You are on page 1of 8

Heart Fail Rev (2011) 16:5–12

DOI 10.1007/s10741-010-9186-2

NADPH oxidases and cardiac remodelling


Adam Nabeebaccus • Min Zhang • Ajay M. Shah

Published online: 25 July 2010


 Springer Science+Business Media, LLC 2010

Abstract A heart under chronic stress undergoes cardiac Introduction


remodelling, a process that comprises structural and func-
tional changes including cardiomyocyte hypertrophy, Chronic heart failure is the culmination of a complex
interstitial fibrosis, contractile dysfunction, cell death and interplay between various myocardial structural and func-
ventricular dilatation. Reactive oxygen species (ROS)- tional changes that are elicited in response to specific
dependent modulation of intracellular signalling is impli- stressors such as chronic pressure overload (e.g. due to
cated in the development of cardiac remodelling. Among the hypertension) or myocardial infarction (MI) [1]. These
different ROS sources that are present in the heart, NADPH changes are usually characterised by an initial adaptive
oxidases (NOXs) are particularly important in redox sig- response of global or regional ventricular hypertrophy that
nalling. NOX isoforms are expressed in multiple cell types progresses to a series of maladaptive changes, including
including cardiomyocytes, fibroblasts, endothelial cells and interstitial fibrosis, contractile dysfunction, energetic defi-
inflammatory cells—with the two main isoforms expressed cit, cardiomyocyte death, vascular dysfunction and cham-
in the heart being NOX2 and NOX4. Recent studies indicate ber dilatation. These maladaptive changes are collectively
that NOX-dependent signalling is involved in the develop- termed adverse cardiac remodelling [2].
ment of cardiomyocyte hypertrophy, interstitial fibrosis and The process of remodelling is driven by complex
post-MI remodelling. NOXs may also be involved in the molecular mechanisms among which reactive oxygen
genesis of contractile dysfunction and myocyte apoptosis. species (ROS)-dependent pathways have attracted signifi-
Here, we review the main effects of NOXs in the patho- cant attention [3–5]. ROS include the free radicals super-
genesis of cardiac remodelling and the redox-sensitive sig- oxide anion (O•- •
2 ) and hydroxyl (OH ) as well as the highly
nalling pathways that underlie these effects. The elucidation reactive compound hydrogen peroxide (H2O2). ROS exert
of mechanisms involved in NOX-dependent regulation of biological effects either by causing non-specific oxidative
cardiac remodelling may lead to new therapeutic targets for damage to DNA, proteins, lipids and macromolecules or
heart failure. through specific modulation of cellular signalling pathways
(termed redox signalling). Cellular levels of ROS and
Keyword Cardiac remodelling  NADPH oxidase  therefore their effects are regulated by a variety of specific
Reactive oxygen species  Oxidative stress  and antioxidant systems (e.g. catalase, superoxide dismu-
Redox signalling tases, glutathione peroxidases, peroxiredoxins, thioredoxin
and various vitamins) [3]. Oxidative stress is said to exist
when there is an imbalance between ROS generation and
A. Nabeebaccus  M. Zhang  A. M. Shah (&)
Cardiovascular Division, King’s College London British Heart cellular antioxidant systems. An important concept with
Foundation Centre, London SE5 9PJ, UK respect to redox signalling is the idea that ROS production
e-mail: ajay.shah@kcl.ac.uk occurs in a spatially and temporally regulated fashion
within the cell, so that the specific modulation of signalling
A. Nabeebaccus
Cardiovascular Division, The James Black Centre, King’s pathways results in discrete changes in cellular phenotype
College London, 125 Coldharbour Lane, London SE5 9NU, UK [6].

123
6 Heart Fail Rev (2011) 16:5–12

ROS can be produced by a variety of enzyme systems cytosolic subunits (p47phox, p67phox, p40phox and Rac) [14].
that have been associated with heart failure, including Other NOX isoforms variably require the binding of dis-
xanthine oxidase (XO) [7], mitochondria [8], uncoupled tinct regulatory subunits for their activity [15, 16]. NOX1
nitric oxide synthases (NOSs) [9] and NADPH oxidases binds to p22phox, Rac1 and to homologues of p47phox and
(NOXs) [10]. The latter are a major source of ROS in the p67phox called NOXO1 and NOXA1. NOX3 can function
cardiovascular system and are unique among the enzymes just as a heterodimer with p22phox, but its activity is
mentioned in that they are especially important in redox enhanced by interaction with p47phox and p67phox, although
signalling [11]. Accumulating in vitro and in vivo evidence it does not seem to require Rac [17]. NOX4 also binds to
suggests that NADPH oxidases play important roles in the p22phox but differs from the other NOXs in that it does not
pathogenesis of cardiac remodelling. This includes studies require any other regulatory subunit for ROS production
that have shown an increase in myocardial NADPH oxidase and appears to be constitutionally active, with regulation
expression and activity in experimental and human heart thought to occur mainly at transcriptional level [18].
failure [10], and more recent work that has used genetically Interestingly, recent reports suggest that NOX4 may have a
modified mouse models to provide more direct molecular propensity for predominant H2O2 rather than O•- 2 produc-
evidence for a causative role of NADPH oxidase in the tion, in marked contrast to NOX2 [18–20]. NOX5 is only
different components of cardiac remodelling [11]. These found in humans and not in rodents, does not require
studies suggest that NADPH oxidases are important in p22phox for its activity, and has an extra calmodulin-like EF
modulating redox-sensitive signalling pathways involved in domain in its N-terminal which confers calcium sensitivity.
remodelling. Cardiovascular cells express a variety of NOX isoforms
[11]. NOX1 is found predominantly in vascular smooth
muscle cells, whereas NOX2 is expressed in endothelial
NADPH oxidase structure and isoforms cells, cardiomyocytes and fibroblasts. NOX4 is expressed
in all the above-mentioned cell types, while NOX5 is
NAPDH oxidase was first identified in neutrophils where it reported to be present in human endothelial cells and
generates ROS during phagocytosis and is involved in vascular smooth muscle cells. An important caveat is that
microbicidal activity [12]. Subsequently, a total of five much of the preceding is based on data from cultured cells
NOX enzymes and two related dual oxidase enzymes which may not necessarily reflect the physiological situa-
(DUOX) have been discovered [13]. The NOX enzymes tion in vivo. For example, although NOX4 is readily
are each based on a distinct catalytic subunit (NOX1-5)— detected in cultured cardiomyocytes, its in vivo expression
Fig. 1. The prototypic phagocyte NADPH oxidase consists in the healthy heart is very low. In general, NOXs (in
of a highly glycosylated membrane-spanning NOX2 sub- particular, NOX2) are activated by signalling pathways
unit (originally called gp91phox) that forms a complex with downstream of G-protein coupled receptors such as
a smaller molecular weight protein called p22phox. The angiotensin II and endothelin 1, by cytokines (e.g. TNFa)
heterodimer forms a flavocytochrome that catalyses the and/or mechanical forces. Following activation, ROS
transfer of electrons from NADPH to molecular oxygen, generated by NOXs may modulate diverse signalling
thereby generating O.- 2 . The catalytic activity of the het- pathways and redox-sensitive proteins (reviewed in
erodimer is stimulated by the binding of regulatory Anilkumar et al. [21]).

