Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2015, Vol. 9 Issue (1) : 46-56    https://doi.org/10.1007/s11684-015-0375-1
REVIEW
Triterpenoid inducers of Nrf2 signaling as potential therapeutic agents in sickle cell disease: a review
Amma Owusu-Ansah1,2,Sung Hee Choi1,1_FMD-14249-OAA _FMD-14249-OAA,Agne Petrosiute1,2,John J. Letterio1,2,Alex Yee-Chen Huang1,2,*()
1. Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
2. The Angie Fowler Adolescent & Young Adult Cancer Institute at University Hospitals, Rainbow Babies & Children’s Hospital, Cleveland, OH 44106, USA
 Download: PDF(631 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Sickle cell disease (SCD) is an inherited disorder of hemoglobin in which the abnormal hemoglobin S polymerizes when deoxygenated. This polymerization of hemoglobin S not only results in hemolysis and vaso-occlusion but also precipitates inflammation, oxidative stress and chronic organ dysfunction. Oxidative stress is increasingly recognized as an important intermediate in these pathophysiological processes and is therefore an important target for therapeutic intervention. The transcription factor nuclear erythroid derived- 2 related factor 2 (Nrf2) controls the expression of anti-oxidant enzymes and is emerging as a protein whose function can be exploited with therapeutic intent. This review article is focused on triterpenoids that activate Nrf2, and their potential for reducing oxidative stress in SCD as an approach to prevent organ dysfunction associated with this disease. A brief overview of oxidative stress in the clinical context of SCD is accompanied by a discussion of several pathophysiological mechanisms contributing to oxidative stress. Finally, these mechanisms are then related to current management strategies in SCD that are either utilized currently or under evaluation. The article concludes with a perspective on the potential of the various therapeutic interventions to reduce oxidative stress and morbidity associated with SCD.

Keywords oxidative stress      Nrf2      triterpenoids      sickle cell disease      vaso-occlusion      CDDO-Me     
Corresponding Author(s): Alex Yee-Chen Huang   
Just Accepted Date: 06 November 2014   Online First Date: 12 December 2014    Issue Date: 02 March 2015
 Cite this article:   
Amma Owusu-Ansah,Sung Hee Choi,Agne Petrosiute, et al. Triterpenoid inducers of Nrf2 signaling as potential therapeutic agents in sickle cell disease: a review[J]. Front. Med., 2015, 9(1): 46-56.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-015-0375-1
https://academic.hep.com.cn/fmd/EN/Y2015/V9/I1/46
Fig.1  The Keap1-Nrf2 antioxidant pathway. CDDO-Me is a triterpenoid inhibitor of Keap1, the cytosolic repressor that targets Nrf2 for ubiquitination. Triterpenoid binds to reactive thiol groups on cysteine 151 within the redox sensitive BTB domain of Keap1. Under basal conditions, Nrf2 is bound to Keap1 in the cytoplasm and is ubiquitinated. Under conditions of electrophilic or oxidative stress, Nrf2 dissociates from Keap1 and is protected from degradation. Nrf2 then accumulates in the nucleus where it will heterodimerize with MAF proteins and then bind to the antioxidant response element (ARE), located on the promoter of several genes involved in the phase 2 response such as HO-1, GST, GCLC and NQO1.
Fig.2  Pathophysiological processes in SCD contribute to oxidative stress. A schematic diagram of how hemolysis, vasoconstriction, inflammation and ischemia reperfusion injury interact to create a state of oxidative stress is depicted. As byproducts of hemolysis, both arginase-depleting L arginine and free heme are released from red cells and subsequently diffuse into neighboring cells, leading to iron deposits which catalyze ROS formation. Free heme also recruits neutrophils to the endothelium. Endothelial NOS becomes uncoupled under conditions of NO depletion, generating ROS and reactive nitrogen species (RNS) instead of NO. Vasoconstriction occurs when endothelin-1 (ET1), a vasoconstrictor that opposes the vasodilatory effects of NO, predominates in NO depletion. The inhibitory effect of NO on inflammation and endothelial activation is lost leading to expression of adhesion molecules like VCAM, ICAM, P-selectins and L-selectins that recruit PMN and thrombospondin (TSP) to which reticulocytes adhere via CD36. Sickle red cells expressing externalized phosphatidylserine (PS) on their membranes cause platelet (plt) activation and are phagocytosed by macrophages (M) contributing further to hemolysis. Xanthine oxidase (XO) levels are increased during reperfusion injury, increasing ROS production.
Fig.3  Chemical structures of select SOTs. The structures of SOTs are depicted, including 2-cyano-3,12-dioxooleana-1,9(11)-diene-28-oic acid (CDDO) and its analogs methyl 2-cyano-3,12-dioxooleana-1,9(11)-diene-28-oate(CDDO-Me), 2-cyano-3,12-dioxooleana-1,9-diene-28-oic acid-methylamide (CDDO-MA), 1[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]-imidazole (CDDO-Im), and CDDO trifluoroethylamide (CDDO-TFEA).
1 Wood KC, Granger DN. Sickle cell disease: role of reactive oxygen and nitrogen metabolites. Clin Exp Pharmacol Physiol 2007; 34(9): 926–932
https://doi.org/10.1111/j.1440-1681.2007.04639.x pmid: 17645642
2 Rees DC, Williams TN, Gladwin MT. Sickle-cell disease. Lancet 2010; 376(9757): 2018–2031
https://doi.org/10.1016/S0140-6736(10)61029-X pmid: 21131035
3 Steinberg MH. Pathophysiologically based drug treatment of sickle cell disease. Trends Pharmacol Sci 2006; 27(4): 204–210
https://doi.org/10.1016/j.tips.2006.02.007 pmid: 16530854
4 Nur E, Biemond BJ, Otten HM, Brandjes DP, Schnog JJ; CURAMA Study Group. Oxidative stress in sickle cell disease; pathophysiology and potential implications for disease management. Am J Hematol 2011; 86(6): 484–489
https://doi.org/10.1002/ajh.22012 pmid: 21544855
5 Platt OS, Brambilla DJ, Rosse WF, Milner PF, Castro O, Steinberg MH, Klug PP. Mortality in sickle cell disease. Life expectancy and risk factors for early death. N Engl J Med 1994; 330(23): 1639–1644
https://doi.org/10.1056/NEJM199406093302303 pmid: 7993409
6 Platt OS, Thorington BD, Brambilla DJ, Milner PF, Rosse WF, Vichinsky E, Kinney TR. Pain in sickle cell disease. Rates and risk factors. N Engl J Med 1991; 325(1): 11–16
https://doi.org/10.1056/NEJM199107043250103 pmid: 1710777
7 Watson J, Starman AW, Bilello FP. The significance of the paucity of sickle cells in newborn Negro infants. Am J Med Sci 1948; 215(4): 419–423
https://doi.org/10.1097/00000441-194804000-00008 pmid: 18107723
8 Steinberg MH, Chui DH, Dover GJ, Sebastiani P, Alsultan A. Fetal hemoglobin in sickle cell anemia: a glass half full? Blood 2014; 123(4): 481–485
https://doi.org/10.1182/blood-2013-09-528067 pmid: 24222332
9 Sankaran VG, Orkin SH . The switch from fetal to adult hemoglobin. Cold Spring Harb Perspect Med 2013; 3(1): a011643
10 Chaves MA, Leonart MS, do Nascimento AJ. Oxidative process in erythrocytes of individuals with hemoglobin S. Hematology 2008; 13(3): 187–192
https://doi.org/10.1179/102453308X343356 pmid: 18702879
11 Silva DGH, Belini Junior E, de Almeida EA, Bonini-Domingos CR. Oxidative stress in sickle cell disease: an overview of erythrocyte redox metabolism and current antioxidant therapeutic strategies. Free Radic Biol Med 2013; 65(0): 1101–1109
https://doi.org/10.1016/j.freeradbiomed.2013.08.181 pmid: 24002011
12 Gizi A, Papassotiriou I, Apostolakou F, Lazaropoulou C, Papastamataki M, Kanavaki I, Kalotychou V, Goussetis E, Kattamis A, Rombos I, Kanavakis E. Assessment of oxidative stress in patients with sickle cell disease: the glutathione system and the oxidant-antioxidant status. Blood Cells Mol Dis 2011; 46(3): 220–225
https://doi.org/10.1016/j.bcmd.2011.01.002 pmid: 21334230
13 Liby K, Hock T, Yore MM, Suh N, Place AE, Risingsong R, Williams CR, Royce DB, Honda T, Honda Y, Gribble GW, Hill-Kapturczak N, Agarwal A, Sporn MB. The synthetic triterpenoids, CDDO and CDDO-imidazolide, are potent inducers of heme oxygenase-1 and Nrf2/ARE signaling. Cancer Res 2005; 65(11): 4789–4798
https://doi.org/10.1158/0008-5472.CAN-04-4539 pmid: 15930299
14 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; 74(13): 1526–1539
https://doi.org/10.1055/s-0028-1088302 pmid: 18937164
15 Yates MS, Kensler TW. Chemopreventive promise of targeting the Nrf2 pathway. Drug News Perspect 2007; 20(2): 109–117
https://doi.org/10.1358/dnp.2007.20.2.1083437 pmid: 17440634
16 Lee JM, Li J, Johnson DA, Stein TD, Kraft AD, Calkins MJ, Jakel RJ, Johnson JA. Nrf2, a multi-organ protector? FASEB J 2005; 19(9): 1061–1066
pmid: 15985529
17 Sangokoya C, Telen MJ, Chi JT. microRNA miR-144 modulates oxidative stress tolerance and associates with anemia severity in sickle cell disease. Blood 2010; 116(20): 4338–4348
https://doi.org/10.1182/blood-2009-04-214817 pmid: 20709907
18 Itoh K, Wakabayashi N, Katoh Y, Ishii T, O’Connor T, Yamamoto M. Keap1 regulates both cytoplasmic-nuclear shuttling and degradation of Nrf2 in response to electrophiles. Genes Cells 2003; 8(4): 379–391
https://doi.org/10.1046/j.1365-2443.2003.00640.x pmid: 12653965
19 Zhu H, Itoh K, Yamamoto M, Zweier JL, Li Y. Role of Nrf2 signaling in regulation of antioxidants and phase 2 enzymes in cardiac fibroblasts: protection against reactive oxygen and nitrogen species-induced cell injury. FEBS Lett 2005; 579(14): 3029–3036
https://doi.org/10.1016/j.febslet.2005.04.058 pmid: 15896789
20 Dr?ge W. Free radicals in the physiological control of cell function. Physiol Rev 2002; 82(1): 47–95
pmid: 11773609
21 Junqueira VBC, Barros SB, Chan SS, Rodrigues L, Giavarotti L, Abud RL, Deucher GP. Aging and oxidative stress. Mol Aspects Med 2004; 25(1-2): 5–16
https://doi.org/10.1016/j.mam.2004.02.003 pmid: 15051312
22 Akohoue SA, Shankar S, Milne GL, Morrow J, Chen KY, Ajayi WU, Buchowski MS. Energy expenditure, inflammation, and oxidative stress in steady-state adolescents with sickle cell anemia. Pediatr Res 2007; 61(2): 233–238
https://doi.org/10.1203/pdr.0b013e31802d7754 pmid: 17237728
23 Manfredini V, Lazzaretti LL, Griebeler IH, Santin AP, Brand?o VD, Wagner S, Castro SM, Peralba MdoC, Benfato MS. Blood antioxidant parameters in sickle cell anemia patients in steady state. J Natl Med Assoc 2008; 100(8): 897–902
pmid: 18717139
24 Klings ES, Christman BW, McClung J, Stucchi AF, McMahon L, Brauer M, Farber HW. Increased F2 isoprostanes in the acute chest syndrome of sickle cell disease as a marker of oxidative stress. Am J Respir Crit Care Med 2001; 164(7): 1248–1252
https://doi.org/10.1164/ajrccm.164.7.2101020 pmid: 11673218
25 Nath KA, Grande JP, Haggard JJ, Croatt AJ, Katusic ZS, Solovey A, Hebbel RP. Oxidative stress and induction of heme oxygenase-1 in the kidney in sickle cell disease. Am J Pathol 2001; 158(3): 893–903
https://doi.org/10.1016/S0002-9440(10)64037-0 pmid: 11238038
26 Reiter CD, Wang X, Tanus-Santos JE, Hogg N, Cannon RO 3rd, Schechter AN, Gladwin MT. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat Med 2002; 8(12): 1383–1389
https://doi.org/10.1038/nm1202-799 pmid: 12426562
27 Morris CR. Mechanisms of vasculopathy in sickle cell disease and thalassemia. Hematology Am Soc Hematol Educ Program 2008; 2008 (1): 177–185
pmid: 19074078
28 Schnog JB, Teerlink T, van der Dijs FP, Duits AJ, Muskiet FA; CURAMA Study Group. Plasma levels of asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase inhibitor, are elevated in sickle cell disease. Ann Hematol 2005; 84(5): 282–286
https://doi.org/10.1007/s00277-004-0983-3 pmid: 15599544
29 Wood KC, Hebbel RP, Lefer DJ, Granger DN. Critical role of endothelial cell-derived nitric oxide synthase in sickle cell disease-induced microvascular dysfunction. Free Radic Biol Med 2006; 40(8): 1443–1453
https://doi.org/10.1016/j.freeradbiomed.2005.12.015 pmid: 16631534
30 Kiefmann R, Rifkind JM, Nagababu E, Bhattacharya J. Red blood cells induce hypoxic lung inflammation. Blood 2008; 111(10): 5205–5214
https://doi.org/10.1182/blood-2007-09-113902 pmid: 18270324
31 Brugnara C. Erythrocyte dehydration in pathophysiology and treatment of sickle cell disease. Curr Opin Hematol 1995; 2(2): 132–138
https://doi.org/10.1097/00062752-199502020-00005 pmid: 9371983
32 Hofstra TC, Kalra VK, Meiselman HJ, Coates TD. Sickle erythrocytes adhere to polymorphonuclear neutrophils and activate the neutrophil respiratory burst. Blood 1996; 87(10): 4440–4447
pmid: 8639806
33 Vichinsky E. Emerging ‘A’ therapies in hemoglobinopathies: agonists, antagonists, antioxidants, and arginine. Hematology Am Soc Hematol Educ Program 2012; 2012 (1): 271–275
pmid: 23233591
34 Nur E, Brandjes DP, Teerlink T, Otten HM, Oude Elferink RP, Muskiet F, Evers LM, ten Cate H, Biemond BJ, Duits AJ, Schnog JJ; CURAMA study group. N-acetylcysteine reduces oxidative stress in sickle cell patients. Ann Hematol 2012; 91(7): 1097–1105
https://doi.org/10.1007/s00277-011-1404-z pmid: 22318468
35 Zimmerman SA, Schultz WH, Davis JS, Pickens CV, Mortier NA, Howard TA, Ware RE. Sustained long-term hematologic efficacy of hydroxyurea at maximum tolerated dose in children with sickle cell disease. Blood 2004; 103(6): 2039–2045
https://doi.org/10.1182/blood-2003-07-2475 pmid: 14630791
36 Silva DG, Belini Junior E, Torres LS, Ricci Júnior O, Lobo CC, Bonini-Domingos CR, de Almeida EA. Relationship between oxidative stress, glutathione S-transferase polymorphisms and hydroxyurea treatment in sickle cell anemia. Blood Cells Mol Dis 2011; 47(1): 23–28
https://doi.org/10.1016/j.bcmd.2011.03.004 pmid: 21489839
37 Torres Lde S, da Silva DG, Belini Junior E, de Almeida EA, Lobo CL, Can?ado RD, Ruiz MA, Bonini-Domingos CR. The influence of hydroxyurea on oxidative stress in sickle cell anemia. Rev Bras Hematol Hemoter 2012; 34(6): 421–425
pmid: 23323065
38 Steinberg MH, Barton F, Castro O, Pegelow CH, Ballas SK, Kutlar A, Orringer E, Bellevue R, Olivieri N, Eckman J, Varma M, Ramirez G, Adler B, Smith W, Carlos T, Ataga K, DeCastro L, Bigelow C, Saunthararajah Y, Telfer M, Vichinsky E, Claster S, Shurin S, Bridges K, Waclawiw M, Bonds D, Terrin M. Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment. JAMA 2003; 289(13): 1645–1651
https://doi.org/10.1001/jama.289.13.1645 pmid: 12672732
39 Charache S, Terrin ML, Moore RD, Dover GJ, Barton FB, Eckert SV, McMahon RP, Bonds DR. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. N Engl J Med 1995; 332(20): 1317–1322
https://doi.org/10.1056/NEJM199505183322001 pmid: 7715639
40 Pace BS, Zein S. Understanding mechanisms of gamma-globin gene regulation to develop strategies for pharmacological fetal hemoglobin induction. Dev Dyn 2006; 235(7): 1727–1737
pmid: 16607652
41 Fathallah H, Atweh GF . Induction of fetal hemoglobin in the treatment of sickle cell disease. Hematology Am Soc Hematol Educ Program 2006: 58–62
pmid: 17124041
42 DeSimone J, Heller P, Hall L, Zwiers D. 5-Azacytidine stimulates fetal hemoglobin synthesis in anemic baboons. Proc Natl Acad Sci USA 1982; 79(14): 4428–4431
https://doi.org/10.1073/pnas.79.14.4428 pmid: 6181507
43 Saunthararajah Y, Hillery CA, Lavelle D, Molokie R, Dorn L, Bressler L, Gavazova S, Chen YH, Hoffman R, DeSimone J. Effects of 5-aza-2′-deoxycytidine on fetal hemoglobin levels, red cell adhesion, and hematopoietic differentiation in patients with sickle cell disease. Blood 2003; 102(12): 3865–3870
https://doi.org/10.1182/blood-2003-05-1738 pmid: 12907443
44 Fard AD, Hosseini SA, Shahjahani M, Salari F, Jaseb K. Evaluation of novel fetal hemoglobin inducer drugs in treatment of beta-hemoglobinopathy disorders. Int J Hematol Oncol Stem Cell Res 2013; 7(3): 47–54
pmid: 24505535
45 List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman E, Powell B, Greenberg P, Thomas D, Stone R, Reeder C, Wride K, Patin J, Schmidt M, Zeldis J, Knight R; Myelodysplastic Syndrome-003 Study Investigators. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med 2006; 355(14): 1456–1465
https://doi.org/10.1056/NEJMoa061292 pmid: 17021321
46 Moutouh-de Parseval LA, Verhelle D, Glezer E, Jensen-Pergakes K, Ferguson GD, Corral LG, Morris CL, Muller G, Brady H, Chan K. Pomalidomide and lenalidomide regulate erythropoiesis and fetal hemoglobin production in human CD34+ cells. J Clin Invest 2008; 118(1): 248–258
https://doi.org/10.1172/JCI32322 pmid: 18064299
47 Rodgers GP, Dover GJ, Noguchi CT, Schechter AN, Nienhuis AW. Hematologic responses of patients with sickle cell disease to treatment with hydroxyurea. N Engl J Med 1990; 322(15): 1037–1045
https://doi.org/10.1056/NEJM199004123221504 pmid: 1690857
48 King SB. Nitric oxide production from hydroxyurea. Free Radic Biol Med 2004; 37(6): 737–744
https://doi.org/10.1016/j.freeradbiomed.2004.02.073 pmid: 15304249
49 Gladwin MT, Shelhamer JH, Ognibene FP, Pease-Fye ME, Nichols JS, Link B, Patel DB, Jankowski MA, Pannell LK, Schechter AN, Rodgers GP. Nitric oxide donor properties of hydroxyurea in patients with sickle cell disease. Br J Haematol 2002; 116(2): 436–444
https://doi.org/10.1046/j.1365-2141.2002.03274.x pmid: 11841449
50 Gladwin MT, Kato GJ, Weiner D, Onyekwere OC, Dampier C, Hsu L, Hagar RW, Howard T, Nuss R, Okam MM, Tremonti CK, Berman B, Villella A, Krishnamurti L, Lanzkron S, Castro O, Gordeuk VR, Coles WA, Peters-Lawrence M, Nichols J, Hall MK, Hildesheim M, Blackwelder WC, Baldassarre J, Casella JF; DeNOVO Investigators. Nitric oxide for inhalation in the acute treatment of sickle cell pain crisis: a randomized controlled trial. JAMA 2011; 305(9): 893–902
https://doi.org/10.1001/jama.2011.235 pmid: 21364138
51 Morris CR, Kuypers FA, Lavrisha L, Ansari M, Sweeters N, Stewart M, Gildengorin G, Neumayr L, Vichinsky EP. A randomized, placebo-controlled trial of arginine therapy for the treatment of children with sickle cell disease hospitalized with vaso-occlusive pain episodes. Haematologica 2013; 98(9): 1375–1382
https://doi.org/10.3324/haematol.2013.086637 pmid: 23645695
52 Pace BS, Shartava A, Pack-Mabien A, Mulekar M, Ardia A, Goodman SR. Effects of N-acetylcysteine on dense cell formation in sickle cell disease. Am J Hematol 2003; 73(1): 26–32
https://doi.org/10.1002/ajh.10321 pmid: 12701116
53 Field JJ, Nathan DG, Linden J. Targeting iNKT cells for the treatment of sickle cell disease. Clin Immunol 2011; 140(2): 177–183
https://doi.org/10.1016/j.clim.2011.03.002 pmid: 21429807
54 Morris CR, Suh JH, Hagar W, Larkin S, Bland DA, Steinberg MH, Vichinsky EP, Shigenaga M, Ames B, Kuypers FA, Klings ES. Erythrocyte glutamine depletion, altered redox environment, and pulmonary hypertension in sickle cell disease. Blood 2008; 111(1): 402–410
https://doi.org/10.1182/blood-2007-04-081703 pmid: 17848621
55 Liby KT, Sporn MB. Synthetic oleanane triterpenoids: multifunctional drugs with a broad range of applications for prevention and treatment of chronic disease. Pharmacol Rev 2012; 64(4): 972–1003
https://doi.org/10.1124/pr.111.004846 pmid: 22966038
56 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; 74(3): 537–545
https://doi.org/10.1021/np100826q pmid: 21309592
57 Suh N, Wang Y, Honda T, Gribble GW, Dmitrovsky E, Hickey WF, Maue RA, Place AE, Porter DM, Spinella MJ, Williams CR, Wu G, Dannenberg AJ, Flanders KC, Letterio JJ, Mangelsdorf DJ, Nathan CF, Nguyen L, Porter WW, Ren RF, Roberts AB, Roche NS, Subbaramaiah K, Sporn MB. A novel synthetic oleanane triterpenoid, 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid, with potent differentiating, antiproliferative, and anti-inflammatory activity. Cancer Res 1999; 59(2): 336–341
pmid: 9927043
58 Dinkova-Kostova AT, Liby KT, Stephenson KK, Holtzclaw WD, Gao X, Suh N, Williams C, Risingsong R, Honda T, Gribble GW, Sporn MB, Talalay P. Extremely potent triterpenoid inducers of the phase 2 response: correlations of protection against oxidant and inflammatory stress. Proc Natl Acad Sci USA 2005; 102(12): 4584–4589
https://doi.org/10.1073/pnas.0500815102 pmid: 15767573
59 Cleasby A, Yon J, Day PJ, Richardson C, Tickle IJ, Williams PA, Callahan JF, Carr R, Concha N, Kerns JK, Qi H, Sweitzer T, Ward P, Davies TG. Structure of the BTB domain of Keap1 and its interaction with the triterpenoid antagonist CDDO. PLoS ONE 2014; 9(6): e98896
https://doi.org/10.1371/journal.pone.0098896 pmid: 24896564
60 Yates MS, Tran QT, Dolan PM, Osburn WO, Shin S, McCulloch CC, Silkworth JB, Taguchi K, Yamamoto M, Williams CR, Liby KT, Sporn MB, Sutter TR, Kensler TW. Genetic versus chemoprotective activation of Nrf2 signaling: overlapping yet distinct gene expression profiles between Keap1 knockout and triterpenoid-treated mice. Carcinogenesis 2009; 30(6): 1024–1031
https://doi.org/10.1093/carcin/bgp100 pmid: 19386581
61 Thimmulappa RK, Scollick C, Traore K, Yates M, Trush MA, Liby KT, Sporn MB, Yamamoto M, Kensler TW, Biswal S. Nrf2-dependent protection from LPS induced inflammatory response and mortality by CDDO-Imidazolide. Biochem Biophys Res Commun 2006; 351(4): 883–889
https://doi.org/10.1016/j.bbrc.2006.10.102 pmid: 17097057
62 Thimmulappa RK, Fuchs RJ, Malhotra D, Scollick C, Traore K, Bream JH, Trush MA, Liby KT, Sporn MB, Kensler TW, Biswal S. Preclinical evaluation of targeting the Nrf2 pathway by triterpenoids (CDDO-Im and CDDO-Me) for protection from LPS-induced inflammatory response and reactive oxygen species in human peripheral blood mononuclear cells and neutrophils. Antioxid Redox Signal 2007; 9(11): 1963–1970
https://doi.org/10.1089/ars.2007.1745 pmid: 17822364
63 Heiss EH, Schachner D, Werner ER, Dirsch VM. Active NF-E2-related factor (Nrf2) contributes to keep endothelial NO synthase (eNOS) in the coupled state: role of reactive oxygen species (ROS), eNOS, and heme oxygenase (HO-1) levels. J Biol Chem 2009; 284(46): 31579–31586
https://doi.org/10.1074/jbc.M109.009175 pmid: 19797052
64 Cho HY, Reddy SP, Yamamoto M, Kleeberger SR. The transcription factor NRF2 protects against pulmonary fibrosis. FASEB J 2004; 18(11): 1258–1260
pmid: 15208274
65 de Zeeuw D, Akizawa T, Agarwal R, Audhya P, Bakris GL, Chin M, Krauth M, Lambers Heerspink HJ, Meyer CJ, McMurray JJ, Parving HH, Pergola PE, Remuzzi G, Toto RD, Vaziri ND, Wanner C, Warnock DG, Wittes J, Chertow GM. Rationale and trial design of Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes: the Occurrence of Renal Events (BEACON). Am J Nephrol 2013; 37(3): 212–222
https://doi.org/10.1159/000346948 pmid: 23467003
66 Chertow GM, de Zeeuw D; BEACON Steering Committee. Bardoxolone methyl in type 2 diabetes and advanced chronic kidney disease. N Engl J Med 2014; 370(18): 1768
pmid: 24785220
67 Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW, Grossman EB, Krauth M, Ruiz S, Audhya P, Christ-Schmidt H, Wittes J, Warnock DG; BEAM Study Investigators. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N Engl J Med 2011; 365(4): 327–336
https://doi.org/10.1056/NEJMoa1105351 pmid: 21699484
68 Boyd JH, Macklin EA, Strunk RC, DeBaun MR. Asthma is associated with increased mortality in individuals with sickle cell anemia. Haematologica 2007; 92(8): 1115–1118
https://doi.org/10.3324/haematol.11213 pmid: 17650441
[1] Pengju Zhang, Tao Li, Xingyun Wu, Edouard C. Nice, Canhua Huang, Yuanyuan Zhang. Oxidative stress and diabetes: antioxidative strategies[J]. Front. Med., 2020, 14(5): 583-600.
[2] Dixin LI MM , Hongbing ZENG MD , Chunyang JI MM , . Effect of pirfenidone on renal tubulointerstitial fibrosis[J]. Front. Med., 2009, 3(3): 316-322.
[3] Qi RUI, Qin LU, Dayong WANG. Administration with Bushenkangshuai Tang alleviates UV irradiation- and oxidative stress-induced lifespan defects in nematode Caenorhabditis elegans[J]. Front Med Chin, 2009, 3(1): 76-90.
[4] HE Zhongye, GUO Renxuan, LI Yang, XIE Chengyao, LIU Nan, SONG Wen. Alleviation of cell damage in experimental ANP in rats by administration of chondroitin-sulfate reduces[J]. Front. Med., 2007, 1(1): 36-40.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed