Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease

Stacey Ruiz, Pablo E Pergola, Richard A Zager, Nosratola D Vaziri, Stacey Ruiz, Pablo E Pergola, Richard A Zager, Nosratola D Vaziri

Abstract

Oxidative stress and inflammation are mediators in the development and progression of chronic kidney disease (CKD) and its complications, and they are inseparably linked as each begets and amplifies the other. CKD-associated oxidative stress is due to increased production of reactive oxygen species (ROS) and diminished antioxidant capacity. The latter is largely caused by impaired activation of Nrf2, the transcription factor that regulates genes encoding antioxidant and detoxifying molecules. Protective effects of Nrf2 are evidenced by amelioration of oxidative stress, inflammation, and kidney disease in response to natural Nrf2 activators in animal models, while Nrf2 deletion amplifies these pathogenic pathways and leads to autoimmune nephritis. Given the role of impaired Nrf2 activity in CKD-induced oxidative stress and inflammation, interventions aimed at restoring Nrf2 may be effective in retarding CKD progression. Clinical trials of the potent Nrf2 activator bardoxolone methyl showed significant improvement in renal function in CKD patients with type 2 diabetes. However, due to unforeseen complications the BEACON trial, which was designed to investigate the effect of this drug on time to end-stage renal disease or cardiovascular death in patients with advanced CKD, was prematurely terminated. This article provides an overview of the role of impaired Nrf2 activity in the pathogenesis of CKD-associated oxidative stress and inflammation and the potential utility of targeting Nrf2 in the treatment of CKD.

Figures

Figure 1. Production and Metabolism of Reactive…
Figure 1. Production and Metabolism of Reactive Oxygen Species
Normally over 95% of the oxygen consumed in the body is converted to water by acquisition of 2 electrons in a single step. However, for the remaining 5% this process occurs with the transfer of one electron at a time, leading to formation highly reactive and short-lived species, collectively referred to as ROS. The primary ROS produced in the body is superoxide which is formed from single electron reduction of molecular oxygen. The primary sources of superoxide include the mitochondria, endoplasmic reticulum, cyclooxygenase, lipoxygenase, uncoupled nitric oxide synthase (NOS), NAD(P)H oxidase, xanthine oxidase, and cytochrome P450. Antioxidants then act on ROS to generate less reactive species. For example, superoxide dismutase (SOD) converts superoxide into hydrogen peroxide, which is then reduced by catalase (CAT) into water and oxygen and by glutathione peroxidase (GPX) into water and oxidized glutathione. However in pathological states hydrogen peroxide serves as the substrate for formation of highly reactive and cytotoxic oxidants such as hydroxyl radical by catalytically-active iron (Fe2+) and hypochlorous acid by myeloperoxidase. An increase in ROS generation or decrease in antioxidant availability leads to oxidative stress and induction of the pro-inflammatory response, which contribute to disease pathogenesis.
Figure 2. Nrf2 inhibits reactive oxygen species…
Figure 2. Nrf2 inhibits reactive oxygen species and inflammatory pathways that lead to kidney dysfunction
Nrf2 is held in an inactive state bound to Keap1 in the cytoplasm. In response to oxidative stress signals from various sources, the Keap1-Nrf2 complex is disrupted, preventing degradation of Nrf2. As a result, newly synthesized Nrf2 translocates to the nucleus, where it activates the transcription of several antioxidant and detoxifying enzymes. Activation of Nrf2 also suppresses NFκB activity, thereby inhibiting inflammation.
Figure 3. Acute kidney injury leads to…
Figure 3. Acute kidney injury leads to progressive renal disease and decline in anti-inflammatory HO-1 and IL-10
(A) Following the induction of unilateral ischemic injury, a progressive loss of renal mass occurs over a three week period, culminating in a 2/3rds reduction of renal weight (left panel). Progressive tubular injury during this period is underscored by a progressive increase in the mRNA for the biomarker protein NGAL. A role for inflammation in this process is indicated by a progressive increase in pro-inflammatory cytokines, with stepwise increases in TNF-α mRNA expression. Similar results were obtained for a pro-inflammatory chemokine, MCP-1, as well as for a pro-fibrotic cytokine, TGF-β1. (Data presented in modified form from studies presented in reference 117 and from additional unpublished data; RZ). BL, baseline values; 1 d, one day, 1 and 3 weeks (wks) post ischemia. The mRNA values were quantified by RT-PCR and were divided by simultaneously obtained GAPDH levels, used as a “housekeeping” gene. (B) Whereas progressive renal disease and inflammation were noted over 3 weeks post ischemia (as shown in Panel A), there was a relative failure of HO-1 expression. While HO-1 protein levels rose at 1 day post ischemia, the levels then fell over the ensuing 3 weeks. Even more dramatic reductions in anti-inflammatory IL-10 protein levels were observed. Thus, the falling anti-inflammatory protein levels, with rising expression of pro-inflammatory genes (as depicted in Panel A), appear to represent reciprocal changes that tip the balance towards a pro-inflammatory / injury promoting state. These results are from ref. 117 and additional unpublished data from one of the authors; RZ). The HO-1 and IL-10 values are presented after factoring by total protein in the tissue extract.

