Endocrinol Metab.  2022 Feb;37(1):96-111. 10.3803/EnM.2021.1305.

EW-7197 Attenuates the Progression of Diabetic Nephropathy in db/db Mice through Suppression of Fibrogenesis and Inflammation

Affiliations
  • 1Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
  • 2Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
  • 3Cardiovascular Research Group, Khon Kaen University, Khon Kaen, Thailand
  • 4Department of Internal Medicine and Institute of Tissue Regeneration, Soonchunhyang University College of Medicine, Cheonan, Korea
  • 5Institution of Genetic Cohort, Yonsei University Wonju College of Medicine, Wonju, Korea
  • 6Department of Pharmacy, Ewha Womans University College of Pharmacy, Seoul, Korea

Abstract

Background
Diabetic nephropathy (DN) is characterized by albuminuria and accumulation of extracellular matrix (ECM) in kidney. Transforming growth factor-β (TGF-β) plays a central role in promoting ECM accumulation. We aimed to examine the effects of EW-7197, an inhibitor of TGF-β type 1 receptor kinase (ALK5), in retarding the progression of DN, both in vivo, using a diabetic mouse model (db/db mice), and in vitro, in podocytes and mesangial cells.
Methods
In vivo study: 8-week-old db/db mice were orally administered EW-7197 at a dose of 5 or 20 mg/kg/day for 10 weeks. Metabolic parameters and renal function were monitored. Glomerular histomorphology and renal protein expression were evaluated by histochemical staining and Western blot analyses, respectively. In vitro study: DN was induced by high glucose (30 mM) in podocytes and TGF-β (2 ng/mL) in mesangial cells. Cells were treated with EW-7197 (500 nM) for 24 hours and the mechanism associated with the attenuation of DN was investigated.
Results
Enhanced albuminuria and glomerular morphohistological changes were observed in db/db compared to that of the nondiabetic (db/m) mice. These alterations were associated with the activation of the TGF-β signaling pathway. Treatment with EW-7197 significantly inhibited TGF-β signaling, inflammation, apoptosis, reactive oxygen species, and endoplasmic reticulum stress in diabetic mice and renal cells.
Conclusion
EW-7197 exhibits renoprotective effect in DN. EW-7197 alleviates renal fibrosis and inflammation in diabetes by inhibiting downstream TGF-β signaling, thereby retarding the progression of DN. Our study supports EW-7197 as a therapeutically beneficial compound to treat DN.

Keyword

Diabetic nephropathies; Glomerular mesangial cells; Podocytes; Transforming growth factor beta; Activin receptor-like kinase 5

Figure

  • Fig. 1 EW-7197 has no effect on insulin tolerance but in kidney function. (A) Bodyweight was monitored throughout experimental period. (B) Intraperitoneal insulin tolerance test were performed after 10 weeks of EW-7197 treatment. (C) Urinary albumin/creatinine (ACR) ratios were measured 24-hour urine collection in metabolic cage. (D) Kidney weight/body weight in all experimental groups. Each group (n=6–8/group). aP<0.05 vs. db/m controls; bP<0.05 vs. db/db controls; cP<0.05 vs. db/db+EW5 mg/kg/day.

  • Fig. 2 EW-7197 reduces glomerular hypertrophy and fibrosis in db/db mice. (A, B) Glomerular hypertrophy was determined in hematoxylin and eosin (H&E) and periodic acid Schiff (PAS) stained glomeruli from diabetic mice. (F) Changes in glomerular area, glomerular volume, Bowman’s space area, (G) percentage of mesangial matrix and mesangial expansion score. (C, D, H) Sirius red, trichrome staining were performed to check the degree of fibrosis of the glomerular (original magnification, 400×). (E) Ultrastructure of glomerular basement membrane (GBM). The GBM thickness, filtration silt pores, and foot process width of podocytes (I) were analyzed by transmission electron microscopy (TEM) at 15000-X magnification. An arrowhead indicates filtration slit pore; a up-down arrow shows GBM thickness (n=2/group). aP<0.05 vs. db/m controls; bP<0.05 vs. db/db controls; cP<0.05 vs. db/db+EW5 mg/kg/day.

