Journal of Pharmaceutical and Biomedical Sciences

NADPH Oxidase Inhibition of Novel Apocynin Dimer Derivatives

Shusheng Wang, Gaofang Wang, Xianyao Fu, Jianbin Qi, Lingchao Zhu, Sha Li, Jie Jiang

Abstract


Apocynin is a widely studied inhibitor of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, which has been regarded as a target for developing anti-oxidative and anti-inflammatory therapeutic agents. Diapocynin, dimer of apocynin, was found to have higher NADPH oxidase inhibition activity than apocynin. To seek for dimeric apocynin analogues of better efficacy, in our previous work, 14 novel apocynin dimer derivatives were designed and synthesized based on the chemical structure of apocynin dimer analogue JJA-D0. The biological activities of the derivatives were evaluated in RAW 264.7 cells. Allcompounds protected RAW 264.7 cells from lipopolysaccharide (LPS)-induced cytotoxicity, and significantly lowered down the intracellular ROS level induced by LPS. JJA-D26 were more potent than apocynin and JJA-D0, which retained 71.2% of cell viability, and decreased 24.8% of intracellular ROS level at the concentration of 200 ?M. JJA-D26 also greatly inhibited the expression of pro-inflammatory cytokine TNF-a, and significantly reduced the expression of subunits p47phox and gp91phox of NADPH oxidase complex (P < 0.05 and P < 0.01 respectively) compared to apocynin. This work may provide useful information for the research and development of potent anti-oxidation and anti-inflammation drugs targeting NADPH oxidase.

Keywords


apocynin dimer derivatives, NADPH oxidase, reactive oxygen species, anti-oxidation, anti-inflammation

Full Text:

References


Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev.2007;87:245–313.

Rodiño-Janeiro BK, Paradela-Dobarro B, Castiñeiras-Landeira MI, Raposeiras-Roubín S, González-Juanatey JR, Alvarez E. Current status of NADPH oxidase research in cardiovascular pharmacology.Vasc Health Risk Manag. 2013;9:401–428.

Johnson DK, Schillinger KJ, Kwait DM, Hughes CV, McNamara EJ, Ishmael F, et al. Inhibition of NADPH oxidase activation in endothelial cells by ortho-methoxy-substituted catechols.Endothelium. 2002;9:191–203.

Kanegae MP, Condino-Neto A, Pedroza LA, de Almeida AC, Rehder J, da Fonseca LM, et al. Diapocynin versus apocynin as pretranscriptional inhibitors of NADPH oxidase and cytokine production by peripheral blood mononuclear cells. Biochem Biophys Res Commun. 2010;393:551–554.

Babior BM. NADPH oxidase: an update. Blood. 1999;93:1464–1476.

Dumont M, Beal MF. Neuroprotective strategies involving ROS in Alzheimer disease. Free Radic Biol Med. 2011;51:1014–1026.

Madamanchi NR, Vendrov AE, Runge MS. Oxidative stress and vasular disease. Arterioscler Thromb Vasc Biol. 2005;25:29–38.

Guzik TJ, Harrison DG. Vascular NADPH oxidases as drug targets for novel antioxidant strategies. Drug Discov Today.2006;11:524–533.

Lambeth JD, Krause KH, Clark RA. NOX enzymes as novel targets for drug development. Semin Immunopathol. 2008;30:339–363.

Seo JE, Hasan M, Rahaman KA, Kang MJ, Jung BH, Kwon OS. A leading role for NADPH oxidase in an in-vitro study of experimental autoimmune encephalomyelitis. Mol Immunol.

;72:19–27.

Sharma M, Kaur T, Singla SK. Role of mitochondria and NADPH oxidase derived reactive oxygen species in hyperoxaluria induced nephrolithiasis: therapeutic intervention with combinatorial therapy of N-acetyl cysteine and apocynin.Mitochondrion. 2016;27:15–24.

Lin CC, Yang CC, Cho RL, Wang CY, Hsiao LD, Yang CM. Sphingosine 1-phosphate-induced ICAM-1 expression via NADPH oxidase/ROS-dependent NF-?B cascade on human pulmonary alveolar epithelial cells. Front Pharmacol. 2016;7:80.

Kim SY, Moon KA, Jo HY, Jeong S, Seon SH, Jung E, et al. Antiinflammatory effects of apocynin, an inhibitor of NADPH oxidase, in airway inflammation. Immunol Cell Biol. 2012;90:441–448.

Wang Q, Tompkins KD, Simonyi A, Korthuis RJ, Sun AY, Sun GY. Apocynin protects against global cerebral ischemiareperfusion-induced oxidative stress and injury in the gerbil hippocampus. Brain Res. 2006;1090:182–189.

Hamilton CA, Brosnan MJ, Al-Benna S, Berg G, Dominiczak AF. NAD(P)H oxidase inhibition improves endothelial function in rat and human blood vessels. Hypertension. 2002;40:755–762.

Hougee S, Hartog A, Sanders A, Graus YM, Hoijer MA, Garssen J, et al. Oral administration of the NADPH-oxidase inhibitor apocynin partially restores diminished cartilage proteoglycan synthesis and reduces inflammation in mice. Eur J Pharmacol.2006;531:264–269.

Hsu HC, Chang WM, Wu JY, Huang CC, Lu FJ, Chuang YW, et al. Folate deficiency triggered apoptosis of synoviocytes: role of overproduction of reactive oxygen species generated via NADPH oxidase/mitochondrial complex II and calcium perturbation.PLos One. 2016;11:e0146440.

Tang LL, Ye K, Yang XF, Zheng JS. Apocynin attenuates cerebral infarction after transient focal ischaemia in rats. J Int Med Res. 2007;35:517–522.

Cotter MA, Cameron NE. Effect of the NAD(P)H oxidase inhibitor, apocynin, on peripheral nerve perfusion and function in diabetic rats. Life Sci. 2003;73:1813–1824.

Check J, Byrd CL, Menio J, Rippe RA, Hines IN, Wheeler MD. Src kinase participates in LPS-induced activation of NADPH oxidase. Mol Immunol. 2010;47:756–762.

Qin L, Liu Y, Wang T, Wei SJ, Block ML, Wilson B, et al. NADPH oxidase mediates lipopolysaccharide-induced neurotoxicity and proinflammatory gene expression in activated microglia. J Biol Chem. 2004;279:1415–1421.

Kim ID, Ha BJ. Paeoniflorin protects RAW 264.7 macrophages from LPS-induced cytotoxicity and genotoxicity. Toxicol In Vitro. 2009;23:1014–1019.

Lu X, Wan S, Jiang J, Jiang X, Yang W, Yu P, et al. Synthesis and biological evaluations of novel apocynin analogues. Eur J Med Chem. 2011;46:2691–2698.

Jiang J, Kang H, Song X, Huang S, Li S, Xu J. A model of interaction between nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and apocynin analogues by docking method. Int J Mol Sci. 2013;14:807–817.

Barbieri SS, Cavalca V, Eligini S, Brambilla M, Caiani A, Tremoli E, et al. Apocynin prevents cyclooxygenase 2 expression in human monocytes through NADPH oxidase and glutathione redox-dependent mechanisms. Free Radic Biol Med.2004;37:156–165.

Blaser H, Dostert C, Mak TW, Brenner D. TNF and ROS crosstalk in inflammation. Trends Cell Biol. 2016;26:249–261.


Refbacks

  • There are currently no refbacks.


Copyright (c) 2017 Journal of Pharmaceutical and Biomedical Sciences

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.