References

    1. Himmelfarb J, Hakim RM. Oxidative stress in uremia. Curr Opin Nephrol Hypertens. 2003 Nov;12(6):593–8.
    1. Himmelfarb J, Stenvinkel P, Ikizler TA, Hakim RM. The elephant in uremia: oxidant stress as a unifying concept of cardiovascular disease in uremia. Kidney Int. 2002 Nov;62(5):1524–38.
    1. Vaziri ND. Roles of oxidative stress and antioxidant therapy in chronic kidney disease and hypertension. Curr Opin Nephrol Hypertens. 2004 Jan;13(1):93–9.
    1. Vaziri ND. Oxidative stress in uremia: nature, mechanisms, and potential consequences. Semin Nephrol. 2004 Sep;24(5):469–73.
    1. Halliwell B. Biochemistry of oxidative stress. Biochem Soc Trans. 2007 Nov;35(Pt 5):1147–50.
    1. Cachofeiro V, Goicochea M, de Vinuesa SG, Oubina P, Lahera V, Luno J. Oxidative stress and inflammation, a link between chronic kidney disease and cardiovascular disease. Kidney Int Suppl. 2008 Dec;(111):S4–S9.
    1. Rosenthal J, Nocera DG. Role of proton-coupled electron transfer in O-O bond activation. Acc Chem Res. 2007 Jul;40(7):543–53.
    1. Gennis RB. Coupled proton and electron transfer reactions in cytochrome oxidase. Front Biosci. 2004 Jan 1;9:581–91.
    1. Bartz RR, Piantadosi CA. Clinical review: oxygen as a signaling molecule. Crit Care. 2010;14(5):234.
    1. Gutteridge JM, Halliwell B. Antioxidants: Molecules, medicines, and myths. Biochem Biophys Res Commun. 2010 Mar 19;393(4):561–4.
    1. Li W, Khor TO, Xu C, Shen G, Jeong WS, Yu S, et al. Activation of Nrf2-antioxidant signaling attenuates NFkappaB-inflammatory response and elicits apoptosis. Biochem Pharmacol. 2008 Dec 1;76(11):148–59.
    1. Wakabayashi N, Slocum SL, Skoko JJ, Shin S, Kensler TW. When NRF2 talks, who’s listening? Antioxid Redox Signal. 2010 Dec 1;13(11):1649–63.
    1. Kobayashi A, Kang MI, Watai Y, Tong KI, Shibata T, Uchida K, et al. Oxidative and electrophilic stresses activate Nrf2 through inhibition of ubiquitination activity of Keap1. Mol Cell Biol. 2006 Jan;26(1):221–9.
    1. Sykiotis GP, Bohmann D. Stress-activated cap’n’collar transcription factors in aging and human disease. Sci Signal. 2010;3(112):re3.
    1. Uruno A, Motohashi H. The Keap1-Nrf2 system as an in vivo sensor for electrophiles. Nitric Oxide. 2011 Aug 1;25(2):153–60.
    1. Surh YJ, Kundu JK, Na HK. Nrf2 as a master redox switch in turning on the cellular signaling involved in the induction of cytoprotective genes by some chemopreventive phytochemicals. Planta Med. 2008 Oct;74(13):1526–39.
    1. Cullinan SB, Diehl JA. PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. J Biol Chem. 2004 May 7;279(19):20108–17.
    1. Yoh K, Itoh K, Enomoto A, Hirayama A, Yamaguchi N, Kobayashi M, et al. Nrf2-deficient female mice develop lupus-like autoimmune nephritis. Kidney Int. 2001 Oct;60(4):1343–53.
    1. Yoh K, Hirayama A, Ishizaki K, Yamada A, Takeuchi M, Yamagishi S, et al. Hyperglycemia induces oxidative and nitrosative stress and increases renal functional impairment in Nrf2-deficient mice. Genes Cells. 2008 Nov;13(11):1159–70.
    1. Malhotra JD, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress: a vicious cycle or a double-edged sword? Antioxid Redox Signal. 2007 Dec;9(12):2277–93.
    1. Vaziri ND, Bai Y, Ni Z, Quiroz Y, Pandian R, Rodriguez-Iturbe B. Intra-renal angiotensin II/AT1 receptor, oxidative stress, inflammation, and progressive injury in renal mass reduction. J Pharmacol Exp Ther. 2007 Oct;323(1):85–93.
    1. Vio CP, Jeanneret VA. Local induction of angiotensin-converting enzyme in the kidney as a mechanism of progressive renal diseases. Kidney Int Suppl. 2003 Oct;(86):S57–S63.
    1. Kobori H, Nangaku M, Navar LG, Nishiyama A. The intrarenal renin-angiotensin system: from physiology to the pathobiology of hypertension and kidney disease. Pharmacol Rev. 2007 Sep;59(3):251–87.
    1. Matsusaka T, Niimura F, Shimizu A, Pastan I, Saito A, Kobori H, et al. Liver Angiotensinogen Is the Primary Source of Renal Angiotensin II. J Am Soc Nephrol. 2012 Apr 19;
    1. Kim HJ, Sato T, Rodriguez-Iturbe B, Vaziri ND. Role of intrarenal angiotensin system activation, oxidative stress, inflammation, and impaired nuclear factor-erythroid-2-related factor 2 activity in the progression of focal glomerulosclerosis. J Pharmacol Exp Ther. 2011 Jun;337(3):583–90.
    1. Vaziri ND, Dicus M, Ho ND, Boroujerdi-Rad L, Sindhu RK. Oxidative stress and dysregulation of superoxide dismutase and NADPH oxidase in renal insufficiency. Kidney Int. 2003 Jan;63(1):179–85.
    1. Shah SV, Baliga R, Rajapurkar M, Fonseca VA. Oxidants in chronic kidney disease. J Am Soc Nephrol. 2007 Jan;18(1):16–28.
    1. Fujihara CK, Antunes GR, Mattar AL, Malheiros DM, Vieira JM, Jr., Zatz R. Chronic inhibition of nuclear factor-kappaB attenuates renal injury in the 5/6 renal ablation model. Am J Physiol Renal Physiol. 2007 Jan;292(1):F92–F99.
    1. Cho KH, Kim HJ, Rodriguez-Iturbe B, Vaziri ND. Niacin ameliorates oxidative stress, inflammation, proteinuria, and hypertension in rats with chronic renal failure. Am J Physiol Renal Physiol. 2009 Jul;297(1):F106–F113.
    1. Poelstra K, Hardonk MJ, Koudstaal J, Bakker WW. Intraglomerular platelet aggregation and experimental glomerulonephritis. Kidney Int. 1990 Jun;37(6):1500–8.
    1. Gaertner SA, Janssen U, Ostendorf T, Koch KM, Floege J, Gwinner W. Glomerular oxidative and antioxidative systems in experimental mesangioproliferative glomerulonephritis. J Am Soc Nephrol. 2002 Dec;13(12):2930–7.
    1. Boyce NW, Tipping PG, Holdsworth SR. Glomerular macrophages produce reactive oxygen species in experimental glomerulonephritis. Kidney Int. 1989 Mar;35(3):778–82.
    1. Oberle GP, Niemeyer J, Thaiss F, Schoeppe W, Stahl RA. Increased oxygen radical and eicosanoid formation in immune-mediated mesangial cell injury. Kidney Int. 1992 Jul;42(1):69–74.
    1. Kawaguchi M, Yamada M, Wada H, Okigaki T. Roles of active oxygen species in glomerular epithelial cell injury in vitro caused by puromycin aminonucleoside. Toxicology. 1992;72(3):329–40.
    1. Neale TJ, Ullrich R, Ojha P, Poczewski H, Verhoeven AJ, Kerjaschki D. Reactive oxygen species and neutrophil respiratory burst cytochrome b558 are produced by kidney glomerular cells in passive Heymann nephritis. Proc Natl Acad Sci U S A. 1993 Apr 15;90(8):3645–9.
    1. Shah SV. Evidence suggesting a role for hydroxyl radical in passive Heymann nephritis in rats. Am J Physiol. 1988 Mar;254(3 Pt 2):F337–F344.
    1. U.S. Renal Data System . USRDS 2011 Annual Data Report: Atlas of Chronic Kidney Disease and End-Stage Renal Disease in the United States. National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases; Bethesda, MD: 2011.
    1. Ha H, Yu MR, Choi YJ, Kitamura M, Lee HB. Role of high glucose-induced nuclear factor-kappaB activation in monocyte chemoattractant protein-1 expression by mesangial cells. J Am Soc Nephrol. 2002 Apr;13(4):894–902.
    1. Ha H, Kim KH. Pathogenesis of diabetic nephropathy: the role of oxidative stress and protein kinase C. Diabetes Res Clin Pract. 1999 Sep;45(2-3):147–51.
    1. Chen HC, Guh JY, Shin SJ, Tsai JH, Lai YH. Reactive oxygen species enhances endothelin-1 production of diabetic rat glomeruli in vitro and in vivo. J Lab Clin Med. 2000 Apr;135(4):309–15.
    1. Koya D, Hayashi K, Kitada M, Kashiwagi A, Kikkawa R, Haneda M. Effects of antioxidants in diabetes-induced oxidative stress in the glomeruli of diabetic rats. J Am Soc Nephrol. 2003 Aug;14(8 Suppl 3):S250–S253.
    1. Scivittaro V, Ganz MB, Weiss MF. AGEs induce oxidative stress and activate protein kinase C-beta(II) in neonatal mesangial cells. Am J Physiol Renal Physiol. 2000 Apr;278(4):F676–F683.
    1. Yan SD, Schmidt AM, Anderson GM, Zhang J, Brett J, Zou YS, et al. Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J Biol Chem. 1994 Apr 1;269(13):9889–97.
    1. Gorin Y, Block K, Hernandez J, Bhandari B, Wagner B, Barnes JL, et al. Nox4 NAD(P)H oxidase mediates hypertrophy and fibronectin expression in the diabetic kidney. J Biol Chem. 2005 Nov 25;280(47):39616–26.
    1. Satoh M, Fujimoto S, Haruna Y, Arakawa S, Horike H, Komai N, et al. NAD(P)H oxidase and uncoupled nitric oxide synthase are major sources of glomerular superoxide in rats with experimental diabetic nephropathy. Am J Physiol Renal Physiol. 2005 Jun;288(6):F1144–F1152.
    1. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001 Dec 13;414(6865):813–20.
    1. Lee HB, Yu MR, Yang Y, Jiang Z, Ha H. Reactive oxygen species-regulated signaling pathways in diabetic nephropathy. J Am Soc Nephrol. 2003 Aug;14(8 Suppl 3):S241–S245.
    1. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, Kaneda Y, et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature. 2000 Apr 13;404(6779):787–90.
    1. Vaziri ND, Rodriguez-Iturbe B. Mechanisms of disease: oxidative stress and inflammation in the pathogenesis of hypertension. Nat Clin Pract Nephrol. 2006 Oct;2(10):582–93.
    1. Rodriguez-Iturbe B, Vaziri ND, Herrera-Acosta J, Johnson RJ. Oxidative stress, renal infiltration of immune cells, and salt-sensitive hypertension: all for one and one for all. Am J Physiol Renal Physiol. 2004 Apr;286(4):F606–F616.
    1. Wilcox CS. Oxidative stress and nitric oxide deficiency in the kidney: a critical link to hypertension? Am J Physiol Regul Integr Comp Physiol. 2005 Oct;289(4):R913–R935.
    1. Barton CH, Ni Z, Vaziri ND. Enhanced nitric oxide inactivation in aortic coarctation-induced hypertension. Kidney Int. 2001 Sep;60(3):1083–7.
    1. Sindhu RK, Roberts CK, Ehdaie A, Zhan CD, Vaziri ND. Effects of aortic coarctation on aortic antioxidant enzymes and NADPH oxidase protein expression. Life Sci. 2005 Jan 7;76(8):945–53.
    1. Vaziri ND, Ni Z. Expression of NOX-I, gp91phox, p47phox and P67phox in the aorta segments above and below coarctation. Biochim Biophys Acta. 2005 May 25;1723(1-3):321–7.
    1. Yilmaz MI, Carrero JJ, Axelsson J, Lindholm B, Stenvinkel P. Low-grade inflammation in chronic kidney disease patients before the start of renal replacement therapy: sources and consequences. Clin Nephrol. 2007 Jul;68(1):1–9.
    1. Vaziri ND, Yuan J, Rahimi A, Ni Z, Said H, Subramanian VS. Disintegration of colonic epithelial tight junction in uremia: a likely cause of CKD-associated inflammation. Nephrol Dial Transplant. 2011 Nov 29;
    1. Vaziri ND, Wong J, Pahl MV, Piceno YM, Yuan J, DeSantis TZ, et al. Chronic kidney disease alters the composition of intestinal microbial flora. Kidney Int. 2012 (In press)
    1. Aronov PA, Luo FJ, Plummer NS, Quan Z, Holmes S, Hostetter TH, et al. Colonic contribution to uremic solutes. J Am Soc Nephrol. 2011 Sep;22(9):1769–76.
    1. Kim HJ, Vaziri ND. Contribution of impaired Nrf2-Keap1 pathway to oxidative stress and inflammation in chronic renal failure. Am J Physiol Renal Physiol. 2009 Dec 9;
    1. Aminzadeh AM, Sato T, Vaziri ND. Participation of endoplasmic reticulum stress in the pathogenesis of spontaneous glomerulosclerosis - Role of intra-renal angiotensin system. Transl Res. 2012 (In press)
    1. Liu M, Grigoryev DN, Crow MT, Haas M, Yamamoto M, Reddy SP, et al. Transcription factor Nrf2 is protective during ischemic and nephrotoxic acute kidney injury in mice. Kidney Int. 2009 May 13;
    1. Zheng H, Whitman SA, Wu W, Wondrak GT, Wong PK, Fang D, et al. Therapeutic potential of Nrf2 activators in streptozotocin-induced diabetic nephropathy. Diabetes. 2011 Nov;60(11):3055–66.
    1. Tsai PY, Ka SM, Chang JM, Chen HC, Shui HA, Li CY, et al. Epigallocatechin-3-gallate prevents lupus nephritis development in mice via enhancing the Nrf2 antioxidant pathway and inhibiting NLRP3 inflammasome activation. Free Radic Biol Med. 2011 Aug 1;51(3):744–54.
    1. Peng A, Ye T, Rakheja D, Tu Y, Wang T, Du Y, et al. The green tea polyphenol (-)-epigallocatechin-3-gallate ameliorates experimental immune-mediated glomerulonephritis. Kidney Int. 2011 Sep;80(6):601–11.
    1. Sahin K, Tuzcu M, Gencoglu H, Dogukan A, Timurkan M, Sahin N, et al. Epigallocatechin-3-gallate activates Nrf2/HO-1 signaling pathway in cisplatin-induced nephrotoxicity in rats. Life Sci. 2010 Aug 14;87(7-8):240–5.
    1. Molina-Jijon E, Tapia E, Zazueta C, El HM, Zatarain-Barron ZL, Hernandez-Pando R, et al. Curcumin prevents Cr(VI)-induced renal oxidant damage by a mitochondrial pathway. Free Radic Biol Med. 2011 Oct 15;51(8):1543–57.
    1. Chen XL, Dodd G, Thomas S, Zhang X, Wasserman MA, Rovin BH, et al. Activation of Nrf2/ARE pathway protects endothelial cells from oxidant injury and inhibits inflammatory gene expression. Am J Physiol Heart Circ Physiol. 2006 May;290(5):H1862–H1870.
    1. Morimitsu Y, Nakagawa Y, Hayashi K, Fujii H, Kumagai T, Nakamura Y, et al. A sulforaphane analogue that potently activates the Nrf2-dependent detoxification pathway. J Biol Chem. 2002 Feb 1;277(5):3456–63.
    1. Surh YJ, Kundu JK, Na HK, Lee JS. Redox-sensitive transcription factors as prime targets for chemoprevention with anti-inflammatory and antioxidative phytochemicals. J Nutr. 2005 Dec;135(12 Suppl):2993S–3001S.
    1. Vaziri ND. Lipotoxicity and impaired high density lipoprotein-mediated reverse cholesterol transport in chronic kidney disease. J Ren Nutr. 2010 Sep;20(5 Suppl):S35–S43.
    1. Vaziri ND. Dyslipidemia of chronic renal failure: the nature, mechanisms, and potential consequences. Am J Physiol Renal Physiol. 2006 Feb;290(2):F262–F272.
    1. Vaziri ND. Causes of dysregulation of lipid metabolism in chronic renal failure. Semin Dial. 2009 Nov;22(6):644–51.
    1. Moradi H, Pahl MV, Elahimehr R, Vaziri ND. Impaired antioxidant activity of high-density lipoprotein in chronic kidney disease. Transl Res. 2009 Feb;153(2):77–85.
    1. Vaziri ND, Navab M, Fogelman AM. HDL metabolism and activity in chronic kidney disease. Nat Rev Nephrol. 2010 May;6(5):287–96.
    1. Vaziri ND, Moradi H, Pahl MV, Fogelman AM, Navab M. In vitro stimulation of HDL anti-inflammatory activity and inhibition of LDL pro-inflammatory activity in the plasma of patients with end-stage renal disease by an apoA-1 mimetic peptide. Kidney Int. 2009 Aug;76(4):437–44.
    1. Vaziri ND, Deng G, Liang K. Hepatic HDL receptor, SR-B1 and Apo A-I expression in chronic renal failure. Nephrol Dial Transplant. 1999 Jun;14(6):1462–6.
    1. Vaziri ND, Liang K, Parks JS. Down-regulation of hepatic lecithin:cholesterol acyltransferase gene expression in chronic renal failure. Kidney Int. 2001 Jun;59(6):2192–6.
    1. Vaziri ND, Norris K. Lipid disorders and their relevance to outcomes in chronic kidney disease. Blood Purif. 2011;31(1-3):189–96.