Fig. 1 Structure of NOX1-5


and requirement for regulatory
subunits. The structures are
based on predictions as no
crystal structure of any of the
NOX isoforms has yet been
solved. The haem moieties
facilitate the transfer of
electrons from NADPH and
FAD to molecular oxygen

123
Heart Fail Rev (2011) 16:5–12 7

Role in cardiac hypertrophy in adult mice inhibited angiotensin II-induced LVH as a


result of reduced NOX2 activation and ROS production.
In isolated neonatal rat cardiomyocytes exposed to angio- Key NOX-modulated signalling pathways that contrib-
tensin II, NADPH oxidase activation was shown to be ute to the development of LVH appear to be the mitogen-
involved in the ROS-dependent hypertrophic response [22]. activated protein kinases (MAPKs), in particular ERK1/2,
NAPDH oxidase-mediated ROS production was also impli- and the phosphatidylinositol 3-kinase (PI3K)-Akt pathway
cated in a1-adrenoreceptor-stimulated hypertrophy of adult (Fig. 2) [22–27]. For example, a1-adrenoreceptor-stimu-
rat ventricular myocytes [23]. These studies suggested that lated cardiomyocyte hypertrophy involves NADPH oxi-
NOX2 was involved in these effects. In a guinea-pig model of dase-dependent activation of the Ras-MEK1/2-ERK1/2
pressure overload left ventricular hypertrophy (LVH), a sig- pathway [23]. Interestingly, Ras may be a direct target of
nificant and sustained increase in NADPH oxidase activation ROS generated by NOXs as it was shown that a specific
was found that correlated with increased expression of thioredoxin-1-sensitive post-translational oxidative modi-
NOX2, p22phox and p67phox, both in cardiomyocytes and in fication of thiols was involved in its activation [28].
coronary microvascular endothelial cells [24]. Angiotensin II-induced cardiomyocyte hypertrophy was
Studies using NOX2 knockout mice demonstrated more also reported to involve NOX2-dependent Akt activation
compelling evidence for a role of NOX2 in mediating [26]. The Rho GTPase Rac is critical for activation of
hypertrophy. Compared to wild-type controls, NOX2- NOX2, and Rac1-induced hypertrophy of neonatal
deficient mice subjected to short-term subpressor infusion cardiomyocytes involved the activation of ERK1/2 and
of angiotensin II developed less LVH and lower increases JNK signalling pathways [29], although whether this
in molecular markers of hypertrophy, such as atrial natri- involved NOX2 was not directly addressed in this study.
uretic factor and b-myosin heavy chain mRNA [25]. Fur- Apoptosis signal-regulating kinase 1 (ASK-1) is also
thermore, angiotensin II-induced increases in NADPH implicated in NOX-dependent signalling in cardiac
oxidase activity were abolished in NOX2-deficient ani- hypertrophy [27] and in fact mice deficient in ASK-1
mals. Whereas this study was undertaken in mice globally showed impaired angiotensin II-induced JNK and
deficient in NOX2, supporting evidence for a role of car- p38MAPK activation as well as reduced hypertrophy [30].
diomyocyte NOX2 in angiotensin II-induced hypertrophy Activation of the transcription factors AP-1 and NFjB also
was provided by a study in isolated neonatal rat cardio- appears to be involved in NOX-dependent hypertrophy. In
myocytes in which siRNA directed against NOX2 abol- neonatal rat ventricular myocytes, angiotensin II activated
ishing angiotensin II induced superoxide generation and AP-1 through NOX [2, 31, 32], although it remains unclear
cardiomyocyte hypertrophy [26]. Additional in vivo data in whether this is a direct effect or occurs downstream of a
support of the importance of cardiomyocyte NOX2 was NOX2-activated signalling pathway. Cardiomyocyte
provided by Satoh et al. [27] in a study that used inducible hypertrophy induced by GPCR agonists or by TNF-a,
cardiomyocyte-specific Rac1-deficient mice. This group which activates NOX2, is reported to be mediated via
demonstrated that cardiomyocyte-specific deletion of Rac1 NFjB activation [33].

Fig. 2 Schematic showing


major signalling targets of
NADPH oxidase-derived ROS
in cardiac remodelling. RyR
Ryanodine receptor, SERCA
sarcoplasmic reticulum Ca2?
ATPase, MMP matrix
metalloproteinase, CTGF
connective tissue growth factor,
TGFb transforming growth
factor b

123
8 Heart Fail Rev (2011) 16:5–12

In contrast to angiotensin II-induced hypertrophy, NOX2 aldosterone/salt treatment showed a marked reduction in
was found not to be essential for the development of pressure NOX2-associated ROS production with a significant
overload LVH which was unaffected in NOX2 knockout reduction in interstitial fibrosis and perivascular fibrosis
mice [34, 35]. Another study also reported that NOX2 did not even though cardiac hypertrophy was similar to wild-type
inhibit LVH in a model of chronic activation of the renin– mice [39]. In Dahl salt-sensitive rats fed on a high salt diet,
angiotensin system secondary to transgenic overexpression LV fibrosis was associated with an increased expression of
of human renin [36]—which could be related to the pressure p22phox and NOX2 and increased NADPH oxidase activity
overload present in this transgenic mouse model. Interest- [40]. When Dahl sat-sensitive rats fed on a high salt diet
ingly, NADPH oxidase activity was still elevated in were treated with an eNOS enhancer, there was a decrease
NOX2-/- mice after aortic banding, which was attributed to in cardiac subunit expression of NOX2 and p22phox asso-
increased NOX4 expression [22], suggesting that NOX4 ciated with an attenuation in diastolic dysfunction, cardiac
may also play some role in pressure overload LVH. How- hypertrophy and fibrosis [41]. Interstitial fibrosis is also
ever, preliminary results indicate that the two isoforms may significantly attenuated in NOX2-deficient mice subjected
have distinct, non-redundant effects and that one isoform to aortic banding even though they have similar levels of
does not necessarily compensate for the other. The results of hypertrophy as their wild-type counterparts [42]. Taken
detailed studies using cell- and isoform-specific manipula- together, these studies indicate an important role of NOX2
tion of NOX2 and NOX4 will help to further define the in the development of in vivo interstitial cardiac fibrosis
respective roles of these two isoforms. occurring in response to diverse stimuli.
Johar et al. [37] found that interstitial cardiac fibrosis
induced by angiotensin II involved Nox2-dependent upreg-
Interstitial cardiac fibrosis ulation of profibrotic cytokines such as connective tissue
growth factor (CTGF) and profibrotic genes such as pro-
Interstitial cardiac fibrosis is a hallmark of adverse cardiac collagen-1, procollagen-3 and fibronectin (Fig. 2). They also
remodelling and contributes to increased myocardial stiff- found that there was increased Nox2-dependent activation of
ness, impaired diastolic and systolic function, and an NF-jB and MMP2. MMP2 is implicated in cardiac remod-
increased propensity for arrhythmias [1]. A significant body elling and has importance in modulating cardiomyocyte
of data implicates NOX2 NADPH oxidase in the develop- function including causing contractile dysfunction and
ment of interstitial fibrosis in vivo. It was shown that NOX2 apoptosis [43]. It has also been shown to be activated by
knockout mice had markedly reduced fibrosis compared to NOX via JNK/p38/ERK1/2 signalling in cardiac myocytes
wild-type animals in response to either subpressor or pressor exposed to doxorubicin (a cardiotoxic anticancer drug that
infusion of angiotensin II [25, 37]. Interestingly, in the can cause heart failure) [44]. The NOX2-expressing cell
pressor model, interstitial fibrosis was inhibited in NOX2- types that are involved in the development of fibrosis (i.e.
deficient mice even though the extent of LVH was similar to fibroblasts, cardiomyocytes, inflammatory cells and/or
wild-type controls [37], indicating a dissociation between endothelial cells) remain to be defined.
the pathways contributing to fibrosis and cardiomyocyte In contrast to above-mentioned studies, some in vitro
hypertrophy. In the study by Satoh et al. [27] in cardiomy- studies suggest an important role for NOX4 in the devel-
ocyte-specific Rac1 knockout mice, angiotensin II-induced opment of cardiac fibrosis. Cucoranu et al. [45] in a study
fibrosis was also inhibited, suggesting that in this case involving cultured human cardiac fibroblasts found that
cardiomyocytes NOX2 activation drove the fibrosis. NOX4 and 5 mRNAs were abundantly expressed, whereas
NOX2 was also implicated in aldosterone-induced NOX1 and 2 were barely detectable. These authors repor-
interstitial cardiac fibrosis [37]. Furthermore, Johar et al. ted that TGF-b1 upregulated NOX4 expression and pro-
[37] found that angiotensin II-induced in vivo NADPH moted the conversion of fibroblasts to myofibroblasts,
oxidase activation and interstitial fibrosis in wild-type mice which are the main producers of collagen in the extracel-
were inhibited by treatment with the mineralocorticoid lular matrix. This study implied that NOX4 may be
receptor antagonist, spironolactone, despite a lack of responsible for driving cardiac fibrosis in response to TGF-
change in blood pressure or cardiac hypertrophy. This b1 stimulation, the latter being well established as a driver
finding suggests that mineralocorticoid receptor activation of cardiac fibrosis [46]. However, TGF-b may also have
is important downstream of angiotensin II in this model. beneficial actions as evidenced by data that TGF-b inhi-
Studies in a rat model of aldosterone-induced fibrosis and bition is detrimental in mice after MI [47]. It should be
inflammation have also implicated NOX2 activation and noted that NOX2 is expressed in mouse cardiac fibroblasts
increased myocardial oxidative stress in the pathogenesis and that TGF-b1 enhanced expression of p22phox, p47phox
of the fibrosis [38]. In a more recent study, apoptosis sig- and NOX2 as well as collagen formation by these cells,
nal-regulating kinase 1 (ASK1)-deficient mice subjected to which could be reduced by NOX2 siRNA [48]. To

123
Heart Fail Rev (2011) 16:5–12 9

determine whether both NOX2 and NOX4 are involved in neonatal rat cardiomyocytes exposed to angiotensin II,
the development of interstitial fibrosis or whether there are increased NADPH oxidase expression and activity were
differences between mouse and human fibroblasts or reported to contribute to apoptosis [59]. The specific NOX
whether in vitro studies fail to reflect the complex in vivo isoform involved was not identified, but the involvement
setting in which fibrosis develops requires further studies. of p47phox suggests that it may be NOX2. A similar
pro-apoptotic effect of aldosterone in neonatal rat cardio-
myocytes was also suggested to involve NADPH oxidase-
Contractile dysfunction derived ROS production [60]. NADPH oxidase-derived
ROS were also implicated in cardiomyocyte apoptosis
The development of contractile dysfunction is an important induced by the chemotherapeutic agent doxorubicin [61].
feature of cardiac decompensation during chronic exposure Likewise, in cultured adult rat cardiomyocytes exposed to
to stress and a hallmark of adverse cardiac remodelling [1]. high glucose as a model of diabetic cardiomyopathy,
Several studies suggest that NADPH oxidase activation may upregulation of Rac1 and NOX2 oxidase activity contrib-
contribute to the development of contractile dysfunction in uted to apoptosis [62, 63].
cardiac hypertrophy and heart failure. In a guinea-pig model The potential mechanisms involved in NOX-dependent
of LVH, increased myocardial NADPH oxidase activity was pro-apoptotic effects remain to be fully defined [21]. In
shown to contribute to impaired coronary endothelial func- neonatal rat cardiac myocytes exposed to aldosterone, it
tion and thereby to impaired ventricular relaxation [49]. was suggested that NOX-dependent ROS production pro-
Along similar lines, Cheng et al. [50] reported that NADPH moted apoptosis via activation of ASK-1 [60]. In angio-
oxidase activation contributed to increased diastolic stiffness tensin II-induced cardiomyocyte apoptosis, the activation
in the Dahl salt-sensitive rat model of hypertensive heart of p38MAPK, increase in Bcl-2, decrease in Bax and
failure, although the mechanism was proposed to involve caspase-3 activity were proposed to be NOX dependent,
changes in the extracellular matrix. In NOX2-deficient mice based on the use of apocynin [59].
subjected to aortic banding, Grieve et al. [42] found evidence
of significantly better contractile function than wild-type
animals both in whole hearts and at the level of isolated Post-MI remodelling
cardiomyocytes. The contractile dysfunction in wild-type
animals could be acutely improved by antioxidant treatment, Cardiac remodelling occurring after myocardial infarction
suggesting that there were direct effects of increased ROS on involves all the features described previously, namely
contractile function in this setting. NADPH oxidase activa- cardiomyocyte hypertrophy, fibrosis and apoptosis. In
tion has also been implicated in the development of con- addition, a major feature is extracellular matrix remodel-
tractile dysfunction in myocardial depression associated ling and dilatation of the heart. Numerous studies support
with gram-negative sepsis [51, 52], obesity [53] and strep- an important role for increased ROS in this process [64].
tozotocin-induced diabetes [54]. NOX2 and associated oxidase subunit expression have
The mechanisms underlying NOX-dependent contractile been shown to be increased at sites of infarcted myocar-
dysfunction remain to be fully defined. Abnormalities of dium in a rat model [65] as well as in humans who had died
cardiomyocyte excitation–contraction coupling, mitochon- from acute myocardial infarction [66]. In the latter study, at
drial dysfunction, myocyte apoptosis and/or alterations in least part of the expression was in cardiomyocytes. Direct
the extracellular matrix could all contribute depending evidence to support a role for NOX2 in post-MI remodel-
upon the context. With respect to excitation–contraction ling comes from studies in p47phox-deficient and NOX2-
coupling, it is known that ROS can modulate the function deficient mice. Doerries et al. [67] found that p47phox-
of sarcolemmal ion channels, the sarcoplasmic reticulum knockout mice had better preserved contractile function,
(SR) calcium release channel (the ryanodine receptor), the reduced LV remodelling and higher survival than wild-type
SR calcium pump (SERCA 2a) and contractile proteins controls after MI, despite similar infarct size. This was
themselves [55, 56]. Recently, it has also been reported that associated with a reduction in cardiomyocyte hypertrophy,
ROS may modulate CAMkinase II, possibly via NOX [57]. apoptosis and interstitial fibrosis. Similarly, Looi et al. [68]
found that NOX2-deficient mice were also protected
against contractile dysfunction and LV remodelling post-
Cardiomyocyte apoptosis MI when compared to wild-type mice, again despite a
similar infarct size. NOX2-deficient mice also had less
Apoptosis of cardiomyocytes is considered to be an cardiomyocyte hypertrophy, apoptosis and interstitial
important contributor to the development of fibrosis and fibrosis. Frantz et al. [69] also studied the response to MI in
contractile dysfunction on the failing heart [58]. In Nox2-deficient mice but reported no benefit of Nox2

