Volume 14 Issue 3
Mar.  2024
Turn off MathJax
Article Contents
Ke-Gang Linghu, Tian Zhang, Guang-Tao Zhang, Peng Lv, Wen-Jun Zhang, Guan-Ding Zhao, Shi-Hang Xiong, Qiu-Shuo Ma, Ming-Ming Zhao, Meiwan Chen, Yuan-Jia Hu, Chang-Sheng Zhang, Hua Yu. Small molecule deoxynyboquinone triggers alkylation and ubiquitination of Keap1 at Cys489 on Kelch domain for Nrf2 activation and inflammatory therapy[J]. Journal of Pharmaceutical Analysis, 2024, 14(3): 401-415. doi: 10.1016/j.jpha.2023.07.009
Citation: Ke-Gang Linghu, Tian Zhang, Guang-Tao Zhang, Peng Lv, Wen-Jun Zhang, Guan-Ding Zhao, Shi-Hang Xiong, Qiu-Shuo Ma, Ming-Ming Zhao, Meiwan Chen, Yuan-Jia Hu, Chang-Sheng Zhang, Hua Yu. Small molecule deoxynyboquinone triggers alkylation and ubiquitination of Keap1 at Cys489 on Kelch domain for Nrf2 activation and inflammatory therapy[J]. Journal of Pharmaceutical Analysis, 2024, 14(3): 401-415. doi: 10.1016/j.jpha.2023.07.009

Small molecule deoxynyboquinone triggers alkylation and ubiquitination of Keap1 at Cys489 on Kelch domain for Nrf2 activation and inflammatory therapy

doi: 10.1016/j.jpha.2023.07.009
Funds:

This study was supported by the Science and Technology Development Fund, Macao SAR (Grant Nos.: No.0159/2020/A3, No.0058/2020/AMJ, No.0096/2019/A2 and SKL-QRCM(UM)-2023-2025), the Research Committee of the University of Macau (Grant No.: MYRG2022-00189-ICMS), the Guangdong Provincial Special Fund for Marine Economic Development Project (Project No.: GDNRC[2021]48), National Natural Science Foundation of China (Grant No.: 82260801), and K.C. Wong Education Foundation (Grant No.: GJTD-2020-12). We greatly appreciate Ms. Parsa Dar for her language help.

  • Received Date: Apr. 18, 2023
  • Accepted Date: Jul. 13, 2023
  • Rev Recd Date: Jul. 13, 2023
  • Publish Date: Jul. 17, 2023
  • Activation of nuclear factor erythroid 2-related factor 2 (Nrf2) by Kelch-like ECH-associated protein 1 (Keap1) alkylation plays a central role in anti-inflammatory therapy. However, activators of Nrf2 through alkylation of Keap1-Kelch domain have not been identified. Deoxynyboquinone (DNQ) is a natural small molecule discovered from marine actinomycetes. The current study was designed to investigate the anti-inflammatory effects and molecular mechanisms of DNQ via alkylation of Keap1. DNQ exhibited significant anti-inflammatory properties both in vitro and in vivo. The pharmacophore responsible for the anti-inflammatory properties of DNQ was determined to be the α, β-unsaturated amides moieties by a chemical reaction between DNQ and N-acetylcysteine. DNQ exerted anti-inflammatory effects through activation of Nrf2/ARE pathway. Keap1 was demonstrated to be the direct target of DNQ and bound with DNQ through conjugate addition reaction involving alkylation. The specific alkylation site of DNQ on Keap1 for Nrf2 activation was elucidated with a synthesized probe in conjunction with liquid chromatography-tandem mass spectrometry. DNQ triggered the ubiquitination and subsequent degradation of Keap1 by alkylation of the cysteine residue 489 (Cys489) on Keap1-Kelch domain, ultimately enabling the activation of Nrf2. Our findings revealed that DNQ exhibited potent anti-inflammatory capacity through α, β-unsaturated amides moieties active group which specifically activated Nrf2 signal pathway via alkylation/ubiquitination of Keap1-Kelch domain, suggesting the potential values of targeting Cys489 on Keap1-Kelch domain by DNQ-like small molecules in inflammatory therapies.
  • loading
  • [1]
    A.M. Mahmoud, F.L. Wilkinson, M.A. Sandhu, et al., The interplay of oxidative stress and inflammation:Mechanistic insights and therapeutic potential of antioxidants, Oxid. Med. Cell. Longev. 2021 (2021) 1-4.
    [2]
    E.J. Ramos-Gonzalez, O.K. Bitzer-Quintero, G. Ortiz, et al., Relationship between inflammation and oxidative stress and its effect on multiple sclerosis, Neurologia (2021).
    [3]
    L. Baird, A.T. Dinkova-Kostova, The cytoprotective role of the Keap1-Nrf2 pathway, Arch. Toxicol. 85 (2011) 241-272.
    [4]
    T.W. Kensler, N. Wakabayashi, S. Biswal, Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway, Annu. Rev. Pharmacol. Toxicol. 47 (2007) 89-116.
    [5]
    S. Magesh, Y. Chen, L. Hu, Small molecule modulators of Keap1-Nrf2-ARE pathway as potential preventive and therapeutic agents, Med. Res. Rev. 32 (2012) 687-726.
    [6]
    M. Lu, J. Ji, Z. Jiang, et al., The Keap1-Nrf2-ARE pathway as a potential preventive and therapeutic target:An update, Med. Res. Rev. 36 (2016) 924-963.
    [7]
    A. Cuadrado, A.I. Rojo, G. Wells, et al., Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases, Nat. Rev. Drug Discov. 18 (2019) 295-317.
    [8]
    A.T. Dinkova-Kostova, W.D. Holtzclaw, T.W. Kensler, The role of Keap1 in cellular protective responses, Chem. Res. Toxicol. 18 (2005) 1779-1791.
    [9]
    A.T. Dinkova-Kostova, R.V. Kostov, P. Canning, Keap1, the cysteine-based mammalian intracellular sensor for electrophiles and oxidants, Arch. Biochem. Biophys. 617 (2017) 84-93.
    [10]
    G. Rachakonda, Y. Xiong, K.R. Sekhar, et al., Covalent modification at Cys151 dissociates the electrophile sensor Keap1 from the ubiquitin ligase CUL3, Chem. Res. Toxicol. 21 (2008) 705-710.
    [11]
    E.L. Mills, D.G. Ryan, H.A. Prag, et al., Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1, Nature 556 (2018) 113-117.
    [12]
    K.L. Rinehart Jr, H.B. Renfroe, The structure of nybomycin, J. Am. Chem. Soc. 83 (1961) 3729-3731.
    [13]
    J.S. Bair, R. Palchaudhuri, P.J. Hergenrother, Chemistry and biology of deoxynyboquinone, a potent inducer of cancer cell death, J. Am. Chem. Soc. 132 (2010) 5469-5478.
    [14]
    S. Li, X. Tian, S. Niu, et al., Pseudonocardians A-C, new diazaanthraquinone derivatives from a deap-sea actinomycete Pseudonocardia sp. SCSIO 01299, Mar. Drugs 9 (2011) 1428-1439.
    [15]
    X. Tian, L. Long, S. Li, et al., Pseudonocardia antitumoralis sp. nov., a deoxynyboquinone-producing actinomycete isolated from a deep-sea sediment, Int. J. Syst. Evol. Microbiol. 63 (2013), 1936.
    [16]
    E.I. Parkinson, P.J. Hergenrother, Deoxynyboquinones as NQO1-Activated Cancer Therapeutics, Acc. Chem. Res. 48 (2015) 2715-2723.
    [17]
    K. Saleem, W.A. Wani, A. Haque, et al., Synthesis, DNA binding, hemolysis assays and anticancer studies of copper (II), nickel (II) and iron (III) complexes of a pyrazoline-based ligand, Future Med. Chem. 5 (2013) 135-146.
    [18]
    I. Ali, W.A. Wani, K. Saleem, et al., Syntheses, DNA binding and anticancer profiles of L-glutamic acid ligand and its copper (II) and ruthenium (III) complexes, Med. Chem. 9 (2013) 11-21.
    [19]
    I. Ali, W.A. Wani, K. Saleem, et al., Anticancer metallodrugs of glutamic acid sulphonamides:in silico, DNA binding, hemolysis and anticancer studies, RSC Adv. 4 (2014) 29629-29641.
    [20]
    I. Ali, M. Suhail, M. Fazil, et al., Anti-cancer and Anti-oxidant Potencies of Cuscuta reflexa Roxb. Plant Extracts, Am. J. Adv. Drug Deliv. 14 (2019) 92-113.
    [21]
    I. Ali, M. Alsehli, L. Scotti, et al., Progress in polymeric nano-medicines for theranostic cancer treatment, Polymers (Basel). 12 (2020), 598.
    [22]
    I. Ali, W.A. Wani, A. Haque, et al., Glutamic acid and its derivatives:candidates for rational design of anticancer drugs, Future Med. Chem. 5 (2013) 961-978.
    [23]
    I. Ali, W. A. Wani, K. Saleem, et al., Thalidomide:A Banned Drug Resurged into Future Anticancer Drug, Curr. Drug Ther. 7 (2012) 13-23.
    [24]
    K.-G. Linghu, G.D. Zhao, W. Xiong, et al., Comprehensive comparison on the anti-inflammatory effects of three species of Sigesbeckia plants based on NF-κB and MAPKs signal pathways in vitro, J. Ethnopharmacol. 250 (2020), 112530.
    [25]
    K.-G. Linghu, Q.S. Ma, G.D. Zhao, et al., Leocarpinolide B attenuates LPS-induced inflammation on RAW264.7 macrophages by mediating NF-κB and Nrf2 pathways, Eur. J. Pharmacol. 868 (2020), 172854.
    [26]
    T. Toyama, Y. Shinkai, T. Kaji, et al., A convenient method to assess chemical modification of protein thiols by electrophilic metals, J. Toxicol. Sci. 38 (2013) 477-484.
    [27]
    K.-G. Linghu, S.H. Xiong, G.D. Zhao, et al., Sigesbeckia orientalis L. extract alleviated the collagen type II-induced arthritis through inhibiting multi-target-mediated synovial hyperplasia and inflammation, Front. Pharmacol. 11 (2020), 547913.
    [28]
    A. Paine, B. Eiz-Vesper, R. Blasczyk, et al., Signaling to heme oxygenase-1 and its anti-inflammatory therapeutic potential, Biochem. Pharmacol. 80 (2010) 1895-1903.
    [29]
    S. Ryter, Therapeutic potential of heme oxygenase-1 and carbon monoxide in acute organ injury, critical illness, and inflammatory disorders, Antioxidants 9 (2020), 1153.
    [30]
    J.A. Araujo, M. Zhang, F. Yin, Heme oxygenase-1, oxidation, inflammation, and atherosclerosis, Front. Pharmacol. 3 (2012), 119.
    [31]
    D.E. Stack, J.A. Conrad, B. Mahmud, Structural identification and kinetic analysis of the in vitro products formed by reaction of bisphenol A-3, 4-quinone with N-acetylcysteine and glutathione, Chem. Res. Toxicol. 31 (2018) 81-87.
    [32]
    Y. Cheng, J. Rong, Therapeutic potential of heme oxygenase-1/carbon monoxide system against ischemia-reperfusion injury, Curr. Pharm. Des. 23 (2017) 3884-3898.
    [33]
    S. Nishikawa, Y. Inoue, Y. Hori, et al., Anti-inflammatory activity of kurarinone involves induction of HO-1 via the KEAP1/Nrf2 pathway, Antioxidants 9 (2020), 842.
    [34]
    A.L. Eggler, G. Liu, J.M. Pezzuto, et al., Modifying specific cysteines of the electrophile-sensing human Keap1 protein is insufficient to disrupt binding to the Nrf2 domain Neh2, Proc. Natl. Acad. Sci. USA 102 (2005) 10070-10075.
    [35]
    Y. Cheng, L. Cheng, X. Gao, et al., Covalent modification of Keap1 at Cys77 and Cys434 by pubescenoside a suppresses oxidative stress-induced NLRP3 inflammasome activation in myocardial ischemia-reperfusion injury, Theranostics 11 (2021) 861-877.
    [36]
    N. Robledinos-Anton, R. Fernandez-Gines, G. Manda, et al., Activators and inhibitors of NRF2:A review of their potential for clinical development, Oxid. Med. Cell. Longev. 2019 (2019) 1-20.
    [37]
    U. Schulze-Topphoff, M. Varrin-Doyer, K. Pekarek, et al., Dimethyl fumarate treatment induces adaptive and innate immune modulation independent of Nrf2, Proc. Natl. Acad. Sci. U. S. A. 113 (2016) 4777-4782.
    [38]
    E.A. Mills, M.A. Ogrodnik, A. Plave, et al., Emerging understanding of the mechanism of action for dimethyl fumarate in the treatment of multiple sclerosis, Front. Neurol. 9 (2018), 5.
    [39]
    Z. Li, P. Hao, L. Li, et al., Design and synthesis of minimalist terminal alkyne-containing diazirine photo-crosslinkers and their incorporation into kinase inhibitors for cell- and tissue-based proteome profiling, Angew. Chem. 125 (2013) 8713-8718.
    [40]
    F. Hong, K.R. Sekhar, M.L. Freeman, et al., Specific patterns of electrophile adduction trigger Keap1 ubiquitination and Nrf2 activation, J. Biol. Chem. 280 (2005) 31768-31775.
    [41]
    L. Baird, D. Lleres, S. Swift, et al., Regulatory flexibility in the Nrf2-mediated stress response is conferred by conformational cycling of the Keap1-Nrf2 protein complex, PNAS 110 (2013) 15259-15264.
    [42]
    Y. Ying, J. Jin, L. Ye, et al., Phloretin prevents diabetic cardiomyopathy by dissociating Keap1/Nrf2 complex and inhibiting oxidative stress, Front. Endocrinol. 9 (2018), 774.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)

    Article Metrics

    Article views (245) PDF downloads(39) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return