939
Views
57
CrossRef citations to date
0
Altmetric
Review Article

The molecular sources of reactive oxygen species in hypertension

, , , &
Pages 70-77 | Received 09 Oct 2007, Accepted 19 Feb 2008, Published online: 08 Jul 2009

Abstract

In both animal models and humans, increased blood pressure has been associated with oxidative stress in the vasculature, i.e. an excessive endothelial production of reactive oxygen species (ROS), which may be both a cause and an effect of hypertension. In addition to NADPH oxidase, the best characterized source of ROS, several other enzymes may contribute to ROS generation, including nitric oxide synthase, lipoxygenases, cyclo‐oxygenases, xanthine oxidase and cytochrome P450 enzymes. It has been suggested that also mitochondria could be considered a major source of ROS: in situations of metabolic perturbation, increased mitochondrial ROS generation might trigger endothelial dysfunction, possibly contributing to the development of hypertension. However, the use of antioxidants in the clinical setting induced only limited effects on human hypertension or cardiovascular endpoints. More clinical studies are needed to fully elucidate this so called “oxidative paradox” of hypertension.

Introduction

A large body of evidence supports the involvement of endothelial dysfunction in the pathogenesis of both arterial hypertension and atherothrombosis Citation[1], Citation[2]. Endothelial dysfunction is characterized by a decreased bioavailability of nitric oxide (NO), which, under normal conditions, mediates both endothelium‐dependent vasodilation and the regulation of other protective properties of endothelial cells. In human hypertension, endothelial dysfunction would be mainly associated with an increased NO degradation, due to NO interaction with superoxide anions, while in experimental models of hypertension, a decreased NO production has also been observed Citation[3]. A defect of NO activity causes an imbalance in favor of endothelial contracting factors, such as endothelin‐1 (ET‐1), angiotensin II (Ang II), vasoconstrictor prostanoids and superoxide anions Citation[4].

In both animal models and humans, increased blood pressure (BP) and endothelial dysfunction have been associated with increased oxidative stress, i.e. an excessive endothelial production of reactive oxygen (ROS) and nitrogen (RNS) species Citation[5], Citation[6]. Oxidative stress contributes to vascular pathophysiology by promoting increased vascular tone, inflammation, cell growth, as well as the activation of matrix metalloproteinases and the deposition of extracellular matrix proteins, which are processes associated with the vascular phenotype of hypertension Citation[1]. However, it still remains unclear whether elevated levels of ROS (including superoxide, hydrogen peroxide and hydroxil anion) and RNS (such as peroxynitrite) initiate the development of hypertension, are a consequence of hypertension, or both Citation[7].

The present paper will mainly deal with the molecular mechanisms underlying the generation of ROS in excess in endothelial cells, with particular consideration for their implications in hypertension.

Oxidative stress and hypertension

Oxidative stress, an excessive production of ROS outstripping antioxidant defense mechanisms, has been associated with several pathophysiological processes affecting the cardiovascular system including hypertension, hypercholesterolemia, diabetes, hyperhomocysteinemia and cigarette smoking Citation[8–11].

In animal models, oxidative stress has been observed in spontaneous hypertensive rats Citation[12], renovascular hypertension Citation[13], salt‐sensitive hypertension Citation[14], and in a rat model of obesity‐induced hypertension Citation[15]. Although its pathogenesis is complex and multifactorial, human hypertension may be also considered a state of increased oxidative stress Citation[16], Citation[17], associated with a decreased antioxidant activity and a reduced ability to scavenge oxygen‐derived free radicals Citation[6], Citation[18].

Guzik et al. Citation[19] have shown an association involving cardiovascular risk factors, vascular NADPH‐dependent superoxide production and reduced Ach‐induced vasorelaxation. However, other authors have shown a dissociation between altered superoxide production and BP Citation[20–22]. Moreover, results can vary depending on the marker of oxidative stress being investigated Citation[6]. For example, Kumar and Das Citation[23] have observed a higher superoxide and hydrogen peroxide production in hypertensive patients. Similar results have also been obtained by Lacy et al. Citation[24], who found a significant correlation between hydrogen peroxide production and systolic BP. Furthermore, in untreated hypertensives, reductions in superoxide dysmutase (SOD) and glutathione peroxidase activity have been observed compared with controls Citation[25], while the treatment of hypertensive patients with candesartan or amlodipine caused a significant reduction in oxidized glutathione Citation[26]. On the other hand, studies with more specific markers of oxidative stress, such as F2‐isoprostanes measured in plasma and/or urine, have demonstrated no differences between untreated hypertensives and controls Citation[27], Citation[28]. Thus, human studies have not been as conclusive as animal studies.

Redon et al. Citation[29] have recently studied the oxidative status, antioxidant activities and ROS byproducts in whole blood and mononuclear peripheral cells in untreated hypertensive subjects and in normotensive volunteers. The blood and mononuclear cells from hypertensive patients exhibited significant deficiencies of physiological antioxidants and an accumulation of large quantities of peroxidation and DNA oxidation byproducts. However, no significant relationship between oxidative stress and BP values was observed. These data may indicate that factors other than BP alone, inherent to the hypertensive status (such as hyperadrenergic reactivity, enhanced activity of the renin–angiotensin system or hyperinsulinemia), may be responsible for the altered oxidative status, which, in this case, would be a consequence, rather than a causative factor, of hypertension Citation[29].

Molecular sources of ROS in hypertension

Although NADPH oxidase is considered the best characterized cellular source of ROS, there are a number of further possible sources of free radical production within the vasculature. These include NO synthase (NOS), lipoxygenases, cyclo‐oxygenases, xanthine oxidase (XO) and cytochrome P450 enzymes (CYP450) Citation[30], Citation[31]. Mitochondria may also be a significant source of superoxide in hypertension.

Nicotamide adenine dinucleotide phosphate reduced form (NADPH)‐oxidases (Noxs)

Since increased Nox activity has been reported as a major source of superoxide, it may be relevant to consider the potential genetic influences on this enzymatic system, which is specifically designed to modulate cellular redox signaling Citation[22]. Moreno et al. Citation[32] have recently described a variant of the human p22phox (an essential Nox subunit) gene promoter, the 930A/G polymorphism, which is associated with essential hypertension. Further studies have confirmed that among the polymorphisms of the gene encoding p22phox, the 930A/G polymorphism may significantly contribute to the susceptibility of hypertensive patients to oxidative stress Citation[33], Citation[34].

The role of Noxs in cardiovascular health and disease has recently and extensively been reviewed by Cave et al. Citation[22]. Nox isoforms represent an enzyme family containing at least five members, which are known to differ in relation to cell‐ specific expression and intracellular sites of localization Citation[35]. It has recently been suggested that the overexpression of the Nox1 isoform in SMCs may produce a basal increase in vascular ROS production, an Ang II‐induced hypertensive response and medial artery hypertrophy Citation[36]. Conversely, in Nox1‐null mice Ang II‐induced hypertension is mild and without effect on medial hypertrophy Citation[37]. These findings are consistent with the data of Wang et al. Citation[38] showing that a deficit in another NADPH oxidase isoform (Nox2), although without effect on Ang II‐induced hypertension, causes a significant attenuation of medial hypertrophy in mice. Thus, these data could provide important insights on the possibility that abnormalities of vascular structure may precede the development of hypertension Citation[35]. In addition, these studies support the idea that Noxs may be not only an important source of ROS within the vessel wall, but also play a more specific role in the pathogenesis of hypertension.

Xanthine oxidoreductase

Xanthine oxidoreductase (XOR) is a metalloflavoprotein enzyme that exists in two interconvertible although functionally distinct forms, namely xanthine dehydrogenase (XD) and XO. Of them, only the latter generates superoxide by oxidation of hypoxanthine to xanthine, and xanthine to urate (which requires the reduction of molecular O2) Citation[39–42].

XO is activated by proinflammatory mediators and is upregulated by NADPH oxidase activation. In vitro experiments with endothelial cells have shown that incubation with apocynin or disruption of the p47phox gene markedly decrease XO expression and activity Citation[43]. However, the precise in vivo regulation of endothelial XO is still unclear Citation[40].

The putative contribution of XO to ROS elevation in hypertension has been assessed using specific inhibitors, such as allopurinol and oxypurinol. For example, oxypurinol increases endothelial‐dependent arterial relaxation in rats fed a high‐salt diet Citation[44], in spontaneously hypertensive rats (SHR) Citation[45], and in rats overexpressing renin and angiotensinogen genes Citation[46]. These data suggest that XO may be another source of ROS elevation in several disease settings associated with impaired vascular function Citation[39], Citation[42]. XO activation may also contribute to the genesis of redox signaling dysfunction leading to altered gene–protein expression Citation[47]. Finally, polymorphisms of the XO gene have recently been found to be associated with BP levels Citation[48].

Table I. Targets and actions of NO.

Endothelial NOS (eNOS)

eNOS, like all NOSs, catalyzes the oxidation of l‐arginine to l‐citrulline and NO Citation[49]. Under abnormal circumstances, eNOS can become “uncoupled” and reduce molecular oxygen (rather than transferring electrons to l‐arginine), thereby generating superoxide Citation[50–54]. The essential NOS cofactor tetrahydrobiopterin (BH4) plays a key role in regulating NOS function by coupling the reduction of O2 to l‐arginine oxidation Citation[55], Citation[56]. Thus, a relative deficiency of BH4 or arginine may lead to eNOS uncoupling. Further, an increased ROS production by NADPH oxidases can cause the oxidative loss of BH4, with following amplification of oxidative stress through decreased NO production and increased NOS‐dependent superoxide generation Citation[57]. Peroxinitrite itself (a product of the interaction between excess superoxide and NO) may also oxidize BH4, causing eNOS uncoupling Citation[58]. Increased production of superoxide rather than NO, and increased generation of peroxynitrite, have been implicated in the pathophysiology of several cardiovascular diseases, including hypertension Citation[53], Citation[40].

Cytochrome P450 (CYP) enzymes

In a recent editorial Citation[59], Fleming has briefly discussed the available data concerning the link between CYP enzymes and the regulation of vascular tone. CYP enzymes fall into two classes: the epoxygenases, which generate vasodilator epoxy‐eicosa‐trienoic acids (EETs), and the ω/ω‐1 hydroxylases, which generate the vasoconstrictor 20‐hydroxy‐eicosa‐tetraenoic acid (20‐HETE).

Since EETs act as NO, a decrease in their formation and tissue levels can contribute to the development of some forms of experimental hypertension Citation[60], including salt‐induced hypertension in salt‐sensitive Dahl rats Citation[61]. Moreover, during the CYP reaction cycle, when the electrons of the central heme iron are transferred on the activated bound oxygen molecule in a NADPH‐dependent reaction, the CYP epoxygenases generate superoxide anions, hydrogen radicals and hydroxyl radicals Citation[60]. Thus, the CYP epoxygenases expressed in endothelial cells could contribute significantly to the production of ROS within the vascular wall. This production of ROS would be sufficient to impair NO‐mediated vasorelaxation, and to elevate the activity of nuclear factor kB (Nf‐kB) and the expression of vascular cell adhesion molecules (VCAM) Citation[30].

Changes in the production of 20‐HETE, which is currently characterized as a vasoconstrictor metabolite of arachidonic acid, may also be linked to the development of experimental Citation[31], Citation[62] and human hypertension Citation[63], Citation[64]. In particular, Ward et al. Citation[63] found that urinary 20‐HETE is associated with endothelial dysfunction in humans with blunted flow‐induced vasodilatation of the brachial artery. More recently, Gainer et al. Citation[64] reported that a functional variant of the human renal 20‐HETE synthase, CYP 4A11, was associated with essential hypertension in a white population from Tennessee. These studies support the hypothesis that abnormalities in the 20‐HETE pathway could play a role in various forms of human hypertension.

Mitochondria as a source of ROS in hypertension

During aerobic metabolism in the mitochondrial respiratory chain, a small quantity of O2 may be incompletely reduced, and superoxide be produced by complexes I (NADH coenzyme Q reductase) and III (ubiquinol Cyt C reductase). The rate of ROS generation can be modulated by the mitochondrial inner membrane potential Citation[65], Citation[66]. Indeed, mitochondria can be considered a major source of ROS. On the other hand, the increase in oxidative stress associated with atherosclerosis risk factors might cause a mitochondrial oxidative damage, so that mitochondria can also be important targets for ROS Citation[67], Citation[68].

An excessive production of ROS in mitochondria will damage lipids, proteins and mitochondrial DNA (mtDNA), which is located close to the inner membrane and is not protected by histone proteins, as is the case for nuclear DNA Citation[69], Citation[70]. The accumulation of mtDNA mutations can cause cell dysfunction by inducing the synthesis of defective protein subunits of the respiratory chain Citation[71], with further oxidative stress and ROS production. A vicious cycle may therefore be generated, which leads to progressive accumulation of mutations and oxidative damage to mtDNA Citation[72] Interestingly, NO itself at high concentrations can damage mtDNA by stimulating the mitochondrial production of superoxide, hydrogen peroxide and peroxinitrite Citation[73].

mtDNA mutations may also be inherited. For example, variants of the mitochondrial genome have been reported by Schwartz et al. in both African American and white hypertensive families Citation[74], and an excess of maternal transmission of predisposition to hypertension has been found in hypertensive families from three ethnic groups Citation[75].

The participation of mithocondria in the pathogenesis of hypertension is also suggested by the involvement of mitochondrial uncoupling proteins (UCPs) in experimental and human hypertensive states. In particular, mice with doxycycline‐inducible expression of UCP1 in arterial walls develop hypertension and dietary atheroscerosis. UCP1 expression also increases superoxide production and decreases the availability of NO Citation[76]. These results are in agreement with the study by Douette et al. Citation[77], which showed that UCP1 has a dual influence on free radical generation. On the one side, FFA‐activated UCP1 is able to decrease superoxide anion production, demonstrating that a decrease in ROS generation is an obligatory outcome of UCP1 activity. On the other side, an increase in UCP1 content was concomitant with an increase in the basal release of superoxide by mitochodria as a side consequence of the overall increase in oxidative metabolism.

As far as UCP2 is concerned, its content in kidney mitochondria of SHR was lower than in normotensive controls Citation[78], suggesting that a major function of UCP2 would be to increase the proton conductance across the inner mitochondrial membrane, thus reducing superoxide production Citation[79]. Accordingly, a common polymorphism of the UCP2 gene was associated with hypertension in a Japanese population, and with hypertension and obesity in Caucasians Citation[80].

The possible mitochondrial involvement in hypertension is supported by several studies in both animals and humans. Hypertension is associated with the deterioration of mitochondrial energy production in SHRs Citation[78], Citation[81], Citation[82] and mice Citation[76]. The occurrence of mitochondrial dysfunction has been associated with decreases in cytochrome oxidase activity Citation[83], ATP production Citation[76] and inorganic phosphate translocator activity Citation[84].

Similarly to eNOS, mitochondrial NOS (mtNOS) might be involved in hypertension. This hypothesis was not confirmed after measurement of brain mtNOS in SHR Citation[85]. However, further studies will be required to assess the physiological significance of vascular mtNOS and to elucidate its regulatory effects on cellular energy metabolism in mitochondria Citation[86].

Other possible sources of ROS in hypertension

Myeloperoxidase (MPO) may be an important source of ROS in the presence of vascular damage, contributing to exacerbate vascular diseases Citation[87].

Transition metals, like iron and copper, are strong catalysts for oxidation reactions in the presence of hydroperoxides. However, the blood concentration of free transition metals is rather low in vivo. In addition to free iron and copper, biological forms of iron, such as heme, hemoglobin and myoglobin all have the potential to catalyze oxidative reactions in the vessel wall and to oxidize low‐density lipoprotein Citation[88].

Conclusions

Experimental evidence and clinical studies suggest that oxidative stress may be involved in the pathogenesis of hypertension. However, whether elevated levels of ROS initiate the development of hypertension or are a consequence of the disease process is not yet completely understood Citation[6], Citation[7]. Potential sources of free radicals within the vasculature that could be implicated in hypertension include mitochondria, NADPH oxidase, XOR, uncoupled endothelial NOSs, and CYP enzymes. In addition, enzymes such as lipoxygenases and cycloxygenases may also generate ROS, but the evidence for their participation in redox signaling is still scarce Citation[89].

Activation of the renin–angiotensin system has been proposed as a mediator of NADPH oxidase activation and ROS production. In particular, Ang II stimulation of AT1 receptors in the vascular wall leads to the activation of NADPH oxidase. The resultant oxidative stress is considered a unifying mechanism for hypertension and atherosclerosis Citation[90]. Ang II can also directly regulate NADPH activation by phosphorylating and translocating p47phox to the cell membrane Citation[91]. Therefore, it is possible that some effects of the antihypertensive and vasculoprotective actions of AT1 receptor blockers and ACE inhibitors could be attributed to NADPH oxidase inhibition and decreased ROS production Citation[92–94].

Finally, also mechanical stretch may induce p47phox membrane translocation and NADPH oxidase activation in vascular smooth muscle cells Citation[95]. Stretch‐stimulated NADPH oxidase might play an important role in hypertension‐induced vessel wall remodeling through the activation of metalloproteinase 2 (MMP‐2), which is involved in elastic lamina destruction and in the process of arterial stiffening in humans Citation[96], Citation[97].

Oxidative stress may contribute to the pathogenesis and/or maintenance of hypertension according to several possible mechanisms. Ward and Croft Citation[6] have outlined these molecular mechanisms as follows: inactivation of NO by superoxide, generation of vasoconstrictor lipid peroxidation products such as F2‐isoprostanes, depletion of BH4 (the cofactor of NOS), as well as structural and functional alterations within the vascular wall, as recently reported by Kals et al. in patients with peripheral arterial disease Citation[98].

Molecular processes underlying ROS‐induced vascular changes involve activation of redox‐sensitive signaling pathways. Indeed, superoxide and H2O2 stimulate mitogen‐activated protein kinases, tyrosine kinases and transcription factors such as nuclear factor kB (NFkB), activator protein‐1 (AP‐1) and hypoxia‐inducible factor‐1 (HIF‐1) Citation[99], and inactivate protein tyrosine phosphatase Citation[100], Citation[101]. ROS also increase Ca2+, and upregulate protoncogenes and the expression of proinflammatory genes Citation[100].

Despite experimental evidence of the importance of oxidative stress in hypertension‐dependent vascular damage, only a few clinical investigations reported limited effects of antioxidant vitamins on BP Citation[99], Citation[102], Citation[103]. The divergence between experimental models and clinical data has been defined “the oxidative paradox” Citation[104]. Patterson et al. Citation[104] have recently examined several possible explanations for the rather disappointing results of antioxidants. For example, the endpoints of most trials are usually ischemic events: these might be too far from the initial development of hypertension and atherosclerosis, which may typically involve oxidative stress. Another important reason may be that in all clinical trials patients were not recruited on the basis of an elevated ROS formation. In other words, the patients without a significant oxidative stress might not benefit from antioxidant therapy, which highlights the need to identify the high‐risk population with reliable biomarkers. Thus, more clinical studies on antioxidants or other ROS modulators are needed, possibly involving cohorts with evidence of oxidative stress, in order to fully elucidate whether an excess of ROS does play a significant role in the development of human hypertension and its cardiovascular complications.

References

  • Paravicini T. M., Touyz R. M. Redox signalling in hypertension. Cardiovasc Res 2006; 71: 247–258
  • Russo G., Leopold J. A., Loscalzo J. Vasoactive substances: Nitric oxide and endothelial dysfunction in atherosclerosis. Vascul Pharmacol 2002; 38: 259–269
  • Thuillez C., Richard V. Targeting endothelial dysfunction in hypertensive subjects. J Hum Hypertens 2005; 19: 21–25
  • Endemann D. H., Schiffrin E. L. Endothelial dysfunction. J Am Soc Nephrol 2004; 15: 1983–1992
  • Parik T., Allikmets K., Teesalu R., Zilmer M. Evidence for oxidative stress in essential hypertension: Perspective for antioxidant therapy. J Cardiovasc Risk 1996; 3: 49–54
  • Ward N. C., Croft K. D. Hypertension and oxidative stress. Clin Exp Pharmacol Physiol 2006; 33: 872–876
  • Chen X., Touyz R. M., Park J. B., Schiffrin E. L. Antioxidant effects of vitamins C and E are associated with altered activation of vascular NADPH oxidase and superoxide dismutase in stroke‐prone SHR. Hypertension 2001; 38: 606–611
  • Steinberg D., Witztum J. L. Is the oxidative modification hypothesis relevant to human atherosclerosis? Do the antioxidant trials conducted to date refute the hypothesis?. Circulation 2002; 105: 2107–111
  • Chisolm G. M., Steinberg D. The oxidative modification hypothesis of atherogenesis: An overview. Free Radic Biol Med 2000; 28: 1815–1826
  • Cai H., Harrison D. G. Endothelial dysfunction in cardiovascular diseases: The role of oxidant stress. Circ Res 2000; 87: 840–844
  • Feletou M., Vanhoutte P. M. Endothelial dysfunction: A multifaceted disorder. Am J Physiol Heart Circ Physiol 2006; 291: 985–1002
  • Wu L., Juurlink B. H. Increased methylglyoxal and oxidative stress in hypertensive rat vascular smooth muscle cells. Hypertension 2002; 39: 809–814
  • Lerman L. O., Nath K. A., Rodriguez‐Porcel M., Krier J. D., Schwartz R. S., Napoli C., et al. Increased oxidative stress in experimental renovascular hypertension. Hypertension 2001; 37: 541–546
  • Trolliet M. R., Rudd M. A., Loscalzo J. Oxidative stress and renal dysfunction in salt‐sensitive hypertension. Kidney Blood Press Res 2001; 24: 116–123
  • Dobrian A. D., Davies M. J., Schriver S. D., Lauterio T. J., Prewitt R. L. Oxidative stress in a rat model of obesity‐induced hypertension. Hypertension 2001; 37: 554–560
  • Romero J. C., Reckelhoff J. F. Role of angiotensin and oxidative stress in essential hypertension. Hypertension 1999; 34: 943–949
  • Raij L. Nitric oxide in hypertension: Relationship with renal injury and left ventricular hypertrophy. Hypertension 1998; 31: 189–193
  • McIntyre M., Bohr D. F., Dominiczak A. F. Endothelial function in hypertension: The role of superoxide anion. Hypertension 1999; 34: 539–545
  • Guzik T. J., West N. E., Black E., McDonald D., Ratnatunga C., Pillai R., et al. Vascular superoxide production by NADPH oxidase: Association with endothelial dysfunction and clinical risk factors. Circ Res 2000; 86: 85–90
  • Li L., Fink G. D., Watts S. W., Northcott C. A., Galligan J. J., Pagano P. J., et al. Endothelin‐1 increases vascular superoxide via endothelin(A)‐NADPH oxidase pathway in low‐renin hypertension. Circulation 2003; 107: 1053–1058
  • Somers M. J., Mavromatis K., Galis Z. S., Harrison D. G. Vascular superoxide production and vasomotor function in hypertension induced by deoxycorticosterone acetate‐salt. Circulation 2000; 101: 1722–1728
  • Cave A. C., Brewer A. C., Narayanapanicker A., Ray R., Grieve D. J., Walker S., et al. NADPH oxidases in cardiovascular health and disease. Antioxid Redox Signal 2006; 8: 691–728
  • Kumar K. V., Das U. N. Are free radicals involved in the pathobiology of human essential hypertension?. Free Radic Res Commun 1993; 19: 59–66
  • Lacy F., Kailasam M. T., O'Connor D. T., Schmid‐Schonbein G. W., Parmer R. J. Plasma hydrogen peroxide production in human essential hypertension: Role of heredity, gender, and ethnicity. Hypertension 2000; 36: 878–884
  • Pedro‐Botet J., Covas M. I., Martin S., Rubies‐Prat J. Decreased endogenous antioxidant enzymatic status in essential hypertension. J Hum Hypertens 2000; 14: 343–345
  • Muda P., Kampus P., Zilmer M., Ristimäe T., Fischer K., Zilmer K., et al. Effect of antihypertensive treatment with candesartan or amlodipine on glutathione and its redox status, homocysteine and vitamin concentrations in patients with essential hypertension. J Hypertens 2005; 23: 105–112
  • Cracowski J. L., Baguet J. P., Ormezzano O., Bessard J., Stanke‐Labesque F., Bessard G., et al. Lipid peroxidation is not increased in patients with untreated mild‐to‐moderate hypertension. Hypertension 2003; 41: 286–288
  • Ward N. C., Hodgson J. M., Puddey I. B., Mori T. A., Beilin L. J., Croft K. D. Oxidative stress in human hypertension: Association with antihypertensive treatment, gender, nutrition, and lifestyle. Free Radic Biol Med 2004; 36: 226–232
  • Redon J., Oliva M. R., Tormos C., Giner V., Chaves J., Iradi A., et al. Antioxidant activities and oxidative stress byproducts in human hypertension. Hypertension 2003; 41: 1096–1101
  • Fleming I., Michaelis U. R., Bredenkotter D., Fisslthaler B., Dehghani F., Brandes R. P., et al. Endothelium‐derived hyperpolarizing factor synthase (Cytochrome P450 2C9) is a functionally significant source of reactive oxygen species in coronary arteries. Circ Res 2001; 88: 44–51
  • Sarkis A., Roman R. J. Role of cytochrome P450 metabolites of arachidonic acid in hypertension. Curr Drug Metab 2004; 5: 245–256
  • Moreno M. U., San Jose G., Orbe J., Paramo J. A., Beloqui O., Diez J., et al. Preliminary characterisation of the promoter of the human p22(phox) gene: Identification of a new polymorphism associated with hypertension. FEBS Lett 2003; 542: 27–31
  • San Jose G., Moreno M. U., Olivan S., Beloqui O., Fortuno A., Diez J., Zalba G. Functional effect of the p22phox ‐930A/G polymorphism on p22phox expression and NADPH oxidase activity in hypertension. Hypertension 2004; 44: 163–169
  • Zalba G., San Jose G., Moreno M. U., Fortuno A., Diez J. NADPH oxidase‐mediated oxidative stress: Genetic studies of the p22(phox) gene in hypertension. Antioxid Redox Signal 2005; 7: 1327–1336
  • Keaney J. F Jr. Oxidative stress and the vascular wall: NADPH oxidases take center stage. Circulation 2005; 112: 2585–2588
  • Dikalova A., Clempus R., Lassegue B., Cheng G., McCoy J., Dikalov S., et al. Nox1 overexpression potentiates angiotensin II‐induced hypertension and vascular smooth muscle hypertrophy in transgenic mice. Circulation 2005; 112: 2668–2676
  • Matsuno K., Yamada H., Iwata K., Jin D., Katsuyama M., Matsuki M., et al. Nox1 is involved in angiotensin II‐mediated hypertension: A study in Nox1‐deficient mice. Circulation 2005; 112: 2677–2685
  • Wang H. D., Xu S., Johns D. G., Du Y., Quinn M. T., Cayatte A. J., Cohen R. A. Role of NADPH oxidase in the vascular hypertrophic and oxidative stress response to angiotensin II in mice. Circ Res 2001; 88: 947–953
  • Lassegue B., Griendling K. K. Reactive oxygen species in hypertension; An update. Am J Hypertens 2004; 17: 852–860
  • Stocker R., Keaney J. F Jr. New insights on oxidative stress in the artery wall. J Thromb Haemost 2005; 3: 1825–1834
  • Ohara Y., Peterson T. E., Harrison D. G. Hypercholesterolemia increases endothelial superoxide anion production. J Clin Invest 1993; 91: 2546–2551
  • Meneshian A., Bulkley G. B. The physiology of endothelial xanthine oxidase: From urate catabolism to reperfusion injury to inflammatory signal transduction. Microcirculation 2002; 9: 161–175
  • McNally J. S., Davis M. E., Giddens D. P., Saha A., Hwang J., Dikalov S., et al. Role of xanthine oxidoreductase and NADPH oxidase in endothelial superoxide production in response to oscillatory shear stress. Am J Physiol Heart Circ Physiol 2003; 285: 2290–2297
  • Lenda D. M., Boegehold M. A. Effect of a high‐salt diet on oxidant enzyme activity in skeletal muscle microcirculation. Am J Physiol Heart Circ Physiol 2002; 282: 395–402
  • Ulker S., McMaster D., McKeown P. P., Bayraktutan U. Impaired activities of antioxidant enzymes elicit endothelial dysfunction in spontaneous hypertensive rats despite enhanced vascular nitric oxide generation. Cardiovasc Res 2003; 59: 488–500
  • Mervaala E. M., Cheng Z. J., Tikkanen I., Lapatto R., Nurminen K., Vapaatalo H., et al. Endothelial dysfunction and xanthine oxidoreductase activity in rats with human renin and angiotensinogen genes. Hypertension 2001; 37: 414–418
  • Li J. M., Shah A. M. Endothelial cell superoxide generation: Regulation and relevance for cardiovascular pathophysiology. Am J Physiol Regul Integr Comp Physiol 2004; 287: 1014–1030
  • Chaves F. J., Corella D., Blesa S., Mansego M. L., Marín P., Portoles O., et al. Xanthine oxidoreductase polymorphisms: Influence in blood pressure and oxidative stress levels. Pharmacogenet Genomics 2007; 17: 589–596
  • Fleming I., Busse R. Molecular mechanisms involved in the regulation of the endothelial nitric oxide synthase. Am J Physiol Regul Integr Comp Physiol 2003; 284: 1–12
  • Vasquez‐Vivar J., Kalyanaraman B., Martasek P., Hogg N., Masters B. S., Karoui H., et al. Superoxide generation by endothelial nitric oxide synthase: The influence of cofactors. Proc Natl Acad Sci U S A 1998; 95: 9220–9225
  • Forstermann U., Munzel T. Endothelial nitric oxide synthase in vascular disease: From marvel to menace. Circulation 2006; 113: 1708–1714
  • Munzel T., Daiber A., Ullrich V., Mulsch A. Vascular consequences of endothelial nitric oxide synthase uncoupling for the activity and expression of the soluble guanylyl cyclase and the cGMP‐dependent protein kinase. Arterioscler Thromb Vasc Biol 2005; 25: 1551–1557
  • Li H., Wallerath T., Munzel T., Forstermann U. Regulation of endothelial‐type NO synthase expression in pathophysiology and in response to drugs. Nitric Oxide 2002; 7: 149–164
  • Li H., Witte K., August M., Brausch I., Godtel‐Armbrust U., Habermeier A., et al. Reversal of endothelial nitric oxide synthase uncoupling and up‐regulation of endothelial nitric oxide synthase expression lowers blood pressure in hypertensive rats. J Am Coll Cardiol 2006; 47: 2536–2544
  • Tiefenbacher C. P. Tetrahydrobiopterin: A critical cofactor for eNOS and a strategy in the treatment of endothelial dysfunction?. Am J Physiol Heart Circ Physiol 2001; 280: 2484–2488
  • Vasquez‐Vivar J., Kalyanaraman B., Martasek P. The role of tetrahydrobiopterin in superoxide generation from eNOS: Enzymology and physiological implications. Free Radic Res 2003; 37: 121–127
  • Landmesser U., Dikalov S., Price S. R., McCann L., Fukai T., Holland S. M., et al. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J Clin Invest 2003; 111: 1201–1209
  • Laursen J. B., Somers M., Kurz S., McCann L., Warnholtz A., Freeman B. A., et al. Endothelial regulation of vasomotion in apoE‐deficient mice: Implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation 2001; 103: 1282–1288
  • Fleming I. Cytochrome P‐450 under pressure: More evidence for a link between 20‐hydroxyeicosatetraenoic acid and hypertension. Circulation 2005; 111: 5–7
  • Fleming I. Cytochrome p450 and vascular homeostasis. Circ Res 2001; 89: 753–762
  • Sarkis A., Lopez B., Roman R. J. Role of 20‐hydroxyeicosatetraenoic acid and epoxyeicosatrienoic acids in hypertension. Curr Opin Nephrol Hypertens 2004; 13: 205–214
  • Omata K., Abraham N. G., Schwartzman M. L. Renal cytochrome P‐450‐arachidonic acid metabolism: Localization and hormonal regulation in SHR. Am J Physiol 1992; 262: 591–599
  • Ward N. C., Rivera J., Hodgson J., Puddey I. B., Beilin L. J., Falck J. R., et al. Urinary 20‐hydroxyeicosatetraenoic acid is associated with endothelial dysfunction in humans. Circulation 2004; 110: 438–443
  • Gainer J. V., Bellamine A., Dawson E. P., Womble K. E., Grant S. W., Wang Y., et al. Functional variant of CYP4A11 20‐hydroxyeicosatetraenoic acid synthase is associated with essential hypertension. Circulation 2005; 111: 63–69
  • Li Y., Zhu H., Trush M. A. Detection of mitochondria‐derived reactive oxygen species production by the chemilumigenic probes lucigenin and luminol. Biochim Biophys Acta 1999; 1428: 1–12
  • Holmuhamedov E. L., Jovanovic S., Dzeja P. P., Jovanovic A., Terzic A. Mitochondrial ATP‐sensitive K+ channels modulate cardiac mitochondrial function. Am J Physiol 1998; 275: 1567–1576
  • Esposito L. A., Melov S., Panov A., Cottrell B. A., Wallace D. C. Mitochondrial disease in mouse results in increased oxidative stress. Proc Natl Acad Sci USA 1999; 96: 4820–4825
  • Wallace D. C. Mitochondrial genetics: A paradigm for aging and degenerative diseases?. Science 1992; 256: 628–632
  • Croteau D. L., Bohr V. A. Repair of oxidative damage to nuclear and mitochondrial DNA in mammalian cells. J Biol Chem 1997; 272: 25409–25412
  • Van Remmen H., Richardson A. Oxidative damage to mitochondria and aging. Exp Gerontol 2001; 36: 957–968
  • Wei Y. H., Lu C. Y., Lee H. C., Pang C. Y., Ma Y. S. Oxidative damage and mutation to mitochondrial DNA and age‐dependent decline of mitochondrial respiratory function. Ann N Y Acad Sci 1998; 854: 155–170
  • Puddu P., Puddu G. M., Galletti L., Cravero E., Muscari A. Mitochondrial dysfunction as an initiating event in atherogenesis: A plausible hypothesis. Cardiology 2005; 103: 137–141
  • Rachek L. I., Grishko V. I., Ledoux S. P., Wilson G. L. Role of nitric oxide‐induced mtDNA damage in mitochondrial dysfunction and apoptosis. Free Radic Biol Med 2006; 40: 754–762
  • Schwartz F., Duka A., Sun F., Cui J., Manolis A., Gavras H. Mitochondrial genome mutations in hypertensive individuals. Am J Hypertens 2004; 17: 629–635
  • DeStefano A. L., Gavras H., Heard‐Costa N., Bursztyn M., Manolis A., Farrer L. A., et al. Maternal component in the familial aggregation of hypertension. Clin Genet 2001; 60: 13–21
  • Bernal‐Mizrachi C., Gates A. C., Weng S., Imamura T., Knutsen R. H., DeSantis P., et al. Vascular respiratory uncoupling increases blood pressure and atherosclerosis. Nature 2005; 435: 502–506
  • Douette P., Gerkens P., Navet R., Leprince P., De Pauw E., Sluse F. E. Uncoupling protein 1 affects the yeast mitoproteome and oxygen free radical production. Free Radic Biol Med 2006; 40: 303–315
  • De Cavanagh E. M., Toblli J. E., Ferder L., Piotrkowski B., Stella I., Inserra F. Renal mitochondrial dysfunction in spontaneously hypertensive rats is attenuated by losartan but not by amlodipine. Am J Physiol Regul Integr Comp Physiol 2006; 290: 1616–1625
  • Brand M. D., Affourtit C., Esteves T. C., Green K., Lambert A. J., Miwa S., et al. Mitochondrial superoxide: Production, biological effects, and activation of uncoupling proteins. Free Radic Biol Med 2004; 37: 755–767
  • Ji Q., Ikegami H., Fujisawa T., Kawabata Y., Ono M., Nishino M., et al. A common polymorphism of uncoupling protein 2 gene is associated with hypertension. J Hypertens 2004; 22: 97–102
  • Atlante A., Seccia T. M., Pierro P., Vulpis V., Marra E., Pirrelli A., et al. ATP synthesis and export in heart left ventricle mitochondria from spontaneously hypertensive rat. Int J Mol Med 1998; 1: 709–716
  • Doroshchuk A. D., Postnov A. Iu., Afanas'eva G. V., Budnikov E. Iu., Postnov Iu. V. [Decreased ATP‐synthesis ability of brain mitochondria in spontaneously hypertensive rats.]. Kardiologiia 2004; 44: 64–65
  • Chen L., Tian X., Song L. Biochemical and biophysical characteristics of mitochondria in the hypertrophic hearts from hypertensive rats. Chin Med J (Engl) 1995; 108: 361–366
  • Seccia T. M., Atlante A., Vulpis V., Marra E., Passarella S., Pirrelli A. Abnormal transport of inorganic phosphate in left ventricular mitochondria from spontaneously hypertensive rats. Cardiologia 1999; 44: 719–725
  • Calderon‐Cortes E., Clemente‐Guerrero M., Sierra‐Campos E., Cortes‐Rojo C., Gaona‐Zamudio F. J., Villalobos‐Molina R., et al. Functional characterization of brain mitochondrial nitric oxide synthase during hypertension and aging. Amino Acids 2006; 30: 73–80
  • Kato K., Giulivi C. Critical overview of mitochondrial nitric‐oxide synthase. Front Biosci 2006; 1: 2725–2738
  • Zhang C., Yang J., Jacobs J. D., Jennings L. K. Interaction of myeloperoxidase with vascular NAD(P)H oxidase‐derived reactive oxygen species in vasculature: Implications for vascular diseases. Am J Physiol Heart Circ Physiol 2003; 285: H2563–H2572
  • Miller Y. I., Felikman Y., Shaklai N. Hemoglobin induced apolipoprotein B crosslinking in low‐density lipoprotein peroxidation. Arch Biochem Biophys 1996; 326: 252–260
  • Droge W. Free radicals in the physiological control of cell function. Physiol Rev 2002; 82: 47–95
  • Zalba G., San José G., Moreno M. U., Fortuño M. A., Fortuño A., Beaumont F. J., et al. Oxidative stress in arterial hypertension: Role of NAD(P)H oxidase. Hypertension 2001; 38: 1395–1399
  • Touyz R. M., Yao G., Schiffrin E. L. c‐Src induces phosphorylation and translocation of p47phox: Role in superoxide generation by angiotensin II in human vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2003; 23: 981–987
  • Ghiadoni L., Magagna A., Versari D., Kardasz I., Huang Y., Taddei S., et al. Different effect of antihypertensive drugs on conduit artery endothelial function. Hypertension 2003; 41: 1281–1286
  • Yoshida J., Yamamoto K., Mano T., Sakata Y., Nishikawa N., Nishio M., et al. AT1 receptor blocker added to ACE inhibitor provides benefits at advanced stage of hypertensive diastolic heart failure. Hypertension 2004; 43: 686–691
  • Puddu P., Puddu G. M., Cravero E., Muscari A. Different effects of antihypertensive drugs on endothelial dysfunction. Acta Cardiol 2004; 59: 555–564
  • Grote K., Flach I., Luchtefeld M., Akin E., Holland S. M., Drexler H., et al. Mechanical stretch enhances mRNA expression and proenzyme release of matrix metalloproteinase‐2 (MMP‐2) via NAD(P)H oxidase‐derived reactive oxygen species. Circ Res 2003; 92: e80–e86
  • Yasmin, McEniery C. M., Wallace S., Dakham Z., Pulsalkar P., Maki‐Petaja K., et al. Matrix metalloproteinase‐9 (MMP‐9), MMP‐2, and serum elastase activity are associated with systolic hypertension and arterial stiffness. Arterioscler Thromb Vasc Biol 2005; 25: 372
  • Boutouyrie P., Tropeano A. I., Asmar R., Gautier I., Benetos A., Lacolley P., et al. Aortic stiffness is an independent predictor of primary coronary events in hypertensive patients: A longitudinal study. Hypertension 2002; 39: 10–15
  • Kals J., Kampus P., Kals M., Zilmer K., Kullisaar T., Teesalu R., et al. Impact of oxidative stress on arterial elasticity in patients with atherosclerosis. Am J Hypertens 2006; 19: 902–908
  • Touyz R. M. Reactive oxygen species, vascular oxidative stress, and redox signaling in hypertension: What is the clinical significance?. Hypertension 2004; 44: 248–252
  • Touyz R. M., Tabet F., Schiffrin E. L. Redox‐dependent signalling by angiotensin II and vascular remodelling in hypertension. Clin Exp Pharmacol Physiol 2003; 30: 860–866
  • Chiarugi P., Cirri P. Redox regulation of protein tyrosine phosphatases during receptor tyrosine kinase signal transduction. Trends Biochem Sci 2003; 28: 509–514
  • Mullan B. A., Young I. S., Fee H., McCance D. R. Ascorbic acid reduces blood pressure and arterial stiffness in type 2 diabetes. Hypertension 2002; 40: 804–809
  • Chen J., He J., Hamm L., Batuman V., Whelton P. K. Serum antioxidant vitamins and blood pressure in the United States population. Hypertension 2002; 40: 810–816
  • Patterson C., Madamanchi N. R., Runge M. S. The oxidative paradox: Another piece in the puzzle. Circ Res 2000; 87: 1074–1076

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.