123
10 Heart Fail Rev (2011) 16:5–12

deletion. However, it should be noted that these authors did References


not use littermate controls nor did they include a sham-
operated group for either genotype. Dandapat et al. [70] 1. Cohn JN, Ferrari R, Sharpe N (2000) Cardiac remodeling—
concepts and clinical implications: a consensus paper from an
found that overexpression of TGF-b1 in mouse cardio- international forum on cardiac remodeling. Behalf of an inter-
myocytes reduced NADPH oxidase activation and ROS national forum on cardiac remodeling. J Am Coll Cardiol
production after hypoxia–reoxygenation, with a concomi- 35(3):569–582
tant reduction in apoptosis. These authors also found that in 2. Opie LH, Commerford PJ, Gersh BJ, Pfeffer MA (2006) Con-
troversies in ventricular remodelling. Lancet 367(9507):356–367
vivo rodent hearts transfected with TGF-b1 and then 3. Giordano FJ (2005) Oxygen, oxidative stress, hypoxia, and heart
exposed to ischaemia–reperfusion injury developed failure. J Clin Invest 115(3):500–508
reduced NADPH oxidase activation and smaller infarct 4. Takimoto E, Kass DA (2007) Role of oxidative stress in cardiac
size compared to non-transfected rodents. TGF-b1 is gen- hypertrophy and remodeling. Hypertension 49(2):241–248
5. Hori M, Nishida K (2009) Oxidative stress and left ventricular
erally regarded to be pro-fibrotic; however, in this model remodelling after myocardial infarction. Cardiovasc Res 81(3):
the effect was cardioprotective both in vitro and in vivo and 457–464
involved NADPH oxidase although the NOX isoform and 6. Droge W (2002) Free radicals in the physiological control of cell
underlying mechanisms remain unclear. NADPH oxidase function. Physiol Rev 82(1):47–95
7. Cappola TP, Kass DA, Nelson GS, Berger RD, Rosas GO,
activation has also been reported to contribute to the Kobeissi ZA, Marban E, Hare JM (2001) Allopurinol improves
exacerbation of post-MI heart failure in mice with type 2 myocardial efficiency in patients with idiopathic dilated cardio-
diabetes compared to non-diabetic mice [71]. myopathy. Circulation 104(20):2407–2411
8. Ide T, Tsutsui H, Hayashidani S, Kang D, Suematsu N, Nakamura
K, Utsumi H, Hamasaki N, Takeshita A (2001) Mitochondrial
DNA damage and dysfunction associated with oxidative stress in
Conclusions and clinical implications failing hearts after myocardial infarction. Circ Res 88(5):529–535
9. Takimoto E, Champion HC, Li M, Ren S, Rodriguez ER, Tavazzi
B, Lazzarino G, Paolocci N, Gabrielson KL, Wang Y, Kass DA
The NADPH oxidases have emerged as important players (2005) Oxidant stress from nitric oxide synthase-3 uncoupling
in modulating redox-sensitive signalling pathways stimulates cardiac pathologic remodeling from chronic pressure
involved in the processes of cardiac remodelling. They load. J Clin Invest 115(5):1221–1231
influence several aspects of this complex process, including 10. Heymes C, Bendall JK, Ratajczak P, Cave AC, Samuel JL,
Hasenfuss G, Shah AM (2003) Increased myocardial NADPH
cardiomyocyte hypertrophy, contractile dysfunction, oxidase activity in human heart failure. J Am Coll Cardiol
apoptosis and interstitial fibrosis. In vivo studies with NOX 41(12):2164–2171
enzyme-deficient mice have begun to demonstrate the 11. Cave AC, Brewer AC, Narayanapanicker A, Ray R, Grieve DJ,
functional relevance of these alterations in signalling. In Walker S, Shah AM (2006) NADPH oxidases in cardiovascular
health and disease. Antioxid Redox Signal 8(5–6):691–728
this regard, it would be attractive to selectively target 12. Babior BM, Lambeth JD, Nauseef W (2002) The neutrophil
detrimental aspects of the process of adverse cardiac NADPH oxidase. Arch Biochem Biophys 397(2):342–344
remodelling, such as fibrosis or cardiomyocyte apoptosis. 13. Lambeth JD (2004) NOX enzymes and the biology of reactive
In fact, some of the beneficial effects of existing therapies oxygen. Nat Rev Immunol 4(3):181–189
14. Sumimoto H, Miyano K, Takeya R (2005) Molecular composi-
for heart failure, such as ACE inhibitors and angiotensin tion and regulation of the nox family NAD(P)H oxidases. Bio-
receptor blockers, could in part relate to inhibition of chem Biophys Res Commun 338(1):677–686
NOX2-dependent signalling since angiotensin II is a key 15. Lambeth JD, Kawahara T, Diebold B (2007) Regulation of nox
activator of the enzyme. Statins (3-hydroxy-3-methylglu- and duox enzymatic activity and expression. Free Radic Biol Med
43(3):319–331
taryl coenzyme A reductase inhibitors) also inhibit NOX2 16. Brown DI, Griendling KK (2009) Nox proteins in signal trans-
activation although evidence that they are beneficial in duction. Free Radic Biol Med 47(9):1239–1253
clinical heart failure is still lacking [72]. The complexity of 17. Hordijk PL (2006) Regulation of NADPH oxidases: the role of
regulation of NOX enzymes may paradoxically provide rac proteins. Circ Res 98(4):453–462
18. Martyn KD, Frederick LM, von Loehneysen K, Dinauer MC,
additional ways of therapeutically manipulating enzyme Knaus UG (2006) Functional analysis of Nox4 reveals unique
activity. A better understanding of the biochemical func- characteristics compared to other NADPH oxidases. Cell Signal
tion of these fascinating enzymes as well as their roles in 18(1):69–82
cardiac remodelling may lead to new therapeutic targets 19. Serrander L, Cartier L, Bedard K, Banfi B, Lardy B, Plastre O,
Sienkiewicz A, Forro L, Schlegel W, Krause KH (2007) NOX4
based on modulation of NOX activity. activity is determined by mRNA levels and reveals a unique
pattern of ROS generation. Biochem J 406(1):105–114
Acknowledgments The authors’ work is supported by the British 20. Dikalov SI, Dikalova AE, Bikineyeva AT, Schmidt HH, Harrison
Heart Foundation (RG/08/011/25922, CH/99001; RE/08/003), a DG, Griendling KK (2008) Distinct roles of Nox1 and Nox4 in
Leducq Foundation Transtlantic Network of Excellence Award, and basal and angiotensin II-stimulated superoxide and hydrogen
EU FP6 grant LSHM-CT-2005-018833 (EUGeneHeart). peroxide production. Free Radic Biol Med 45(9):1340–1351