  • Fig. 3 EW-7197 protects against nephrin loss from diabetic glomeruli and podocytes. (A) Immunohistochemistry of renal nephrin, (B) the percentage of positively stained area. Original magnification is 400× (scale bar=50 μm). (C, D) By Western blot protein expression of nephrin were analyzed from diabetic mice (n=4–5/group). (E, F) After treatment with high glucose (HG, 30 mM) and EW (500 nM) in podocytes, the protein expression of synaptopodin and nephrin was confirmed by Western blot. (G, H) F-actin stained with fluorescein isothiocyanate (FITC)-phalloidin and positive stained area. aP<0.05 vs. db/m controls; bP<0.05 vs. db/db controls; cP<0.05 vs. db/db+EW5 mg/kg/day; dP<0.05 vs. controls; eP<0.05 vs. high glucose.

  • Fig. 4 EW-7197 down-regulates the expression on fibrogenic mediators in diabetic mice. (A) Immunohistochemistry of renal collagen IV and fibronectin, (B) positively stained area. Original magnification is 400× (scale bar=50 μm). (C, D) By Western blot of renal cortex protein expression of col IV and fibronectin. (E, F) The expression of Smad2/3, p-Smad 2/3, vascular endothelial growth factor (VEGF), and connective tissue growth factor (CTGF) as proteins related to fibrosis was confirmed, respectively from diabetic mice (n=4–5/group). aP<0.05 vs. db/m controls; bP<0.05 vs. db/db controls; cP<0.05 vs. db/db+EW5 mg/kg/day.

  • Fig. 5 EW-7197 down-regulates the expression of fibrosis markers in podocyte and mesangial cell lines. (A, B) Podocyte fibrosis marker; fibronectin, transforming growth factor-β1 (TGF-β1), and vascular endothelial growth factor (VEGF) was detected by Western blots and quantified values. (C, D) Changes in extracellular matrix (ECM) through fibronectin, α-smooth muscle actin (α-SMA), and collagen type IV were detected by mesangial cell Western blot. (E, F) Immunofluorescence of fibronectin and collagen type IV and positive stained area. MES-13, mouse mesangial cells. aP<0.05 vs. controls; bP<0.05 vs. high glucose; cP<0.05 vs. controls; dP<0.05 vs. TGF-β.

  • Fig. 6 EW-7197 inhibits the inflammatory response and prevents cell death. (A, B) Protein expression in the renal cortex of diabetic mice, a marker related to the inflammatory response; phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells (p-NF-κB), NF-κB, monocyte chemoattractant protein-1 (MCP-1), and intercellular adhesion molecule 1 (ICAM-1). (C, D) Expression of proteins such as, Bcl2 Associated X (Bax) and caspase-3 were investigated using Western blot and fold change relative to control. Anti-apoptotic B-cell lymphoma/leukemia-2 (Bcl2) an apoptosis-related marker, in podocytes. (E, F, G, H) Expression of apoptosis-related proteins and immunofluorescence staining in mesangial cells. MES-13, mouse mesangial cells. aP<0.05 vs. db/m controls; bP<0.05 vs. db/db controls; cP<0.05 vs. controls; dP<0.05 vs. high glucose; eP<0.05 vs. controls; fP<0.05 vs. TGF-β.

  • Fig. 7 EW-7197 ameliorates renal reactive oxygen species (ROS) and endoplasmic reticulum (ER) stress. (A, B) Expression of ROS related proteins such as NAD(P)H oxidase 4 (NOX4) and ER stress markers; nuclear factor erythroid 2-related factor (Nrf2) and, transcription factor 6 alpha (ATF6α) in podocytes. (C, D) ROS measurements in podocytes with CM-H2DCF-DA. (E, F) Expression of NOX4 and ER stress-related proteins in mesangial cells. (G, H) ROS generation in mesangial cells by immunofluorescence staining with NOX4 and 8-hydroxydeoxyguanosine (8-OHdG). (I, J) The expression of Nrf2, heme oxygenase-1 (HO-1), and Lamin B was confirmed by extracting the protein from the nucleus. HG, high glucose; MES-13, mouse mesangial cells; TGF-β, transforming growth factor-β. aP<0.05 vs. controls; bP<0.05 vs. high glucose; cP<0.05 vs. controls; dP<0.05 vs. TGF-β.


