Associate editor: A. Christopoulos
NADPH oxidases in the vasculature: Molecular features, roles in disease and pharmacological inhibition

https://doi.org/10.1016/j.pharmthera.2008.08.005Get rights and content

Abstract

Until the 1970s, reactive oxygen species (ROS) were considered merely harmful by-products of aerobic respiration and the driving force behind the evolution of an array of cellular antioxidant enzymes with the purpose of rapidly metabolising ROS to minimise their oxidising effects. However, the perception that ROS are only harmful to cells has since been questioned by a burgeoning body of evidence pointing to the existence of enzymes with the dedicated function of generating ROS. NADPH oxidases represent the only known family of enzymes whose sole purpose is to generate ROS. Members of this enzyme family are expressed across mammalian and non-mammalian cells, and influence a multitude of biological functions including host defence and redox signalling. However, although ROS are deliberately generated by NADPH oxidases during normal cell physiology, the observations that their expression and activity is markedly upregulated in the blood vessel wall in a number of cardiovascular ‘high-risk’ states (e.g. hypertension, hypercholesterolemia) implicates them in the oxidative stress that gives rise to artery disease and ultimately heart attacks and strokes. These observations highlight the fact that NADPH oxidases are important therapeutic targets in cardiovascular disease and that, hence, there is clearly a need for the development of selective inhibitors of these enzymes. Here we highlight the structural and biochemical characteristics of the NADPH oxidase family and then comprehensively review the literature on the currently available pharmacological inhibitors of these enzymes with a particular emphasis on their mechanisms of action, isoform selectivity and therapeutic potential in cardiovascular disease.

Section snippets

Introduction: reactive oxygen species: roles in physiology and pathophysiology

Life as an aerobic organism is a double-edged sword. On the one hand, oxygen is an abundant source of readily available and transferable energy that supports the complex array of biochemical reactions required for even the most basic of cellular functions. On the other hand, oxygen is an oxidising agent that could potentially damage the cellular infrastructure (e.g. nucleotides, proteins, lipids and carbohydrates) required for these very same processes. In actuality, ground state molecular

NADPH oxidases are major sources of reactive oxygen species in blood vessels

Within mammalian cells, several enzyme systems are capable of transferring electrons to molecular oxygen to produce superoxide, including NADH dehydrogenase and ubiquinone–cytochrome bc1 of the mitochondrial electron transport chain (Boveris and Cadenas, 1975, Turrens, 2003, Orrenius et al., 2007), nitric oxide synthases (NOS) (Vasquez-Vivar et al., 1998, Vasquez-Vivar and Kalyanaraman, 2000, Alp and Channon, 2004), cyclooxygenases (Simmons et al., 2004), lipoxygenases (Wittwer & Hersberger,

Molecular description of vascular NADPH oxidases

The purpose of the present discussion is not to provide an exhaustive review of the molecular biology and biochemistry of the various NADPH oxidase family members. Indeed this has been achieved recently in several excellent reviews (Vignais, 2002, Groemping and Rittinger, 2005, Sumimoto et al., 2005). Rather, here we aim to highlight some of the key molecular sites involved in activation of the NADPH oxidases, especially those, which are unique to specific isoforms, and may therefore be

Cellular and subcellular localisation of NADPH oxidase isoforms in the vascular wall

ROS have been implicated in numerous processes associated with atherogenesis including endothelial dysfunction, activation of inflammatory signalling pathways, upregulation of adhesion molecules, proliferation of vascular smooth muscle cells and oxidation of lipoproteins. Hence, identification of the cellular and enzymatic source(s) of ROS that contribute to each of these processes is essential for the development of effective therapies to reduce vascular oxidative stress and its many sequelae.

NADPH oxidases: causes and not just symptoms of vascular disease

The previous discussion, based largely on in vitro observations, suggest that Nox4-oxidases and constitutively active isoforms of Nox2-oxidases are important sources of intracellular ROS in the cells of the vascular wall under basal conditions. By contrast, in pathophysiological settings, inducible isoforms of Nox1- (in VSMCs) and Nox2- (in endothelial cells, fibroblasts and invading phagocytes) oxidases are upregulated. Importantly, these observations are generally supported in vivo. Hence, in

Inhibitors of NADPH oxidase and their limitations

The recent emergence of evidence implicating Nox enzymes in oxidative stress-related disorders of the cardiovascular system highlights the substantial clinical potential of Nox inhibitors. However, clearly much remains to be learned about the function and tissue distribution of these enzymes, and the stimuli which activate them. Furthermore, given that Nox enzymes have different subcellular locations and are likely to influence the redox milieu of cells, it is critical that Nox inhibitors do

Pleiotropic actions of three commonly employed cardiovascular drug classes

A number of clinically important drugs used for the treatment of hypertension, hypercholesterolaemia and coronary artery disease such as the statins, AT1 receptor antagonists and the ACE inhibitors have been shown to decrease NADPH oxidase-derived superoxide and ROS production, and thus are likely to mediate some of their beneficial actions via these mechanisms. The purpose of this section is not to provide an exhaustive review of these classes of drugs but to highlight briefly how these

Perspectives

From the previous discussion of the various currently available inhibitors, it is apparent that none of these compounds show any specificity towards a particular isoform of NADPH oxidase (Table 1). Nevertheless, these compounds are valuable for assessing the roles of these enzyme systems in in vitro and in vivo assays. For example, we suggest that if the intention of a study was to inhibit superoxide/ROS production generated by all NADPH oxidases within a particular in vitro system (i.e. cell

Concluding remarks

NADPH oxidases are ubiquitously expressed across many phyla and species and subserve a range of crucial redox-sensitive physiological and biological functions through the generation of ROS. The processes that lead to NADPH oxidase activation are complex and finely regulated; however, dysregulation of its expression and activation coupled with unregulated or inefficient ROS removal often leads to oxidative stress and an altered cellular phenotype, contributing to development of major

References (406)

  • BanfiB. et al.

    A Ca2+-activated NADPH oxidase in testis, spleen, and lymph nodes

    J Biol Chem

    (2001)
  • BelAibaR.S. et al.

    NOX5 variants are functionally active in endothelial cells

    Free Radic Biol Med

    (2007)
  • BetteP. et al.

    A comparative biochemical, pharmacological and immunological study of Clostridium novyi alpha-toxin, C. difficile toxin B and C. sordellii lethal toxin

    Toxicon

    (1991)
  • BeyE.A. et al.

    In vitro knockout of human p47phox blocks superoxide anion production and LDL oxidation by activated human monocytes

    J Lipid Res

    (2000)
  • BissonnetteS.A. et al.

    Phosphatidylinositol 3-phosphate-dependent and -independent functions of p40phox in activation of the neutrophil NADPH oxidase

    J Biol Chem

    (2008)
  • Bonnefont-RousselotD. et al.

    An intracellular modulation of free radical production could contribute to the beneficial effects of metformin towards oxidative stress

    Metabolism

    (2003)
  • BoverisA. et al.

    Mitochondrial production of superoxide anions and its relationship to the antimycin insensitive respiration

    FEBS Letters

    (1975)
  • BrandesR.P. et al.

    The vascular NADPH oxidase subunit p47phox is involved in redox-mediated gene expression

    Free Radic Biol Med

    (2002)
  • BrandschR. et al.

    Covalent flavinylation of 6-hydroxy-d-nicotine oxidase involves an energy-requiring process

    FEBS Letters

    (1987)
  • BurrittJ.B. et al.

    Phage display epitope mapping of human neutrophil flavocytochrome b558. Identification of two juxtaposed extracellular domains

    J Biol Chem

    (2001)
  • CachiaO. et al.

    Alpha-tocopherol inhibits the respiratory burst in human monocytes. Attenuation of p47phox membrane translocation and phosphorylation

    J Biol Chem

    (1998)
  • CaiH. et al.

    The vascular NAD(P)H oxidases as therapeutic targets in cardiovascular diseases

    Trends Pharmacol Sci

    (2003)
  • ChengG. et al.

    Homologs of gp91phox: cloning and tissue expression of Nox3, Nox4, and Nox5

    Gene

    (2001)
  • ChengG. et al.

    Nox1-dependent reactive oxygen generation is regulated by Rac1

    J Biol Chem

    (2006)
  • ChengG. et al.

    NOXO1, regulation of lipid binding, localization, and activation of Nox1 by the Phox homology (PX) domain

    J Biol Chem

    (2004)
  • ChengG. et al.

    Alternative mRNA splice forms of NOXO1: differential tissue expression and regulation of Nox1 and Nox3

    Gene

    (2005)
  • ChoiH.S. et al.

    Taurine chloramine inhibits PMA-stimulated superoxide production in human neutrophils perhaps by inhibiting phosphorylation and translocation of p47(phox)

    Int Immunopharmacol

    (2006)
  • ComeraC. et al.

    Gliotoxin from Aspergillus fumigatus affects phagocytosis and the organization of the actin cytoskeleton by distinct signalling pathways in human neutrophils

    Microbes Infect

    (2007)
  • CooperJ.M. et al.

    An animal model of mitochondrial myopathy: a biochemical and physiological investigation of rats treated in vivo with the NADH-CoQ reductase inhibitor, diphenyleneiodonium

    J Neurorol Sci

    (1988)
  • CuiX.L. et al.

    Expression of NADPH oxidase isoform 1 (Nox1) in human placenta: involvement in preeclampsia

    Placenta

    (2006)
  • D'AlessioA. et al.

    Caveolae participate in tumor necrosis factor receptor 1 signaling and internalization in a human endothelial cell line

    Am J Pathol

    (2005)
  • Deby-DupontG. et al.

    Resveratrol and curcumin reduce the respiratory burst of Chlamydia-primed THP-1 cells

    Biochem Biophys Res Commun

    (2005)
  • DeLeoF.R. et al.

    Processing and maturation of flavocytochrome b558 include incorporation of heme as a prerequisite for heterodimer assembly

    J Biol Chem

    (2000)
  • DiatchukV. et al.

    Inhibition of NADPH oxidase activation by 4-(2-Aminoethyl)-benzenesulfonyl fluoride and related compounds

    J Biol Chem

    (1997)
  • DidsburyJ.R. et al.

    Isoprenylation of the low molecular mass GTP-binding proteins rac 1 and rac 2: possible role in membrane localization

    Biochem Biophys Res Commun

    (1990)
  • DoussiereJ. et al.

    Activation of bovine neutrophil oxidase in a cell free system. GTP-dependent formation of a complex between a cytosolic factor and a membrane protein

    Biochem Biophys Res Commun

    (1988)
  • DuerrschmidtN. et al.

    Endothelin-1 induces NAD(P)H oxidase in human endothelial cells

    Biochem Biophys Res Commun

    (2000)
  • AbdelghaffarH. et al.

    The macrolide roxithromycin impairs NADPH oxidase activation and alters translocation of its cytosolic components to the neutrophil membrane in vitro

    Antimicrob Agents Chemother

    (2005)
  • AboA. et al.

    Activation of the NADPH oxidase involves the small GTP-binding protein p21rac1

    Nature

    (1991)
  • AgerberthB. et al.

    Amino acid sequence of PR-39. Isolation from pig intestine of a new member of the family of proline–arginine-rich antibacterial peptides

    Eur J Biochem

    (1991)
  • AgoT. et al.

    NAD(P)H oxidases in rat basilar arterial endothelial cells

    Stroke

    (2005)
  • AgoT. et al.

    Nox4 as the major catalytic component of an endothelial NAD(P)H oxidase

    Circulation

    (2004)
  • AgoT. et al.

    Phosphorylation of p47phox directs phox homology domain from SH3 domain toward phosphoinositides, leading to phagocyte NADPH oxidase activation

    PNAS

    (2003)
  • AgwuhK.N. et al.

    Pharmacokinetics and pharmacodynamics of the tetracyclines including glycylcyclines

    J Antimicrob Chemother

    (2006)
  • Al-MehdiA.B. et al.

    Endothelial NADPH oxidase as the source of oxidants in lungs exposed to ischemia or high K+

    Circ Res

    (1998)
  • AlpN.J. et al.

    Regulation of endothelial nitric oxide synthase by tetrahydrobiopterin in vascular disease

    Arterioscler Thromb Vasc Biol

    (2004)
  • AndersonR.

    Erythromycin and roxithromycin potentiate human neutrophil locomotion in vitro by inhibition of leukoattractant-activated superoxide generation and autooxidation

    J Infect Dis

    (1989)
  • AshrafianH. et al.

    Perhexiline

    Cardiovasc Drug Rev

    (2007)
  • BabiorB.M. et al.

    Biological defense mechanisms. The production by leukocytes of superoxide, a potential bactericidal agent

    J Clin Invest

    (1973)
  • BaeJ.S. et al.

    Receptors of the protein C activation and activated protein C signaling pathways are colocalized in lipid rafts of endothelial cells

    Proc Natl Acad Sci

    (2007)
  • Cited by (0)

    View full text