    1. Kim HJ, Moradi H, Yuan J, Norris K, Vaziri ND. Renal mass reduction results in accumulation of lipids and dysregulation of lipid regulatory proteins in the remnant kidney. Am J Physiol Renal Physiol. 2009 Jun;296(6):F1297–F1306.
    1. Moradi H, Yuan J, Ni Z, Norris K, Vaziri ND. Reverse cholesterol transport pathway in experimental chronic renal failure. Am J Nephrol. 2009;30(2):147–54.
    1. Bea F, Hudson FN, Chait A, Kavanagh TJ, Rosenfeld ME. Induction of glutathione synthesis in macrophages by oxidized low-density lipoproteins is mediated by consensus antioxidant response elements. Circ Res. 2003 Mar 7;92(4):386–93.
    1. Cho S, Hazama M, Urata Y, Goto S, Horiuchi S, Sumikawa K, et al. Protective role of glutathione synthesis in response to oxidized low density lipoprotein in human vascular endothelial cells. Free Radic Biol Med. 1999 Mar;26(5-6):589–602.
    1. Ishii T, Itoh K, Ruiz E, Leake DS, Unoki H, Yamamoto M, et al. Role of Nrf2 in the regulation of CD36 and stress protein expression in murine macrophages: activation by oxidatively modified LDL and 4-hydroxynonenal. Circ Res. 2004 Mar 19;94(5):609–16.
    1. Dinkova-Kostova AT, Liby KT, Stephenson KK, Holtzclaw WD, Gao X, Suh N, et al. Extremely potent triterpenoid inducers of the phase 2 response: correlations of protection against oxidant and inflammatory stress. Proc Natl Acad Sci U S A. 2005 Mar 22;102(12):4584–9.
    1. Honda T, Rounds BV, Bore L, Finlay HJ, Favaloro FG, Jr., Suh N, et al. Synthetic oleanane and ursane triterpenoids with modified rings A and C: a series of highly active inhibitors of nitric oxide production in mouse macrophages. J Med Chem. 2000 Nov 2;43(22):4233–46.
    1. Sporn MB, Liby KT, Yore MM, Fu L, Lopchuk JM, Gribble GW. New synthetic triterpenoids: potent agents for prevention and treatment of tissue injury caused by inflammatory and oxidative stress. J Nat Prod. 2011 Mar 25;74(3):537–45.
    1. Levonen AL, Landar A, Ramachandran A, Ceaser EK, Dickinson DA, Zanoni G, et al. Cellular mechanisms of redox cell signalling: role of cysteine modification in controlling antioxidant defences in response to electrophilic lipid oxidation products. Biochem J. 2004 Mar 1;378(Pt 2):373–82.
    1. Cernuda-Morollon E, Pineda-Molina E, Canada FJ, Perez-Sala D. 15-Deoxy-Delta 12,14-prostaglandin J2 inhibition of NF-kappaB-DNA binding through covalent modification of the p50 subunit. J Biol Chem. 2001 Sep 21;276(38):35530–6.
    1. Cuzzocrea S, Wayman NS, Mazzon E, Dugo L, Di PR, Serraino I, et al. The cyclopentenone prostaglandin 15-deoxy-Delta(12,14)-prostaglandin J(2) attenuates the development of acute and chronic inflammation. Mol Pharmacol. 2002 May;61(5):997–1007.
    1. Kawamoto Y, Nakamura Y, Naito Y, Torii Y, Kumagai T, Osawa T, et al. Cyclopentenone prostaglandins as potential inducers of phase II detoxification enzymes. 15-deoxy-delta(12,14)-prostaglandin j2-induced expression of glutathione S-transferases. J Biol Chem. 2000 Apr 14;275(15):11291–9.
    1. Rossi A, Kapahi P, Natoli G, Takahashi T, Chen Y, Karin M, et al. Anti-inflammatory cyclopentenone prostaglandins are direct inhibitors of IkappaB kinase. Nature. 2000 Jan 6;403(6765):103–8.
    1. Straus DS, Pascual G, Li M, Welch JS, Ricote M, Hsiang CH, et al. 15-deoxy-delta 12,14-prostaglandin J2 inhibits multiple steps in the NF-kappa B signaling pathway. Proc Natl Acad Sci U S A. 2000 Apr 25;97(9):4844–9.
    1. Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 2007;47:89–116.
    1. Ahmad R, Raina D, Meyer C, Kharbanda S, Kufe D. Triterpenoid CDDO-Me Blocks the NF-{kappa}B Pathway by Direct Inhibition of IKKbeta on Cys-179. J Biol Chem. 2006 Nov 24;281(47):35764–9.
    1. Rivero A, Mora C, Muros M, Garcia J, Herrera H, Navarro-Gonzalez JF. Pathogenic perspectives for the role of inflammation in diabetic nephropathy. Clin Sci (Lond) 2009 Mar;116(6):479–92.
