rodent kidneys. Histochem Cell Biol. 2005;123(4-5):335-46.,2828 Rossini M, Cheunsuchon B, Donnert E, Ma LJ, Thomas JW, Neilson EG, et al. Immunolocalization of fibroblast growth factor-1 (FGF-1), its receptor (FGFR-1), and fibroblast-specific protein-1 (FSP-1) in inflammatory renal disease. Kidney Int. 2005;68(6):2621-8.].

A common marker used to demonstrate the EMT process is vimentin. Studies have shown that injured tubular epithelial cells strongly express vimentin however, this is regarded as evidence of regenerating activity rather than proof of EMT [2929 Grone HJ, Weber K, Grone E, Helmchen U, Osborn M. Coexpression of keratin and vimentin in damaged and regenerating tubular epithelia of the kidney. Am J Pathol. 1987;129(1):1-8.,3030 Witzgall R, Brown D, Schwarz C, Bonventre J V. Localization of proliferating cell nuclear antigen, vimentin, c-Fos, and clusterin in the postischemic kidney. Evidence for a heterogenous genetic response among nephron segments, and a large pool of mitotically active and dedifferentiated cells. J Clin Invest. 1994;93(5):2175-88.,3131 Zhu MQ, De Broe ME NE. Vimentin expression and distal tubular damage in the rat kidney. Exp Nephrol. 1996;4(3):172-83.]. Zheng and coauthors confirmed the importance of vimentin expression in the process of EMT as an inducer of allograft kidney failure in chronic allograft nephropathy [3232 Zheng Wang, Alex Divanyan, Frances L. Jourd’heuil, Robert D. Goldman, Karen M. Ridge, David Jourd’heuil RIL-S. Vimentin expression is required for the development of EMT-related renal fibrosis following unilateral ureteral obstruction in mice. Am J Physiol Ren Physiol. 2018;315(4):F769-F780.]. These authors, established the role of vimentin in renal fibrosis via unilateral ureteral obstruction in vimentin knock out mice and compared the amount of fibrosis with the control group. A similar phenomenon was demonstrated in cultured human proximal renal tubular cells after vimentin expression was dimmed using lentivirus-driven inhibition of vimentin and subsequent treatment of the same with TGF-β which initiates the process of EMT. They concluded that inhibition of vimentin halts the process of fibrosis following unilateral ureteral obstruction, possibly by down streaming the signaling pathways [3232 Zheng Wang, Alex Divanyan, Frances L. Jourd’heuil, Robert D. Goldman, Karen M. Ridge, David Jourd’heuil RIL-S. Vimentin expression is required for the development of EMT-related renal fibrosis following unilateral ureteral obstruction in mice. Am J Physiol Ren Physiol. 2018;315(4):F769-F780.].

A survey of the literature showed that α-SMA has also been suggested as a potential marker for EMT. Studies have shown that α-SMA positive cells are seen in the interstitium in injured tubules whereas the normal tubules were negative for this marker [3333 Kimura M, Asano M, Abe K, Miyazaki M, Suzuki T, Hishida A. Role of atrophic changes in proximal tubular cells in the peritubular deposition of type IV collagen in a rat renal ablation model. Nephrol Dial Transplant. 2005;20(8):1559-65.,3434 Loffing J, Loffing-Cueni D, Hegyi I, Kaplan MR, Hebert SC, Le Hir M, et al. Thiazide treatment of rats provokes apoptosis in distal tubule cells. Kidney Int. 1996;50(4):1180-90.].

In a controversial paper by Humphreys BD and coauthors [3535 Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre J V, et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol [Internet]. 2010;176(1):85-97. Available from: http://dx.doi.org/10.2353/ajpath.2010.090517
http://dx.doi.org/10.2353/ajpath.2010.09...
] and Iwano M and coauthors [3636 Iwano M, Okada H, Neilson EG, Iwano M, Plieth D, Danoff TM, et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis Find the latest version : Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110(3):341-50.] regarding the origin of myofibroblasts in kidney fibrosis, the authors used high-resolution markers for renal tubular cells origin linked to nephrogenesis and demonstrated that interstitial cells differentiate into smooth muscle actin positive myofibroblasts during fibrosis, Concerning Humphreys BD and coauthors [3535 Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre J V, et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol [Internet]. 2010;176(1):85-97. Available from: http://dx.doi.org/10.2353/ajpath.2010.090517
http://dx.doi.org/10.2353/ajpath.2010.09...
] and Iwano M and coauthors [3636 Iwano M, Okada H, Neilson EG, Iwano M, Plieth D, Danoff TM, et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis Find the latest version : Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110(3):341-50.], Fragiadaki M and coauthors [44 Fragiadaki M, Mason RM. Epithelial-mesenchymal transition in renal fibrosis - evidence for and against. Int J Exp Pathol. 2011;92(3):143-50.], suggested that to get an accurate picture of EMT it is essential to use two promoters along with different fluorescent proteins to confirm the presence of EMT in fibrosis and hence the original theory of EMT in renal fibrosis should not be rejected.

Thus a variety of immunohistochemistry (IHC) markers were used in the past as surrogates for direct detection of collagen synthesis by immunolabelling [3737 Rastaldi MP, Ferrario F, Giardino L, Dell’antonio G, Grillo C, Grillo P, et al. Epithelial-mesenchymal transition of tubular epithelial cells in human renal biopsies. Kidney Int. 2002;62(1):137-46.]. However, a definite marker for the demonstration of the EMT process is yet to be identified and this requires further research to identify definite markers to confirm the process of EMT.

EMT in Kidney Fibrosis- Clinical Perspectives

Chronic kidney disease (CKD) is characterized by the development of progressive renal tubular interstitial fibrosis, usually as a result of a primary renal disorder [3333 Kimura M, Asano M, Abe K, Miyazaki M, Suzuki T, Hishida A. Role of atrophic changes in proximal tubular cells in the peritubular deposition of type IV collagen in a rat renal ablation model. Nephrol Dial Transplant. 2005;20(8):1559-65.]. This process is irreversible which progressively leads to end-stage renal disease (ESRD). The initial insult is in the glomerulus, which then progresses to the tubulointerstitium, resulting in the replacement of the entire functional unit of the kidney to a fibrous scar. CKD has a high global prevalence which is estimated to be 11 to 13% [3838 Yang CW, Harris DCH, Luyckx VA, Nangaku M, Hou FF, Garcia Garcia G, et al. Global case studies for chronic kidney disease/end-stage kidney disease care. Kidney Int Suppl [Internet]. 2020;10(1):e24-48. Available from: https://doi.org/10.1016/j.kisu.2019.11.010
https://doi.org/10.1016/j.kisu.2019.11.0...
]. In India, the age-adjusted incidence of ESRD due to CKD is 229 per million population with >100,000 new patients entering the renal transplantation program annually [3939 Singh AK, Farag YMK, Mittal B V., Subramanian KK, Reddy SRK, Acharya VN, et al. Epidemiology and risk factors of chronic kidney disease in India - Results from the SEEK (Screening and Early Evaluation of Kidney Disease) study. BMC Nephrol. 2013;14(1):1-10.]. This increasing trend poses a major healthcare problem, especially in resource-limited countries.

Various surrogate markers such as albuminuria, doubling of serum creatinine and the evidence of a 50% reduction in eGFR are used in monitoring the progression of CKD to ESRD. However, the decline in renal function is not linear to the glomerular filtration rate (GFR) or proteinuria. Therefore, the prediction of a rapid or slow progression to chronic disease is completely dependent on either evidence in kidney biopsy or the use of newly developed biomarkers such as kidney injury molecule-1, neutrophil gelatinase-associated protein, apolipoprotein A-IV and soluble urokinase receptor [4040 Jianyong Zhong, Hai-Chun Yang ABF. A perspective on chronic kidney disease progression. Am J Physiol Ren Physiol. 2017;312(3): F375-F384.].

There is limited evidence in support of EMT in vivo in hypertensive nephropathy [4141 Yang F, Huang XR, Chung ACK, Hou CC, Lai KN, Lan HY. Essential role for Smad3 in angiotensin II-induced tubular epithelial-mesenchymal transition. J Pathol. 2010;221(4):390-401.,4242 Seccia T, Caroccia B, Piazza M, Rossi GP. The Key Role of Epithelial to Mesenchymal Transition ( EMT ) in Hypertensive Kidney Disease. 2019;]. In the rat cell culture models, angiotensin II was found to cause the transformation of epithelial cells to elongated, spindle-shaped mesenchymal cells [4343 Carvajal G, Rodríguez-Vita J, Rodrigues-Díez R, Sánchez-López E, Rupérez M, Cartier C, et al. Angiotensin II activates the Smad pathway during epithelial mesenchymal transdifferentiation. Kidney Int. 2008;74(5):585-95.], with loss of E-cadherin and gain of α-SMA in the transformed cells [4444 Burns WC, Thomas MC. Angiotensin II and its role in tubular epithelial to mesenchymal transition associated with chronic kidney disease. Cells Tissues Organs. 2010;193(1-2):74-84.]. This supports the role of angiotensin II as a driving factor in EMT in hypertensive nephropathy.

Histopathological evaluation requires standardization of immunohistochemical markers, which can depict the progression of the disease in a phase-wise manner depending on the initiation or the level of EMT in the renal biopsy during the various stages of CKD. Additionally, interstitial fibrosis and tubular atrophy (IFTA score) is used to quantify renal fibrosis which helps in the management of patients with CKD [4545 Farris AB, Alpers CE. What is the best way to measure renal fibrosis: A pathologist’s perspective. Kidney Int Suppl. 2014;4(1):9-15.].

Rastaldi and coauthors [3737 Rastaldi MP, Ferrario F, Giardino L, Dell’antonio G, Grillo C, Grillo P, et al. Epithelial-mesenchymal transition of tubular epithelial cells in human renal biopsies. Kidney Int. 2002;62(1):137-46.] demonstrated EMT in 133 human renal biopsies from the spectrum of renal diseases with the help of IHC markers (ZO-1, α SMA, CK, and vimentin) on renal tubular epithelial cells. With the progression of the disease, there was a loss of cytokeratin and ZO-1 and gain of α SMA and vimentin thus showing a linear correlation with the severity of the disease. J. Yao and coauthors [4646 Yao J, Ke Z, Wang X, Peng F, Li B, Wu R. Epithelial-mesenchymal transition and apoptosis of renal tubular epithelial cells are associated with disease progression in patients with IgA nephropathy. Mol Med Rep. 2014;10(1):39-44.] studied 74 human renal biopsies in patients with IgA nephropathy and demonstrated EMT by using the IHC markers α- SMA and vimentin. They showed that there was an increased expression of tubular and interstitial α- SMA and vimentin with the severity of the disease. This proves that the process of EMT is responsible for the disease progression leading to CKD. Thus, early detection of EMT and targeted antifibrotic therapy is the key in the management of renal diseases associated with fibrosis.

The Current Trend in EMT Related Anti-Fibrotic Therapy

Numerous treatment modalities have been developed in the recent past to inhibit the progression of fibrosis. The three basic strategies used are a) Inhibition & Prevention of EMT, b) Removal of fibroblastic cells, and c) Re-transdifferentiation of myofibroblasts to epithelial cells [2121 Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, et al. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med. 2016;241(1):1-13.].

a) Inhibition and prevention of EMT

To date, efforts have been made to inhibit EMT with the help of antagonists directed against EMT-inducing cytokines. The current important cytokine in the process of EMT-related tissue fibrosis is TGF-β. TGF- β has a wide range of biological functions that can affect all cellular functions involved in wound healing. Therefore, as a therapeutic agent the level of TGF- β should be adjusted to prevent its effects on other cells [4747 Hawinkels LJAC, Ten Dijke P. Exploring anti-TGF-β therapies in cancer and fibrosis. Growth Factors. 2011;29(4):140-52.].

The targeted therapy is aimed at limiting the TGF-β signaling pathway as well as the reversal of established fibrosis [4848 Chen YL, Zhang X, Bai J, Gai L, Ye XL, Zhang L, et al. Sorafenib ameliorates bleomycin-induced pulmonary fibrosis: Potential roles in the inhibition of epithelial-mesenchymal transition and fibroblast activation. Cell Death Dis. 2013;4(6):1-11.]. Various molecular methods are available to disrupt the signaling pathway of TGF-β with the inhibitor of TGF-β mRNA expression. They include TGF- β1 anti-sense oligodeoxynucleotide and small molecule receptor kinase inhibitor [4949 Connolly EC, Freimuth J, Akhurst RJ. Complexities of TGF-β Targeted Cancer Therapy. Int J Biol Sci. 2012;8(7).]. Another method used is the sequestration of the ligand by soluble receptor ectodomain construct (ligand trap). These ligands occupy the same binding site of TGF-β receptors I and II and compete for receptor binding [4747 Hawinkels LJAC, Ten Dijke P. Exploring anti-TGF-β therapies in cancer and fibrosis. Growth Factors. 2011;29(4):140-52.].

Hepatocyte growth factor (HGF) balances the effect of TGF- β and acts directly without interfering with the signaling pathway. Therefore, it has an antifibrotic property by reducing the levels of TGF- β in the lung [5050 Gazdhar A, Temuri A, Knudsen L, Gugger M, Schmid RA, Ochs M, et al. Targeted gene transfer of hepatocyte growth factor to alveolar type II epithelial cells reduces lung fibrosis in rats. Hum Gene Ther. 2013;24(1):105-16.].

Another chemical compound that can significantly inhibit the effects of TGF- β and reactive oxygen species production is zinc. Zinc inhibits the TGF- β/Smad pathway in the process of EMT in rat peritoneal mesothelial cells [5151 Zhang X, Wang J, Fan Y, Yang L, Wang L, Ma J. Zinc supplementation attenuates high glucose-induced epithelial-to-mesenchymal transition of peritoneal mesothelial cells. Biol Trace Elem Res. 2012;150(1-3):229-35.].

Other processes such as eliminating the local immune cell response or blocking the effects of inflammatory cytokines can effectively inhibit the EMT process [2121 Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, et al. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med. 2016;241(1):1-13.].

In pathological stress, the levels of intra-renal IL-15 drops drastically and protects physiological function and thus leading to the survival of the renal epithelial cells [5252 Eini H, Tejman-Yarden N, Lewis EC, Chaimovitz C, Zlotnik M, Douvdevani A. Association between renal injury and reduced interleukin-15 and interleukin-15 receptor levels in acute kidney injury. J Interf Cytokine Res. 2010;30(1):1-8.]. The effectiveness of recombinant human IL-15 to inhibit TGF- β1-induced type 2 EMT has been demonstrated in a human cell model [2424 Devocelle A, Lecru L, François H, Desterke C, Gallerne C, Eid P, et al. Inhibition of TGF- β 1 Signaling by IL-15: A Novel Role for IL-15 in the Control of Renal Epithelial-Mesenchymal Transition: IL-15 Counteracts TGF- β 1-Induced EMT in Renal Fibrosis. Int J Cell Biol. 2019.].

b) Removal of fibroblasts and/or myofibroblastic cells

Once fibrosis is established, it is practically impossible for the kidney to revert back to a normal function. Hence, novel strategies are aimed at preventing further damage by promoting apoptosis of fibroblast/myofibroblast in the damaged area via various signaling pathways like Rho/Rho-kinase and phosphatidylinositol-3-kinase/Akt signaling pathways. The end products of these pathways i.e. nerve growth factor and basic fibroblast growth factors respectively are used to stall the process of fibrosis [5353 Abe M, Yokoyama Y, Ishikawa O. A possible mechanism of basic fibroblast growth factor-promoted scarless wound healing: The induction of myofibroblast apoptosis. Eur J Dermatology. 2012;22(1):46-53., 5454 Micera A, Puxeddu I, Balzamino BO, Bonini S, Levi-Schaffer F. Chronic Nerve Growth Factor Exposure Increases Apoptosis in a Model of In Vitro Induced Conjunctival Myofibroblasts. PLoS One. 2012;7(10):1-13.].

Carvalho and coauthors [5555 Nunes De Carvalho S, Da Cunha Lira D, Costa Cortez EA, De Andrade DC, Thole AA, Stumbo AC, et al. Bone marrow cell transplantation is associated with fibrogenic cells apoptosis during hepatic regeneration in cholestatic rats. Biochem Cell Biol. 2013;91(2):88-94.] demonstrated a novel method of removal of fibroblast through bone marrow mononuclear cell transplantation in cases of liver fibrosis secondary to bile duct ligation. The plausible pathogenesis includes apoptosis of fibroblasts and regeneration of hepatocytes through possible stimulation of cytokines.

c) Re-transdifferentiation of myofibroblast to epithelial cells

Myofibroblasts are not terminally differentiated cells and can easily revert to their origin in due course of time if the inducing signals are inhibited [2121 Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, et al. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med. 2016;241(1):1-13.]. In an experimental model, TGF- β-induced EMT was reversed in humans and rat alveolar epithelial-like cell lines by using HGF which is an inhibitor of the TGF- β signaling pathway, Smad7 [5656 Shukla MN, Rose JL, Ray R, Lathrop KL, Ray A, Ray P. Hepatocyte growth factor inhibits epithelial to myofibroblast transition in lung cells via Smad7. Am J Respir Cell Mol Biol. 2009;40(6):643-53.]. Chemical compounds such as procyanidins and proanthocyanidins have anti-inflammatory and antioxidant features and are able to reverse the EMT process [5757 Arora P, Ansari S, Nazish I. Study of antiobesity effects of ethanolic and water extracts of grapes seeds. J Complement Integr Med. 2011;8(1):1-14.]. Human amniotic membrane stromal extracts and prostaglandins E2 also induces the trans-differentiation of myofibroblasts into fibroblasts [2121 Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, et al. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med. 2016;241(1):1-13.].

Stem cells have a wide range of paracrine activity while maintaining the original cellular micro-environment. It has the capacity to replenish cells that are lost in tissue fibrosis [2121 Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, et al. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med. 2016;241(1):1-13.]. Akram and coauthors [5858 Akram KM, Samad S, Spiteri MA, Forsyth NR. Mesenchymal stem cells promote alveolar epithelial cell wound repair in vitro through distinct migratory and paracrine mechanisms. Respir Res. 2013;14(1):1-16.] described the clinical application of intravenous and local administration of mesenchymal stem cells in the treatment of tissue fibrosis. Du and coauthors [5959 Du T, Zou X, Cheng J, Wu S, Zhong L, Ju G, et al. Human Wharton’s jelly-derived mesenchymal stromal cells reduce renal fibrosis through induction of native and foreign hepatocyte growth factor synthesis in injured tubular epithelial cells. Stem Cell Res Ther. 2013;4(3):1-13.] demonstrated the use of Wharton's jelly-derived mesenchymal stem cells to delay the renal tubular EMT and thus alleviate renal fibrosis in an experimental model. Recent studies suggest that mesenchymal stem cell-derived from human adipose tissue play an important role in therapy for renal fibrosis. Adipose-derived mesenchymal stem cells (AMSC) through the autocrine and paracrine mediated release of angiogenic cytokines and growth factors improve the endogenous repair mechanism by increasing the peritubular capillary density and relieving tissue hypoxia [6060 Li S, Wang Y, Wang Z, Chen L, Zuo B, Liu C, et al. Enhanced renoprotective effect of GDNF- modified adipose-derived mesenchymal stem cells on renal interstitial fibrosis. 2021;1-17.].

Glial cell line-derived neurotrophic factor (GDNF) is known for its neuroprotective effects in hypoxic-ischemic encephalopathy [6161 Tao XU. Neuroprotective Effects of Electroacupuncture on Hypoxic-Ischemic Encephalopathy in Newborn Rats Are Associated with Increased Expression of GDNF-RET and Protein Kinase B. 2015;(266013).]. Recent studies show that GDNF -/- mutant mice develop renal agenesis and dysgenesis suggesting that they play a critical role in renal development [6262 Cortés D, Carballo-Molina OA, Castellanos-Montiel MJ, Velasco I. The non-survival effects of Glial cell line-derived neurotrophic factor on neural cells. Front Mol Neurosci. 2017;10(August):1-13.]. GDNF-modified AMSC helps in the repair of the injured microcirculation in the kidney by promoting more angiogenesis through the release of growth factors, thus protecting the kidney microenvironment from hypoxic injury [6060 Li S, Wang Y, Wang Z, Chen L, Zuo B, Liu C, et al. Enhanced renoprotective effect of GDNF- modified adipose-derived mesenchymal stem cells on renal interstitial fibrosis. 2021;1-17.].

CONCLUSION

The current knowledge on the role of EMT based on in vitro experiments shows that various cytokines including TGFβ play a major role in renal fibrosis. However, existing immunohistochemical markers have not been able to establish this mechanism. Multiple multidirectional in vitro experiments with in-vivo models are required to understand this process and the quest for appropriate IHC markers to demonstrate EMT in fibrogenesis should continue. This can aid in targeted antifibrotic therapy which reduces the burden of CKD.

Acknowledgement:

The authors are very grateful to Dr Prakash Peralam Yegneswaran, for help in the preparation of the line chart in this review.

  • Funding: This research received no external funding.

REFERENCES

  • 1
    Hay ED. An overview of epithelio-mesenchymal transformation. Acta Anat (Basel). 1995;154:8-20.
  • 2
    Kalluri R WAR. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420-8.
  • 3
    Acloque H, Adams MS, Fishwick K, Bronner-Fraser M, Nieto MA. Epithelial-mesenchymal transitions: The importance of changing cell state in development and disease. J Clin Invest. 2009;119(6):1438-49.
  • 4
    Fragiadaki M, Mason RM. Epithelial-mesenchymal transition in renal fibrosis - evidence for and against. Int J Exp Pathol. 2011;92(3):143-50.
  • 5
    Pasupulati AK, Nishad, R, Nakuluri, K, Motrapu, M. Epithelial-mesenchymal Transition of Glomerular Podocytes: Implications in Proteinuria. MGM J Med Sci. 2017;4(1):26-34.
  • 6
    May CJ, Saleem M, Welsh GI. Podocyte dedifferentiation : a specialized process for a specialized cell. 2014;5(October):1-8.
  • 7
    Ling LI, Chen L, Zhang C, Gui S, Zhao H, Li Z. High glucose induces podocyte epithelial ‑ to ‑ mesenchymal transition by demethylation ‑ mediated enhancement of MMP9 expression. 2018;5642-51.
  • 8
    Dejana E, Hirschi KK, Simons M. The molecular basis of endothelial cell plasticity. Nat Commun [Internet]. 2017;8:1-11. Available from: http://dx.doi.org/10.1038/ncomms14361
    » http://dx.doi.org/10.1038/ncomms14361
  • 9
    Lovisa S, Fletcher-sananikone E, Sugimoto H, Hensel J, Hertig A, Taduri G, et al. Endothelial-to-mesenchymal transition compromises vascular integrity to induce Myc-mediated metabolic reprogramming in kidney fibrosis. 2021;13(635):1-35.
  • 10
    Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007;13(8):952-61.
  • 11
    Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol. 2008;19(12):2282-7.
  • 12
    Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007;67(21):10123-8.
  • 13
    Chen PY, Qin L, Baeyens N, Li G, Afolabi T, Budatha M, et al. Endothelial-to-mesenchymal transition drives atherosclerosis progression. J Clin Invest. 2015;125(12):4514-28.
  • 14
    Neilson EG. Mechanisms of disease: Fibroblasts - A new look at an old problem. Nat Clin Pract Nephrol. 2006;2(2):101-8.
  • 15
    Qi W, Chen X, Poronnik P, Pollock CA. The renal cortical fibroblast in renal tubulointerstitial fibrosis. 2006;38:1-5.
  • 16
    Gregorio J Di, Robuffo I, Spalletta S, Giambuzzi G, Iuliis V De, Toniato E, et al. The Epithelial-to-Mesenchymal Transition as a Possible Therapeutic Target in Fibrotic Disorders. 2020;8(December):1-32.
  • 17
    Pan B, Liu G, Jiang Z, Zheng D. Regulation of renal fibrosis by macrophage polarization. Cell Physiol Biochem. 2015;35(3):1062-9.
  • 18
    Liu Y. New insights into epithelial-mesenchymal transition in kidney fibrosis. J Am Soc Nephrol. 2010;21(2):212-22.
  • 19
    Yang J, Liu Y. Dissection of key events in tubular epithelial to myofibroblast transition and its implications in renal interstitial fibrosis. Am J Pathol. 2001;159(4):1465-75. Available from: http://dx.doi.org/10.1016/S0002-9440(10)62533-3
    » http://dx.doi.org/10.1016/S0002-9440(10)62533-3
  • 20
    Liu Y. Epithelial to Mesenchymal Transition in Renal Fibrogenesis: Pathologic Significance, Molecular Mechanism, and Therapeutic Intervention. J Am Soc Nephrol. 2004;15(1):1-12.
  • 21
    Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, et al. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med. 2016;241(1):1-13.
  • 22
    Zhao Y, Qiao X, Tan TK, Zhao H, Zhang Y, Liu L, et al. Matrix metalloproteinase 9-dependent Notch signaling contributes to kidney fibrosis through peritubular endothelial-mesenchymal transition. Nephrol Dial Transplant. 2017;32(5):781-91.
  • 23
    Shinozaki M, Hirahashi J, Lebedeva T, Liew FY, Salant DJ, Maron R, et al. IL-15, a survival factor for kidney epithelial cells, counteracts apoptosis and inflammation during nephritis. J Clin Invest. 2002;109(7):951-60.
  • 24
    Devocelle A, Lecru L, François H, Desterke C, Gallerne C, Eid P, et al. Inhibition of TGF- β 1 Signaling by IL-15: A Novel Role for IL-15 in the Control of Renal Epithelial-Mesenchymal Transition: IL-15 Counteracts TGF- β 1-Induced EMT in Renal Fibrosis. Int J Cell Biol. 2019.
  • 25
    Strutz F, Okada H, Lo CW, Danoff T, Carone RL, Tomaszewski JE, et al. Identification and characterization of a fibroblast marker: FSP1. J Cell Biol. 1995;130(2):393-405.
  • 26
    Inoue T, Plieth D, Venkov CD, Xu C, Neilson EG. Antibodies against macrophages that overlap in specificity with fibroblasts. Kidney Int. 2005;67(6):2488-93.
  • 27
    Le Hir M, Hegyi I, Cueni-Loffing D, Loffing J, Kaissling B. Characterization of renal interstitial fibroblast-specific protein 1/S100A4-positive cells in healthy and inflamed rodent kidneys. Histochem Cell Biol. 2005;123(4-5):335-46.
  • 28
    Rossini M, Cheunsuchon B, Donnert E, Ma LJ, Thomas JW, Neilson EG, et al. Immunolocalization of fibroblast growth factor-1 (FGF-1), its receptor (FGFR-1), and fibroblast-specific protein-1 (FSP-1) in inflammatory renal disease. Kidney Int. 2005;68(6):2621-8.
  • 29
    Grone HJ, Weber K, Grone E, Helmchen U, Osborn M. Coexpression of keratin and vimentin in damaged and regenerating tubular epithelia of the kidney. Am J Pathol. 1987;129(1):1-8.
  • 30
    Witzgall R, Brown D, Schwarz C, Bonventre J V. Localization of proliferating cell nuclear antigen, vimentin, c-Fos, and clusterin in the postischemic kidney. Evidence for a heterogenous genetic response among nephron segments, and a large pool of mitotically active and dedifferentiated cells. J Clin Invest. 1994;93(5):2175-88.
  • 31
    Zhu MQ, De Broe ME NE. Vimentin expression and distal tubular damage in the rat kidney. Exp Nephrol. 1996;4(3):172-83.
  • 32
    Zheng Wang, Alex Divanyan, Frances L. Jourd’heuil, Robert D. Goldman, Karen M. Ridge, David Jourd’heuil RIL-S. Vimentin expression is required for the development of EMT-related renal fibrosis following unilateral ureteral obstruction in mice. Am J Physiol Ren Physiol. 2018;315(4):F769-F780.
  • 33
    Kimura M, Asano M, Abe K, Miyazaki M, Suzuki T, Hishida A. Role of atrophic changes in proximal tubular cells in the peritubular deposition of type IV collagen in a rat renal ablation model. Nephrol Dial Transplant. 2005;20(8):1559-65.
  • 34
    Loffing J, Loffing-Cueni D, Hegyi I, Kaplan MR, Hebert SC, Le Hir M, et al. Thiazide treatment of rats provokes apoptosis in distal tubule cells. Kidney Int. 1996;50(4):1180-90.
  • 35
    Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre J V, et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol [Internet]. 2010;176(1):85-97. Available from: http://dx.doi.org/10.2353/ajpath.2010.090517
    » http://dx.doi.org/10.2353/ajpath.2010.090517
  • 36
    Iwano M, Okada H, Neilson EG, Iwano M, Plieth D, Danoff TM, et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis Find the latest version : Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110(3):341-50.
  • 37
    Rastaldi MP, Ferrario F, Giardino L, Dell’antonio G, Grillo C, Grillo P, et al. Epithelial-mesenchymal transition of tubular epithelial cells in human renal biopsies. Kidney Int. 2002;62(1):137-46.
  • 38
    Yang CW, Harris DCH, Luyckx VA, Nangaku M, Hou FF, Garcia Garcia G, et al. Global case studies for chronic kidney disease/end-stage kidney disease care. Kidney Int Suppl [Internet]. 2020;10(1):e24-48. Available from: https://doi.org/10.1016/j.kisu.2019.11.010
    » https://doi.org/10.1016/j.kisu.2019.11.010
  • 39
    Singh AK, Farag YMK, Mittal B V., Subramanian KK, Reddy SRK, Acharya VN, et al. Epidemiology and risk factors of chronic kidney disease in India - Results from the SEEK (Screening and Early Evaluation of Kidney Disease) study. BMC Nephrol. 2013;14(1):1-10.
  • 40
    Jianyong Zhong, Hai-Chun Yang ABF. A perspective on chronic kidney disease progression. Am J Physiol Ren Physiol. 2017;312(3): F375-F384.
  • 41
    Yang F, Huang XR, Chung ACK, Hou CC, Lai KN, Lan HY. Essential role for Smad3 in angiotensin II-induced tubular epithelial-mesenchymal transition. J Pathol. 2010;221(4):390-401.
  • 42
    Seccia T, Caroccia B, Piazza M, Rossi GP. The Key Role of Epithelial to Mesenchymal Transition ( EMT ) in Hypertensive Kidney Disease. 2019;
  • 43
    Carvajal G, Rodríguez-Vita J, Rodrigues-Díez R, Sánchez-López E, Rupérez M, Cartier C, et al. Angiotensin II activates the Smad pathway during epithelial mesenchymal transdifferentiation. Kidney Int. 2008;74(5):585-95.
  • 44
    Burns WC, Thomas MC. Angiotensin II and its role in tubular epithelial to mesenchymal transition associated with chronic kidney disease. Cells Tissues Organs. 2010;193(1-2):74-84.
  • 45
    Farris AB, Alpers CE. What is the best way to measure renal fibrosis: A pathologist’s perspective. Kidney Int Suppl. 2014;4(1):9-15.
  • 46
    Yao J, Ke Z, Wang X, Peng F, Li B, Wu R. Epithelial-mesenchymal transition and apoptosis of renal tubular epithelial cells are associated with disease progression in patients with IgA nephropathy. Mol Med Rep. 2014;10(1):39-44.
  • 47
    Hawinkels LJAC, Ten Dijke P. Exploring anti-TGF-β therapies in cancer and fibrosis. Growth Factors. 2011;29(4):140-52.
  • 48
    Chen YL, Zhang X, Bai J, Gai L, Ye XL, Zhang L, et al. Sorafenib ameliorates bleomycin-induced pulmonary fibrosis: Potential roles in the inhibition of epithelial-mesenchymal transition and fibroblast activation. Cell Death Dis. 2013;4(6):1-11.
  • 49
    Connolly EC, Freimuth J, Akhurst RJ. Complexities of TGF-β Targeted Cancer Therapy. Int J Biol Sci. 2012;8(7).
  • 50
    Gazdhar A, Temuri A, Knudsen L, Gugger M, Schmid RA, Ochs M, et al. Targeted gene transfer of hepatocyte growth factor to alveolar type II epithelial cells reduces lung fibrosis in rats. Hum Gene Ther. 2013;24(1):105-16.
  • 51
    Zhang X, Wang J, Fan Y, Yang L, Wang L, Ma J. Zinc supplementation attenuates high glucose-induced epithelial-to-mesenchymal transition of peritoneal mesothelial cells. Biol Trace Elem Res. 2012;150(1-3):229-35.
  • 52
    Eini H, Tejman-Yarden N, Lewis EC, Chaimovitz C, Zlotnik M, Douvdevani A. Association between renal injury and reduced interleukin-15 and interleukin-15 receptor levels in acute kidney injury. J Interf Cytokine Res. 2010;30(1):1-8.
  • 53
    Abe M, Yokoyama Y, Ishikawa O. A possible mechanism of basic fibroblast growth factor-promoted scarless wound healing: The induction of myofibroblast apoptosis. Eur J Dermatology. 2012;22(1):46-53.
  • 54
    Micera A, Puxeddu I, Balzamino BO, Bonini S, Levi-Schaffer F. Chronic Nerve Growth Factor Exposure Increases Apoptosis in a Model of In Vitro Induced Conjunctival Myofibroblasts. PLoS One. 2012;7(10):1-13.
  • 55
    Nunes De Carvalho S, Da Cunha Lira D, Costa Cortez EA, De Andrade DC, Thole AA, Stumbo AC, et al. Bone marrow cell transplantation is associated with fibrogenic cells apoptosis during hepatic regeneration in cholestatic rats. Biochem Cell Biol. 2013;91(2):88-94.
  • 56
    Shukla MN, Rose JL, Ray R, Lathrop KL, Ray A, Ray P. Hepatocyte growth factor inhibits epithelial to myofibroblast transition in lung cells via Smad7. Am J Respir Cell Mol Biol. 2009;40(6):643-53.
  • 57
    Arora P, Ansari S, Nazish I. Study of antiobesity effects of ethanolic and water extracts of grapes seeds. J Complement Integr Med. 2011;8(1):1-14.
  • 58
    Akram KM, Samad S, Spiteri MA, Forsyth NR. Mesenchymal stem cells promote alveolar epithelial cell wound repair in vitro through distinct migratory and paracrine mechanisms. Respir Res. 2013;14(1):1-16.
  • 59
    Du T, Zou X, Cheng J, Wu S, Zhong L, Ju G, et al. Human Wharton’s jelly-derived mesenchymal stromal cells reduce renal fibrosis through induction of native and foreign hepatocyte growth factor synthesis in injured tubular epithelial cells. Stem Cell Res Ther. 2013;4(3):1-13.
  • 60
    Li S, Wang Y, Wang Z, Chen L, Zuo B, Liu C, et al. Enhanced renoprotective effect of GDNF- modified adipose-derived mesenchymal stem cells on renal interstitial fibrosis. 2021;1-17.
  • 61
    Tao XU. Neuroprotective Effects of Electroacupuncture on Hypoxic-Ischemic Encephalopathy in Newborn Rats Are Associated with Increased Expression of GDNF-RET and Protein Kinase B. 2015;(266013).
  • 62
    Cortés D, Carballo-Molina OA, Castellanos-Montiel MJ, Velasco I. The non-survival effects of Glial cell line-derived neurotrophic factor on neural cells. Front Mol Neurosci. 2017;10(August):1-13.

Edited by

Editor-in-Chief: Alexandre Rasi Aoki
Associate Editor: Sinvaldo Baglie

Publication Dates

  • Publication in this collection
    08 Aug 2022
  • Date of issue
    2022

History

  • Received
    22 Apr 2021
  • Accepted
    11 Feb 2022
Instituto de Tecnologia do Paraná - Tecpar Rua Prof. Algacyr Munhoz Mader, 3775 - CIC, 81350-010 Curitiba PR Brazil, Tel.: +55 41 3316-3052/3054, Fax: +55 41 3346-2872 - Curitiba - PR - Brazil
E-mail: babt@tecpar.br