123
Heart Fail Rev (2011) 16:5–12 11

21. Anilkumar N, Sirker A, Shah AM (2009) Redox sensitive sig- 38. Sun Y, Zhang J, Lu L, Chen SS, Quinn MT, Weber KT (2002)
naling pathways in cardiac remodeling, hypertrophy and failure. Aldosterone-induced inflammation in the rat heart: role of oxi-
Front Biosci 14:3168–3187 dative stress. Am J Pathol 161(5):1773–1781
22. Nakagami H, Takemoto M, Liao JK (2003) NADPH oxidase- 39. Nakamura T, Kataoka K, Fukuda M, Nako H, Tokutomi Y, Dong
derived superoxide anion mediates angiotensin II-induced cardiac YF, Ichijo H, Ogawa H, Kim-Mitsuyama S (2009) Critical role of
hypertrophy. J Mol Cell Cardiol 35(7):851–859 apoptosis signal-regulating kinase 1 in aldosterone/salt-induced
23. Xiao L, Pimentel DR, Wang J, Singh K, Colucci WS, Sawyer DB cardiac inflammation and fibrosis. Hypertension 54(3):544–551
(2002) Role of reactive oxygen species and NAD(P)H oxidase in 40. Yamada T, Nagata K, Cheng XW, Obata K, Saka M, Miyachi M,
alpha(1)-adrenoceptor signaling in adult rat cardiac myocytes. Naruse K, Nishizawa T, Noda A, Izawa H, Kuzuya M, Okumura
Am J Physiol Cell Physiol 282(4):C926–C934 K, Murohara T, Yokota M (2009) Long-term administration of
24. Li JM, Gall NP, Grieve DJ, Chen M, Shah AM (2002) Activation nifedipine attenuates cardiac remodeling and diastolic heart
of NADPH oxidase during progression of cardiac hypertrophy to failure in hypertensive rats. Eur J Pharmacol 615(1–3):163–170
failure. Hypertension 40(4):477–484 41. Westermann D, Riad A, Richter U, Jager S, Savvatis K, Schuc-
25. Bendall JK, Cave AC, Heymes C, Gall N, Shah AM (2002) hardt M, Bergmann N, Tolle M, Nagorsen D, Gotthardt M,
Pivotal role of a gp91(phox)-containing NADPH oxidase in Schultheiss HP, Tschope C (2009) Enhancement of the endo-
angiotensin II-induced cardiac hypertrophy in mice. Circulation thelial NO synthase attenuates experimental diastolic heart fail-
105(3):293–296 ure. Basic Res Cardiol 104(5):499–509
26. Hingtgen SD, Tian X, Yang J, Dunlay SM, Peek AS, Wu Y, Sharma 42. Grieve DJ, Byrne JA, Siva A, Layland J, Johar S, Cave AC, Shah
RV, Engelhardt JF, Davisson RL (2006) Nox2-containing NADPH AM (2006) Involvement of the nicotinamide adenosine dinucle-
oxidase and akt activation play a key role in angiotensin II-induced otide phosphate oxidase isoform Nox2 in cardiac contractile
cardiomyocyte hypertrophy. Physiol Genomics 26(3):180–191 dysfunction occurring in response to pressure overload. J Am
27. Satoh M, Ogita H, Takeshita K, Mukai Y, Kwiatkowski DJ, Liao Coll Cardiol 47(4):817–826
JK (2006) Requirement of Rac1 in the development of cardiac 43. Kandasamy AD, Chow AK, Ali MA, Schulz R (2010) Matrix
hypertrophy. Proc Natl Acad Sci USA 103(19):7432–7437 metalloproteinase-2 and myocardial oxidative stress injury:
28. Kuster GM, Pimentel DR, Adachi T, Ido Y, Brenner DA, Cohen beyond the matrix. Cardiovasc Res 85(3):413–423
RA, Liao R, Siwik DA, Colucci WS (2005) Alpha-adrenergic 44. Spallarossa P, Altieri P, Garibaldi S, Ghigliotti G, Barisione C,
receptor-stimulated hypertrophy in adult rat ventricular myocytes Manca V, Fabbi P, Ballestrero A, Brunelli C, Barsotti A (2006)
is mediated via thioredoxin-1-sensitive oxidative modification of Matrix metalloproteinase-2 and -9 are induced differently by
thiols on ras. Circulation 111(9):1192–1198 doxorubicin in H9c2 cells: the role of MAP kinases and
29. Clerk A, Pham FH, Fuller SJ, Sahai E, Aktories K, Marais R, NAD(P)H oxidase. Cardiovasc Res 69(3):736–745
Marshall C, Sugden PH (2001) Regulation of mitogen-activated 45. Cucoranu I, Clempus R, Dikalova A, Phelan PJ, Ariyan S,
protein kinases in cardiac myocytes through the small G protein Dikalov S, Sorescu D (2005) NAD(P)H oxidase 4 mediates
Rac1. Mol Cell Biol 21(4):1173–1184 transforming growth factor-beta1-induced differentiation of car-
30. Izumiya Y, Kim S, Izumi Y, Yoshida K, Yoshiyama M, Ma- diac fibroblasts into myofibroblasts. Circ Res 97(9):900–907
tsuzawa A, Ichijo H, Iwao H (2003) Apoptosis signal-regulating 46. Sakata Y, Chancey AL, Divakaran VG, Sekiguchi K, Sivasubr-
kinase 1 plays a pivotal role in angiotensin II-induced cardiac amanian N, Mann DL (2008) Transforming growth factor-beta
hypertrophy and remodeling. Circ Res 93(9):874–883 receptor antagonism attenuates myocardial fibrosis in mice with
31. Wu S, Gao J, Ohlemeyer C, Roos D, Niessen H, Kottgen E, cardiac-restricted overexpression of tumor necrosis factor. Basic
Gessner R (2005) Activation of AP-1 through reactive oxygen Res Cardiol 103(1):60–68
species by angiotensin II in rat cardiomyocytes. Free Radic Biol 47. Frantz S, Hu K, Adamek A, Wolf J, Sallam A, Maier SK, Lonning
Med 39(12):1601–1610 S, Ling H, Ertl G, Bauersachs J (2008) Transforming growth factor
32. Tu VC, Sun H, Bowden GT, Chen QM (2007) Involvement of beta inhibition increases mortality and left ventricular dilatation
oxidants and AP-1 in angiotensin II-activated NFAT3 transcrip- after myocardial infarction. Basic Res Cardiol 103(5):485–492
tion factor. Am J Physiol Cell Physiol 292(4):C1248–C1255 48. Hu C, Dandapat A, Sun L, Khan JA, Liu Y, Hermonat PL, Mehta
33. Higuchi Y, Otsu K, Nishida K, Hirotani S, Nakayama H, Yam- JL (2008) Regulation of TGFbeta1-mediated collagen formation
aguchi O, Matsumura Y, Ueno H, Tada M, Hori M (2002) by LOX-1: Studies based on forced overexpression of TGFbeta1
Involvement of reactive oxygen species-mediated NF-kappa B in wild-type and lox-1 knock-out mouse cardiac fibroblasts.
activation in TNF-alpha-induced cardiomyocyte hypertrophy. J Biol Chem 283(16):10226–10231
J Mol Cell Cardiol 34(2):233–240 49. MacCarthy PA, Grieve DJ, Li JM, Dunster C, Kelly FJ, Shah AM
34. Byrne JA, Grieve DJ, Bendall JK, Li JM, Gove C, Lambeth JD, (2001) Impaired endothelial regulation of ventricular relaxation
Cave AC, Shah AM (2003) Contrasting roles of NADPH oxidase in cardiac hypertrophy: role of reactive oxygen species and
isoforms in pressure-overload versus angiotensin II-induced car- NADPH oxidase. Circulation 104(24):2967–2974
diac hypertrophy. Circ Res 93(9):802–805 50. Cheng XW, Okumura K, Kuzuya M, Jin Z, Nagata K, Obata K,
35. Maytin M, Siwik DA, Ito M, Xiao L, Sawyer DB, Liao R, Col- Inoue A, Hirashiki A, Takeshita K, Unno K, Harada K, Shi GP,
ucci WS (2004) Pressure overload-induced myocardial hyper- Yokota M, Murohara T (2009) Mechanism of diastolic stiffening
trophy in mice does not require gp91phox. Circulation 109(9): of the failing myocardium and its prevention by angiotensin
1168–1171 receptor and calcium channel blockers. J Cardiovasc Pharmacol
36. Touyz RM, Mercure C, He Y, Javeshghani D, Yao G, Callera GE, 54(1):47–56
Yogi A, Lochard N, Reudelhuber TL (2005) Angiotensin II- 51. Peng T, Lu X, Feng Q (2005) Pivotal role of gp91phox-con-
dependent chronic hypertension and cardiac hypertrophy are taining NADH oxidase in lipopolysaccharide-induced tumor
unaffected by gp91phox-containing NADPH oxidase. Hyperten- necrosis factor-alpha expression and myocardial depression.
sion 45(4):530–537 Circulation 111(13):1637–1644
37. Johar S, Cave AC, Narayanapanicker A, Grieve DJ, Shah AM 52. Zhu H, Shan L, Peng T (2009) Rac1 mediates sex difference in
(2006) Aldosterone mediates angiotensin II-induced interstitial cardiac tumor necrosis factor-alpha expression via NADPH oxi-
cardiac fibrosis via a Nox2-containing NADPH oxidase. FASEB dase-ERK1/2/p38 MAPK pathway in endotoxemia. J Mol Cell
J 20(9):1546–1548 Cardiol 47(2):264–274

123
12 Heart Fail Rev (2011) 16:5–12

53. Li SY, Yang X, Ceylan-Isik AF, Du M, Sreejayan N, Ren J 63. Thandavarayan RA, Watanabe K, Ma M, Gurusamy N,
(2006) Cardiac contractile dysfunction in Lep/Lep obesity is Veeraveedu PT, Konishi T, Zhang S, Muslin AJ, Kodama M,
accompanied by NADPH oxidase activation, oxidative modifi- Aizawa Y (2009) Dominant-negative p38alpha mitogen-activated
cation of sarco(endo)plasmic reticulum Ca2?-ATPase and myosin protein kinase prevents cardiac apoptosis and remodeling after
heavy chain isozyme switch. Diabetologia 49(6):1434–1446 streptozotocin-induced diabetes mellitus. Am J Physiol Heart
54. Guo Z, Xia Z, Jiang J, McNeill JH (2007) Downregulation of Circ Physiol 297(3):H911–H919
NADPH oxidase, antioxidant enzymes, and inflammatory 64. Sun Y (2007) Oxidative stress and cardiac repair/remodeling
markers in the heart of streptozotocin-induced diabetic rats by following infarction. Am J Med Sci 334(3):197–205
N-acetyl-L-cysteine. Am J Physiol Heart Circ Physiol 292(4): 65. Fukui T, Yoshiyama M, Hanatani A, Omura T, Yoshikawa J, Abe
H1728–H1736 Y (2001) Expression of p22-phox and gp91-phox, essential
55. Zima AV, Blatter LA (2006) Redox regulation of cardiac calcium components of NADPH oxidase, increases after myocardial
channels and transporters. Cardiovasc Res 71(2):310–321 infarction. Biochem Biophys Res Commun 281(5):1200–1206
56. Yano M, Okuda S, Oda T, Tokuhisa T, Tateishi H, Mochizuki M, 66. Krijnen PA, Meischl C, Hack CE, Meijer CJ, Visser CA, Roos D,
Noma T, Doi M, Kobayashi S, Yamamoto T, Ikeda Y, Ohkusa T, Niessen HW (2003) Increased Nox2 expression in human
Ikemoto N, Matsuzaki M (2005) Correction of defective inter- cardiomyocytes after acute myocardial infarction. J Clin Pathol
domain interaction within ryanodine receptor by antioxidant is a 56(3):194–199
new therapeutic strategy against heart failure. Circulation 112(23): 67. Doerries C, Grote K, Hilfiker-Kleiner D, Luchtefeld M, Schaefer
3633–3643 A, Holland SM, Sorrentino S, Manes C, Schieffer B, Drexler H,
57. Palomeque J, Rueda OV, Sapia L, Valverde CA, Salas M, Petroff Landmesser U (2007) Critical role of the NAD(P)H oxidase
MV, Mattiazzi A (2009) Angiotensin II-induced oxidative stress subunit p47phox for left ventricular remodeling/dysfunction and
resets the Ca2? dependence of Ca2?-calmodulin protein kinase II survival after myocardial infarction. Circ Res 100(6):894–903
and promotes a death pathway conserved across different species. 68. Looi YH, Grieve DJ, Siva A, Walker SJ, Anilkumar N, Cave AC,
Circ Res 105(12):1204–1212 Marber M, Monaghan MJ, Shah AM (2008) Involvement of Nox2
58. Dorn GW 2nd (2009) Apoptotic and non-apoptotic programmed NADPH oxidase in adverse cardiac remodeling after myocardial
cardiomyocyte death in ventricular remodelling. Cardiovasc Res infarction. Hypertension 51(2):319–325
81(3):465–473 69. Frantz S, Brandes RP, Hu K, Rammelt K, Wolf J, Scheuermann
59. Qin F, Patel R, Yan C, Liu W (2006) NADPH oxidase is involved H, Ertl G, Bauersachs J (2006) Left ventricular remodeling after
in angiotensin II-induced apoptosis in H9C2 cardiac muscle cells: myocardial infarction in mice with targeted deletion of the
effects of apocynin. Free Radic Biol Med 40(2):236–246 NADPH oxidase subunit gp91PHOX. Basic Res Cardiol 101(2):
60. Hayashi H, Kobara M, Abe M, Tanaka N, Gouda E, Toba H, 127–132
Yamada H, Tatsumi T, Nakata T, Matsubara H (2008) Aldoste- 70. Dandapat A, Hu CP, Li D, Liu Y, Chen H, Hermonat PL, Mehta
rone nongenomically produces NADPH oxidase-dependent JL (2008) Overexpression of TGFbeta1 by adeno-associated virus
reactive oxygen species and induces myocyte apoptosis. Hyper- type-2 vector protects myocardium from ischemia-reperfusion
tens Res 31(2):363–375 injury. Gene Ther 15(6):415–423
61. Gilleron M, Marechal X, Montaigne D, Franczak J, Neviere R, 71. Matsushima S, Kinugawa S, Yokota T, Inoue N, Ohta Y, Ham-
Lancel S (2009) NADPH oxidases participate to doxorubicin- aguchi S, Tsutsui H (2009) Increased myocardial NAD(P)H
induced cardiac myocyte apoptosis. Biochem Biophys Res oxidase-derived superoxide causes the exacerbation of postinfarct
Commun 388(4):727–731 heart failure in type 2 diabetes. Am J Physiol Heart Circ Physiol
62. Shen E, Li Y, Li Y, Shan L, Zhu H, Feng Q, Arnold JM, Peng T 297(1):H409–H416
(2009) Rac1 is required for cardiomyocyte apoptosis during 72. Sadek MM, Haddad T, Haddad H (2009) The role of statins in
hyperglycemia. Diabetes 58(10):2386–2395 chronic heart failure. Curr Opin Cardiol 24(2):167–171

123

You might also like