Cited by  1 articles

Beneficial Effects of a Curcumin Derivative and Transforming Growth Factor-β Receptor I Inhibitor Combination on Nonalcoholic Steatohepatitis
Kyung Bong Ha, Eun Soo Lee, Na Won Park, Su Ho Jo, Soyeon Shim, Dae-Kee Kim, Chan Mug Ahn, Choon Hee Chung
Diabetes Metab J. 2023;47(4):500-513.    doi: 10.4093/dmj.2022.0110.


Reference

1. Elmarakby AA, Sullivan JC. Relationship between oxidative stress and inflammatory cytokines in diabetic nephropathy. Cardiovasc Ther. 2012; 30:49–59.
Article
2. Susztak K, Raff AC, Schiffer M, Bottinger EP. Glucose-induced reactive oxygen species cause apoptosis of podocytes and podocyte depletion at the onset of diabetic nephropathy. Diabetes. 2006; 55:225–33.
Article
3. He T, Xiong J, Nie L, Yu Y, Guan X, Xu X, et al. Resveratrol inhibits renal interstitial fibrosis in diabetic nephropathy by regulating AMPK/NOX4/ROS pathway. J Mol Med (Berl). 2016; 94:1359–71.
Article
4. Sedeek M, Callera G, Montezano A, Gutsol A, Heitz F, Szyndralewiez C, et al. Critical role of Nox4-based NADPH oxidase in glucose-induced oxidative stress in the kidney: implications in type 2 diabetic nephropathy. Am J Physiol Renal Physiol. 2010; 299:F1348–58.
Article
5. Jerums G, Panagiotopoulos S, Premaratne E, MacIsaac RJ. Integrating albuminuria and GFR in the assessment of diabetic nephropathy. Nat Rev Nephrol. 2009; 5:397–406.
Article
6. Maezawa Y, Takemoto M, Yokote K. Cell biology of diabetic nephropathy: roles of endothelial cells, tubulointerstitial cells and podocytes. J Diabetes Investig. 2015; 6:3–15.
Article
7. Li JJ, Kwak SJ, Jung DS, Kim JJ, Yoo TH, Ryu DR, et al. Podocyte biology in diabetic nephropathy. Kidney Int Suppl. 2007; 106:S36–42.
Article
8. Schena FP, Gesualdo L. Pathogenetic mechanisms of diabetic nephropathy. J Am Soc Nephrol. 2005; 16(Suppl 1):S30–3.
Article
9. Kaneto H, Katakami N, Kawamori D, Miyatsuka T, Sakamoto K, Matsuoka TA, et al. Involvement of oxidative stress in the pathogenesis of diabetes. Antioxid Redox Signal. 2007; 9:355–66.
Article
10. Bucala R, Vlassara H. Advanced glycosylation endproducts in diabetic renal disease: clinical measurement, pathophysiological significance, and prospects for pharmacological inhibition. Blood Purif. 1995; 13:160–70.
Article
11. Wolf G, Ziyadeh FN. Cellular and molecular mechanisms of proteinuria in diabetic nephropathy. Nephron Physiol. 2007; 106:p26–31.
Article
12. Inoguchi T, Sonta T, Tsubouchi H, Etoh T, Kakimoto M, Sonoda N, et al. Protein kinase C-dependent increase in reactive oxygen species (ROS) production in vascular tissues of diabetes: role of vascular NAD(P)H oxidase. J Am Soc Nephrol. 2003; 14(8 Suppl 3):S227–32.
Article
13. Haneda M, Kikkawa R, Sugimoto T, Koya D, Araki S, Togawa M, et al. Abnormalities in protein kinase C and MAP kinase cascade in mesangial cells cultured under high glucose conditions. J Diabetes Complications. 1995; 9:246–8.
Article
14. Londero TM, Giaretta LS, Farenzena LP, Manfro RC, Canani LH, Lavinsky D, et al. Microvascular complications of posttransplant diabetes mellitus in kidney transplant recipients: a longitudinal study. J Clin Endocrinol Metab. 2019; 104:557–67.
Article
15. Feng D, Ngov C, Henley N, Boufaied N, Gerarduzzi C. Characterization of matricellular protein expression signatures in mechanistically diverse mouse models of kidney injury. Sci Rep. 2019; 9:16736.
Article
16. Vallon V, Richter K, Blantz RC, Thomson S, Osswald H. Glomerular hyperfiltration in experimental diabetes mellitus: potential role of tubular reabsorption. J Am Soc Nephrol. 1999; 10:2569–76.
17. Yamamoto T, Nakamura T, Noble NA, Ruoslahti E, Border WA. Expression of transforming growth factor beta is elevated in human and experimental diabetic nephropathy. Proc Natl Acad Sci U S A. 1993; 90:1814–8.
Article
18. Voelker J, Berg PH, Sheetz M, Duffin K, Shen T, Moser B, et al. Anti-TGF-β1 antibody therapy in patients with diabetic nephropathy. J Am Soc Nephrol. 2017; 28:953–62.
19. Zhu Y, Usui HK, Sharma K. Regulation of transforming growth factor beta in diabetic nephropathy: implications for treatment. Semin Nephrol. 2007; 27:153–60.
20. Whiteside CI, Dlugosz JA. Mesangial cell protein kinase C isozyme activation in the diabetic milieu. Am J Physiol Renal Physiol. 2002; 282:F975–80.
Article
21. Lee TS, Saltsman KA, Ohashi H, King GL. Activation of protein kinase C by elevation of glucose concentration: proposal for a mechanism in the development of diabetic vascular complications. Proc Natl Acad Sci U S A. 1989; 86:5141–5.
Article
22. Hong YA, Lim JH, Kim MY, Kim TW, Kim Y, Yang KS, et al. Fenofibrate improves renal lipotoxicity through activation of AMPK-PGC-1α in db/db mice. PLoS One. 2014; 9:e96147.
Article
23. Juarez P, Vilchis-Landeros MM, Ponce-Coria J, Mendoza V, Hernandez-Pando R, Bobadilla NA, et al. Soluble betaglycan reduces renal damage progression in db/db mice. Am J Physiol Renal Physiol. 2007; 292:F321–9.
24. Park SA, Kim MJ, Park SY, Kim JS, Lee SJ, Woo HA, et al. EW-7197 inhibits hepatic, renal, and pulmonary fibrosis by blocking TGF-β/Smad and ROS signaling. Cell Mol Life Sci. 2015; 72:2023–39.
Article
25. Kim MJ, Park SA, Kim CH, Park SY, Kim JS, Kim DK, et al. TGF-β type I receptor kinase inhibitor EW-7197 suppresses cholestatic liver fibrosis by inhibiting HIF1α-induced epithelial mesenchymal transition. Cell Physiol Biochem. 2016; 38:571–88.
Article
26. Jin CH, Krishnaiah M, Sreenu D, Subrahmanyam VB, Rao KS, Lee HJ, et al. Discovery of N-((4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-yl)methyl)-2-fluoroaniline (EW-7197): a highly potent, selective, and orally bioavailable inhibitor of TGF-β type I receptor kinase as cancer immunotherapeutic/antifibrotic agent. J Med Chem. 2014; 57:4213–38.
27. Kim SY, Park S, Lee SW, Lee JH, Lee ES, Kim M, et al. RIPK3 contributes to Lyso-Gb3-induced podocyte death. Cells. 2021; 10:245.
Article
28. Seok SJ, Lee ES, Kim GT, Hyun M, Lee JH, Chen S, et al. Blockade of CCL2/CCR2 signalling ameliorates diabetic nephropathy in db/db mice. Nephrol Dial Transplant. 2013; 28:1700–10.
Article
29. Tesch GH, Lim AK. Recent insights into diabetic renal injury from the db/db mouse model of type 2 diabetic nephropathy. Am J Physiol Renal Physiol. 2011; 300:F301–10.
30. Cortes P, Mendez M, Riser BL, Guerin CJ, Rodriguez-Barbero A, Hassett C, et al. F-actin fiber distribution in glomerular cells: structural and functional implications. Kidney Int. 2000; 58:2452–61.
Article
31. Michael AF. The glomerular mesangium. Contrib Nephrol. 1984; 40:7–16.
Article
32. Ravindran S, Pasha M, Agouni A, Munusamy S. Microparticles as potential mediators of high glucose-induced renal cell injury. Biomolecules. 2019; 9:348.
Article