    1. Navarro-Gonzalez JF, Mora-Fernandez C. Inflammatory pathways. Contrib Nephrol. 2011;170:113–23.
    1. Hong DS, Kurzrock R, Supko JG, He X, Naing A, Wheler J, et al. A Phase I First-in-Human Trial of Bardoxolone Methyl in Patients with Advanced Solid Tumors and Lymphomas. Clin Cancer Res. 2012 May 25;
    1. Rossing P. Diabetic nephropathy: worldwide epidemic and effects of current treatment on natural history. Curr Diab Rep. 2006 Dec;6(6):479–83.
    1. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001 Sep 20;345(12):861–9.
    1. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med. 2001 Sep 20;345(12):851–60.
    1. Vilayur E, Harris DC. Emerging therapies for chronic kidney disease: what is their role? Nat Rev Nephrol. 2009 Jul;5(7):375–83.
    1. Pergola P, Krauth M, Huff W, Ferguson D, Ruiz S, Meyer C, et al. Effect of Bardoxolone on Kidney Function in Patients with T2D and Stage 3b - 4 CKD. Am J Nephrol. 2011;33:469–76.
    1. Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW, Grossman EB, et al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N Engl J Med. 2011 Jul 28;365(4):327–36.
    1. Navarro-Gonzalez JF, Mora-Fernandez C, Muros de FM, Garcia-Perez J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol. 2011 Jun;7(6):327–40.
    1. Shah SV, Rajapurkar MM, Baliga R. The role of catalytic iron in acute kidney injury. Clin J Am Soc Nephrol. 2011 Oct;6(10):2329–31.
    1. Aksu U, Demirci C, Ince C. The pathogenesis of acute kidney injury and the toxic triangle of oxygen, reactive oxygen species and nitric oxide. Contrib Nephrol. 2011;174:119–28.
    1. Bonventre JV, Yang L. Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest. 2011 Nov;121(11):4210–21.
    1. Lee DW, Faubel S, Edelstein CL. Cytokines in acute kidney injury (AKI) Clin Nephrol. 2011 Sep;76(3):165–73.
    1. Kinsey GR, Okusa MD. Pathogenesis of acute kidney injury: foundation for clinical practice. Am J Kidney Dis. 2011 Aug;58(2):291–301.
    1. Miyazaki T, Kirino Y, Takeno M, Samukawa S, Hama M, Tanaka M, et al. Expression of heme oxygenase-1 in human leukemic cells and its regulation by transcriptional repressor Bach1. Cancer Sci. 2010 Jun;101(6):1409–16.
    1. Igarashi K, Sun J. The heme-Bach1 pathway in the regulation of oxidative stress response and erythroid differentiation. Antioxid Redox Signal. 2006 Jan;8(1-2):107–18.
    1. Keum YS, Han YH, Liew C, Kim JH, Xu C, Yuan X, et al. Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. Pharm Res. 2006 Nov;23(11):2586–94.
    1. Tenhunen R, Marver HS, Schmid R. The enzymatic conversion of heme to bilirubin by microsomal heme oxygenase. Proc Natl Acad Sci U S A. 1968 Oct;61(2):748–55.
    1. Nath KA, Balla G, Vercellotti GM, Balla J, Jacob HS, Levitt MD, et al. Induction of heme oxygenase is a rapid, protective response in rhabdomyolysis in the rat. J Clin Invest. 1992 Jul;90(1):267–70.
    1. Nath KA. Heme oxygenase-1: a provenance for cytoprotective pathways in the kidney and other tissues. Kidney Int. 2006 Aug;70(3):432–43.
    1. Inguaggiato P, Gonzalez-Michaca L, Croatt AJ, Haggard JJ, Alam J, Nath KA. Cellular overexpression of heme oxygenase-1 up-regulates p21 and confers resistance to apoptosis. Kidney Int. 2001 Dec;60(6):2181–91.
    1. Agarwal A, Balla J, Alam J, Croatt AJ, Nath KA. Induction of heme oxygenase in toxic renal injury: a protective role in cisplatin nephrotoxicity in the rat. Kidney Int. 1995 Oct;48(4):1298–307.
    1. Vogt BA, Alam J, Croatt AJ, Vercellotti GM, Nath KA. Acquired resistance to acute oxidative stress. Possible role of heme oxygenase and ferritin. Lab Invest. 1995 Apr;72(4):474–83.
    1. Agarwal A, Nick HS. Renal response to tissue injury: lessons from heme oxygenase-1 GeneAblation and expression. J Am Soc Nephrol. 2000 May;11(5):965–73.
    1. Nath KA. Heme oxygenase-1: a redoubtable response that limits reperfusion injury in the transplanted adipose liver. J Clin Invest. 1999 Dec;104(11):1485–6.
    1. Zarjou A, Kim J, Traylor AM, Sanders PW, Balla J, Agarwal A, et al. Paracrine effects of mesenchymal stem cells in cisplatin-induced renal injury require heme oxygenase-1. Am J Physiol Renal Physiol. 2011 Jan;300(1):F254–F262.
    1. Pittock ST, Norby SM, Grande JP, Croatt AJ, Bren GD, Badley AD, et al. MCP-1 is up-regulated in unstressed and stressed HO-1 knockout mice: Pathophysiologic correlates. Kidney Int. 2005 Aug;68(2):611–22.
    1. Tracz MJ, Juncos JP, Croatt AJ, Ackerman AW, Grande JP, Knutson KL, et al. Deficiency of heme oxygenase-1 impairs renal hemodynamics and exaggerates systemic inflammatory responses to renal ischemia. Kidney Int. 2007 Nov;72(9):10735–80.
    1. Stocker R, Yamamoto Y, McDonagh AF, Glazer AN, Ames BN. Bilirubin is an antioxidant of possible physiological importance. Science. 1987 Feb 27;235(4792):1043–6.
    1. Nakahira K, Kim HP, Geng XH, Nakao A, Wang X, Murase N, et al. Carbon monoxide differentially inhibits TLR signaling pathways by regulating ROS-induced trafficking of TLRs to lipid rafts. J Exp Med. 2006 Oct 2;203(10):2377–89.
    1. Pamplona A, Ferreira A, Balla J, Jeney V, Balla G, Epiphanio S, et al. Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimental cerebral malaria. Nat Med. 2007 Jun;13(6):703–10.
    1. Tracz MJ, Alam J, Nath KA. Physiology and pathophysiology of heme: implications for kidney disease. J Am Soc Nephrol. 2007 Feb;18(2):414–20.
    1. Dulak J, Deshane J, Jozkowicz A, Agarwal A. Heme oxygenase-1 and carbon monoxide in vascular pathobiology: focus on angiogenesis. Circulation. 2008 Jan 15;117(2):231–41.
    1. Abraham NG, Kappas A. Pharmacological and clinical aspects of heme oxygenase. Pharmacol Rev. 2008 Mar;60(1):79–127.
    1. Wu QQ, Wang Y, Senitko M, Meyer C, Wigley WC, Ferguson DA, et al. Bardoxolone Methyl (BARD) ameliorates ischemic AKI and increases expression of protective genes - Nrf2, PPAR{gamma}, and HO-1. Am J Physiol Renal Physiol. 2011 Feb 2;
    1. Aleksunes LM, Goedken MJ, Rockwell CE, Thomale J, Manautou JE, Klaassen CD. Transcriptional regulation of renal cytoprotective genes by Nrf2 and its potential use as a therapeutic target to mitigate cisplatin-induced nephrotoxicity. J Pharmacol Exp Ther. 2010 Oct;335(1):2–12.
    1. Lo LJ, Go AS, Chertow GM, McCulloch CE, Fan D, Ordonez JD, et al. Dialysis-requiring acute renal failure increases the risk of progressive chronic kidney disease. Kidney Int. 2009 Oct;76(8):893–9.
    1. Zager RA, Johnson AC, Becker K. Acute unilateral ischemic renal injury induces progressive renal inflammation, lipid accumulation, histone modification, and “end-stage” kidney disease. Am J Physiol Renal Physiol. 2011 Dec;301(6):F1334–F1345.
    1. Zager RA, Johnson AC. Progressive histone alterations and proinflammatory gene activation: consequences of heme protein/iron-mediated proximal tubule injury. Am J Physiol Renal Physiol. 2010 Mar;298(3):F827–F837.
    1. Naito M, Zager RA, Bomsztyk K. BRG1 increases transcription of proinflammatory genes in renal ischemia. J Am Soc Nephrol. 2009 Aug;20(8):1787–96.
    1. Zager RA, Johnson AC. Renal ischemia-reperfusion injury upregulates histone-modifying enzyme systems and alters histone expression at proinflammatory/profibrotic genes. Am J Physiol Renal Physiol. 2009 May;296(5):F1032–F1041.
    1. Naito M, Bomsztyk K, Zager RA. Endotoxin mediates recruitment of RNA polymerase II to target genes in acute renal failure. J Am Soc Nephrol. 2008 Jul;19(7):1321–30.
    1. Zager RA, Johnson AC, Hanson SY, Lund S. Ischemic proximal tubular injury primes mice to endotoxin-induced TNF-alpha generation and systemic release. Am J Physiol Renal Physiol. 2005 Aug;289(2):F289–F297.

Source: PubMed

3
Abonner