Inhibition of human vascular NADPH oxidase by apocynin derived oligophenols

https://doi.org/10.1016/j.bmc.2009.05.061Get rights and content

Abstract

Enzymatic oxidation of apocynin, which may mimic in vivo metabolism, affords a large number of oligomers (apocynin oxidation products, AOP) that inhibit vascular NADPH oxidase. In vitro studies of NADPH oxidase activity were performed to identify active inhibitors, resulting in a trimer hydroxylated quinone (IIIHyQ) that inhibited NADPH oxidase with an IC50 = 31 nM. Apocynin itself possessed minimal inhibitory activity. NADPH oxidase is believed to be inhibited through prevention of the interaction between two NADPH oxidase subunits, p47phox and p22phox. To that end, while apocynin was unable to block the interaction of his-tagged p47phox with a surface immobilized biotinylated p22phox peptide, the IIIHyQ product strongly interfered with this interaction (apparent IC50 = 1.6 μM). These results provide evidence that peroxidase-generated AOP, which consist of oligomeric phenols and quinones, inhibit critical interactions that are involved in the assembly and activation of human vascular NADPH oxidase.

Introduction

Recent years have seen substantial improvement in our understanding of the role of superoxide anion (O2-) in eliciting oxidative stress and vascular diseases.1, 2, 3, 4, 5 The production of O2- is catalyzed by a variety of enzymes, including xanthine oxidase, cytochromes P450, lipoxygenase, enzymes in the mitochondrial respiratory chain, and NADPH oxidases.6 The latter, in particular, have been identified as the major source of O2- in vascular endothelial cells (VECs). Excessive production of O2- in VECs leads to increased oxidative stress and endothelial dysfunction. This in turn can result in a diverse array of cardiovascular diseases, including atherosclerosis, hypertension, diabetes, heart failure, stroke, and restenosis.1, 7, 8, 9, 10, 11

The most well-studied NADPH oxidase in humans is found in neutrophils. In the resting state, the neutrophil enzyme consists of at least six partially dissociated components.12 Tight regulation of NADPH oxidase activity is achieved by at least two mechanisms; the association of the cytosolic subunits and the modulation of reversible protein–protein and protein–membrane interactions.13 Despite near 100% homology with the neutrophil NADPH oxidase,14, 15 the exact assembly and activation of VEC NADPH oxidase is poorly understood.16 Nevertheless, current evidence suggests that phosphorylation of key serine residues in p47phox facilitates interaction of a Src homology 3 (SH3) domain of this protein with a proline-rich region (PRR) of p22phox, thereby forming the active membrane-associated NADPH oxidase complex17, 18 (Scheme 1).

The catalytic subunit of NADPH oxidase (Nox) has several isoforms and, at least three of them are expressed in VEC (Nox1, Nox2, and Nox4). Their precise activation mechanisms and cellular regulation remain unclear.19, 20 Nox1 appears to be of only minor importance in the generation of VEC reactive oxygen species (ROS). Nox4, however, is abundantly expressed in endothelial cells, more than Nox2.21 Nevertheless, Li et. al.21 showed that, despite low expression relative to Nox4, under starvation conditions Nox2 was upregulated ∼8-fold and, as a consequence of this nutrient deprivation-induced oxidative stress, the production of O2- increased ∼2.3-fold. Importantly, Nox2 requires assembly of p47phox with other cytosolic subunits prior to translocation to the membrane to form the active NADPH oxidase complex.22, 23

Due to the key role VEC NADPH oxidase appears to play in vascular diseases, identification of selective inhibitors is of great interest. Along these lines, several inhibitors have been identified, including nitrovasodilators,24 the flavonoid derivative 6,8-diallyl-5,7-dihydroxy-2-(2-allyl-3-hydroxy-4-methoxyphenyl)-1-H-benzo-[b]-pyran-4-one,25 and peptides such as the antibiotic PR-39.26 Interestingly, PRR regions are also known to bind polyphenols (such as flavonoids).27 It is not surprising, therefore, that phenolics have been found to have NADPH oxidase inhibitory activity.

Apocynin (4′-hydroxy-3′-methoxyacetophenone)28, 29 is a particularly interesting phenol that has been used as inhibitor of NADPH oxidase. While apocynin itself was found to have low activity in vitro29, 30 metabolism in vivo converts the phenol into active metabolites that inhibit the enzyme.30, 31, 32, 33, 34 This may be due to peroxidase catalysis29, 30, 35, 36 leading to disruption of the p47phox–p22phox interaction, which is required for translocation of the cytosolic enzyme components to the membrane leading to activation of the enzyme complex. In the current work, we demonstrate that several oligomeric apocynin oxidation products generated by peroxidases are extremely potent inhibitors of VEC NADPH oxidase in vitro. Moreover, a strong correlation exists between the inhibition of VEC NADPH oxidase in endothelial cell-based assays and disruption of the interaction of EC p47phox–p22phox in cell-free assays. These results provide additional mechanistic insight into the nature and function of active metabolites of apocynin.

Section snippets

Results and discussion

Under conditions of oxidative stress, overactive NADPH oxidase in the vasculature generates O2-, thereby leading to increased levels of H2O2. In the presence of peroxidases in the blood, for example, myeloperoxidase, and reducing substrates of peroxidases, such as phenols, peroxidatic reactions can occur. To mimic this scenario, we used a simple commercially available peroxidase from soybean (SBP) to catalyze the oxidation of apocynin in the presence of H2O2 following our earlier published

Experimental

Apocynin, SBP, solvents, H2O2, LDL, superoxide dismutase (SOD), low density lipoprotein (LDL), cytochrome c, Tween 20, 3,3′,5,5′-tetramethyl-benzidine (TMB), sodium caseinate, fetal bovine serum, heparin, and endothelial growth supplement were purchased from Sigma–Aldrich. Endothelial cells and medium were purchased from ATCC. Escherichia coli BL21 (DE3), IPTG and Ni-affinity column (Probond system) were purchased from Invitrogen. Antibodies were purchased from Upstate. High-affinity

Acknowledgments

We are grateful to NIH (AT002115) for the financial support of this project and to Dr. Christopher Bjornsson, Director of Microscopy and Imaging Core Facility (Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute), for his help with the confocal fluorescent microscopy analysis.

References and notes (48)

  • J. Li et al.

    Mol. Cell Cardiol.

    (2001)
  • Q. Li et al.

    J. Biol. Chem.

    (1999)
  • L. Walch et al.

    Atherosclerosis

    (2006)
  • B.M. Babior

    Blood

    (1999)
  • T. Finkel

    Curr. Opin. Cell Biol.

    (2003)
  • T.C. Meng et al.

    Mol. Cell.

    (2002)
  • J.W. Meyer et al.

    FEBS Lett.

    (2000)
  • J.M. Li et al.

    J. Biol. Chem.

    (2002)
  • K.D. Martyn et al.

    Cell. Signal.

    (2006)
  • J. Li et al.

    Free Radical Biol.

    (2007)
  • M.P.P. Kanegae et al.

    Biochem. Pharmacol.

    (2007)
  • J. Yu et al.

    Curr. Vasc. Pharmacol.

    (2008)
  • V.F. Ximenes et al.

    Arch. Biochem. Biophys.

    (2007)
  • J.M. Simons et al.

    Free Radical Biol. Med.

    (1990)
  • R. Takeya et al.

    J. Biol. Chem.

    (2003)
  • K. Lapouge et al.

    J. Biol. Chem.

    (2002)
  • I. Morozov et al.

    J. Biol. Chem.

    (1998)
  • J. Park

    Biochem. Biophys. Res. Commun.

    (1996)
  • I. Dahan et al.

    J. Biol. Chem.

    (2002)
  • R.M. Touyz

    Hypertension

    (2004)
  • J.A. Holland et al.

    J. Cell. Physiol.

    (1996)
  • B. Lassegue et al.

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2003)
  • Y. Taniyama et al.

    Hypertension

    (2003)
  • H. Seguchi et al.

    J. Electron Microsc.

    (2002)
  • Cited by (0)

    View full text