33. Mima A. Inflammation and oxidative stress in diabetic nephropathy: new insights on its inhibition as new therapeutic targets. J Diabetes Res. 2013; 2013:248563.
Article
34. Biernacka A, Dobaczewski M, Frangogiannis NG. TGF-β signaling in fibrosis. Growth Factors. 2011; 29:196–202.
Article
35. Rosen P, Nawroth PP, King G, Moller W, Tritschler HJ, Packer L. The role of oxidative stress in the onset and progression of diabetes and its complications: a summary of a Congress Series sponsored by UNESCO-MCBN, the American Diabetes Association and the German Diabetes Society. Diabetes Metab Res Rev. 2001; 17:189–212.
36. Smith KA, Zhou B, Avdulov S, Benyumov A, Peterson M, Liu Y, et al. Transforming growth factor-β1 induced epithelial mesenchymal transition is blocked by a chemical antagonist of translation factor eIF4E. Sci Rep. 2015; 5:18233.
Article
37. Zeisberg M, Neilson EG. Biomarkers for epithelial-mesenchymal transitions. J Clin Invest. 2009; 119:1429–37.
Article
38. Balasubramanyam M, Lenin R, Monickaraj F. Endoplasmic reticulum stress in diabetes: new insights of clinical relevance. Indian J Clin Biochem. 2010; 25:111–8.
Article
39. Chiang CK, Hsu SP, Wu CT, Huang JW, Cheng HT, Chang YW, et al. Endoplasmic reticulum stress implicated in the development of renal fibrosis. Mol Med. 2011; 17:1295–305.
Article
40. Moon JA, Kim HT, Cho IS, Sheen YY, Kim DK. IN-1130, a novel transforming growth factor-beta type I receptor kinase (ALK5) inhibitor, suppresses renal fibrosis in obstructive nephropathy. Kidney Int. 2006; 70:1234–43.
41. Park CY, Son JY, Jin CH, Nam JS, Kim DK, Sheen YY. EW-7195, a novel inhibitor of ALK5 kinase inhibits EMT and breast cancer metastasis to lung. Eur J Cancer. 2011; 47:2642–53.
Article
42. Sharma K, McCue P, Dunn SR. Diabetic kidney disease in the db/db mouse. Am J Physiol Renal Physiol. 2003; 284:F1138–44.
43. Marques C, Mega C, Goncalves A, Rodrigues-Santos P, Teixeira-Lemos E, Teixeira F, et al. Sitagliptin prevents inflammation and apoptotic cell death in the kidney of type 2 diabetic animals. Mediators Inflamm. 2014; 2014:538737.
Article
44. Meng XM, Tang PM, Li J, Lan HY. TGF-β/Smad signaling in renal fibrosis. Front Physiol. 2015; 6:82.
Article
45. Gill PS, Wilcox CS. NADPH oxidases in the kidney. Antioxid Redox Signal. 2006; 8:1597–607.
Article
46. Kaspar JW, Niture SK, Jaiswal AK. Nrf2:INrf2 (Keap1) signaling in oxidative stress. Free Radic Biol Med. 2009; 47:1304–9.
Article
47. Katsuoka F, Motohashi H, Ishii T, Aburatani H, Engel JD, Yamamoto M. Genetic evidence that small maf proteins are essential for the activation of antioxidant response element-dependent genes. Mol Cell Biol. 2005; 25:8044–51.
Article
48. Michaeloudes C, Chang PJ, Petrou M, Chung KF. Transforming growth factor-β and nuclear factor E2–related factor 2 regulate antioxidant responses in airway smooth muscle cells: role in asthma. Am J Respir Crit Care Med. 2011; 184:894–903.
Article
49. Cao Y, Hao Y, Li H, Liu Q, Gao F, Liu W, et al. Role of endoplasmic reticulum stress in apoptosis of differentiated mouse podocytes induced by high glucose. Int J Mol Med. 2014; 33:809–16.
Article
50. Madhusudhan T, Wang H, Dong W, Ghosh S, Bock F, Thangapandi VR, et al. Defective podocyte insulin signalling through p85-XBP1 promotes ATF6-dependent maladaptive ER-stress response in diabetic nephropathy. Nat Commun. 2015; 6:6496.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr