Está en la página 1de 12

REVIEWS

BLOCKING NO SYNTHESIS:
HOW, WHERE AND WHY?
Patrick Vallance and James Leiper
Nitric oxide (NO) is a key physiological mediator, and the association of disordered NO
generation with many pathological conditions has led to much interest in pharmacologically
modulating NO levels. However, the wide range of processes in which NO has been implicated,
and the fact that increases or decreases in NO levels might be therapeutically desirable
depending on the condition or even at different stages of the same condition, pose
considerable challenges for drug development. Here, we focus on the rationale and potential
for approaches that reduce NO synthesis, which have led to the development of several
compounds that will shortly be entering clinical trials.

Nitric oxide (NO) is a mediator with protean functions. to activate the enzyme by sequestering iron, generating
It is involved in signalling in the cardiovascular, high-affinity binding sites for arginine and the essen-
gastrointestinal, genitourinary, respiratory and nervous tial cofactor tetrahydrobiopterin (BH4), and allowing
systems, and disordered NO generation has been electron transfer from the reductase-domain flavins
implicated in a wide range of diseases1–3. Therapies to the oxygenase-domain haem. Activity is also
based on increasing NO effects or donating exogenous dependent on bound calmodulin3. In iNOS, calmod-
NO hold promise, and have been discussed extensively ulin is tightly bound, whereas in eNOS and nNOS,
elsewhere2. However, several companies are develop- calmodulin binding is dependent on calcium, and
ing compounds that inhibit NO synthesis, and this is enzyme activity is therefore calcium dependent.
the focus of the present article. What is the rationale for Bound calmodulin is thought to enhance the rates of
reducing NO synthesis and which are the approaches electron transfer through the reductase domain to the
that could yield useful medicines? oxygenase domain.
In addition to regulation of NO synthesis by cofac-
Synthesis of NO tors and calcium, the activities of NOSs can be altered by
NO is synthesized from L-arginine in a reaction that is post-translational modifications and by protein–protein
catalysed by a family of enzymes — the NO synthases interactions. For example, eNOS activity is increased
(NOSs; FIG. 1). Three NOS isoforms have been identified by phosphorylation of Ser1179 (REF. 4), and is inhibited
and named according to the cell type or conditions in by interaction with the scaffolding domain of the mem-
Centre for Clinical which they were first described: endothelial NOS brane protein caveolin-1. These important areas of reg-
Pharmacology, (eNOS), neuronal NOS (nNOS) and inducible or ulation of NO synthesis could be targets for drug
British Heart Foundation inflammatory NOS (iNOS)1. These enzymes share action, but a detailed discussion is outside the scope of
(BHF) Laboratories,
Department of Medicine, ~50% sequence homology and catalyse the NADPH- this article (for review, see REF. 5).
University College London, and O2-dependent oxidation of L-arginine to NO and
5 University Street, London citrulline, with N ω-hydroxy-L-arginine formed as an Targets for NO
WC1E 6JJ, UK. intermediate3. NOSs are flavohaem enzymes that are The first described physiological target for NO was
Correspondence to P.V.
e-mail:
active only as dimers. Each monomer has a carboxy- soluble guanylyl cyclase6. Binding of NO to the iron
patrick.vallance@ucl.ac.uk terminal diflavin-reductase domain and an amino- within the haem moiety of guanylyl cyclase produces a
doi:10.1038/nrd960 terminal oxygenase domain. Dimerization is thought conformational change that leads to enzyme activation.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 1 | DECEMBER 2002 | 9 3 9


© 2002 Nature Publishing Group
REVIEWS

NO•

Haem Haem

H2N NH2 BH4 BH4

NH + NADPH, O2 FMN FMN

Zn
H2N COO–
FAD FAD
L-Arginine

H2N O

NH

CAL CAL
H2N COO–

Citrulline
Figure 1 | The NOS pathway. For enzymatic activity, nitric oxide synthase (NOS) enzymes must dimerize and bind the cofactors
tetrahydrobiopterin (BH4), haem, flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). On binding calmodulin (CAL),
the active enzyme catalyses the oxidation of L-arginine to citrulline and nitric oxide (NO) and requires molecular oxygen and NADPH
as co-substrates. Each NOS dimer coordinates a single zinc (Zn) atom.

The subsequent rise in cyclic GMP accounts for many of protective and harmful effects. The interaction of NO
the physiological effects of NO. However, NO has the with respiratory-chain enzymes illustrates many of these
potential to interact directly or indirectly with metals, mechanisms. For example, NO itself can bind reversibly
thiols and oxides, and affect proteins, nucleic acids, lipids to the haem enzyme cytochrome c oxidase, NO+-like
and sugars7 (TABLE 1). Indeed, because NO is a free radical reactions lead to reversible S-nitrosation of mitochon-
(it has an unpaired electron), and the nitrogen can exist drial complex I and generation of ONOO– can irre-
in various oxidation states to generate nitroxyl ions versibly inhibit multiple complexes, as well as aconitase.
(NO–), NO free radicals (NO•), nitrosonium cations These effects activate proton leak and the permeability
(NO+), nitrite ions (NO2–) or nitrate ions (NO3–), the transition pore, leading to cell death11.
biological chemistry of NO is complex, and its potential
effects within biological systems are many. There are NO in physiology
probably four main targets for reactions with NO in To understand the potential effects of pharmacological
cells: metals, reduced thiols, molecular oxygen and other inhibition of NOS isoforms, it is important to identify
reactive oxygen species (for example, superoxide (O2–)). the physiological processes that could be disrupted.
Which reactions are favoured will depend on the con-
centration of the reactants and the reaction rates8. eNOS. In the cardiovascular system, NO generation from
The fastest reaction rates are with superoxide ions to the endothelium is important to maintain the vasculature
form peroxynitrite (ONOO–), a powerful oxidant that in a relaxed state, inhibit the adhesion of platelets and
can modify proteins and lipids by nitration9,10. However, white cells and suppress the replication of smooth-muscle
the concentration of superoxide is usually kept low, and cells12. Pharmacological inhibition of NOS causes vaso-
so this reaction is probably not favoured. Reaction rates constriction, hypertension and enhanced platelet activa-
with metals are also high, but except for certain haem tion, and increases atherogenesis in animal models13. In
proteins, such as soluble guanylyl cyclase, the metal addition, eNos knockout mice are hypertensive, show
centres are generally not available for reaction with NO. exaggerated smooth-muscle replicative responses to vas-
Reactions with thiols are important, as S-nitrosation of cular injury, are more prone to atherogenesis and develop
specific cysteine residues (an NO+-like reaction) can aneurysms14 (FIG. 2, TABLE 2). eNOS is also important to
affect protein function in a reversible manner. Ion promote angiogenesis and regulate the expression of
channels, enzymes, transcription factors and G proteins vascular-endothelial growth factor (VEGF). So, prolonged
can all be S-nitrosated, and structural nitrosation motifs inhibition of eNOS activity is considered detrimental.
are being identified10. This means that NO signalling is
intimately bound to the redox state of the cell, and can nNOS. NO derived from nNOS in peripheral nerves is
affect many signalling pathways. These multiple actions also important in the relaxation of vascular and non-
of NO and dependence on thiol concentration and vascular smooth muscle15. It relaxes sphincters in the gut,
redox state probably explain why NO can have both mediates relaxation of the corpus cavernosum and

940 | DECEMBER 2002 | VOLUME 1 www.nature.com/reviews/drugdisc


© 2002 Nature Publishing Group
REVIEWS

Table 1 | Examples of modification of protein function by NO and related species


Target Reactant Example Effect References
Metal NO• Fe2+ in haem–guanylyl cyclase Enzyme activation 1
Fe–S cluster in aconitase Enzyme inhibition 92
Thiol NO+-like reaction Cys249 on DDAH Enzyme inhibition 91
(S-nitrosation)* Cys3656 on ryanodine receptor Sensitizes channel 93
to calcium
Cys69 on thioredoxin Enzyme activation 48
Tyrosine ONOO– (nitration) Tyrosines in ribonucleotide reductase Enzyme inhibition 94
NO• Tyrosines in prostaglandin H synthase Enzyme activation 95
*Nitrosation of cysteines is specific for certain cysteine residues held in the three-dimensional protein structure in a responsive state.
DDAH, dimethylarginine dimethylaminohydrolase; NO, nitric oxide.

thereby causes penile erection, relaxes the bladder and and inappropriate mounting behaviour, indicating that
urethra and alters responses in airways. Because splice NO derived from nNOS might be an important media-
variants of nNOS are still expressed in some tissues of tor of behavioural inhibition — the effects on behaviour
nNos knockout mice16, and highly selective nNOS are particularly evident in males16. The mice also show
inhibitors have not been used extensively, the data from abnormalities of balance and coordination at night
(non-selective) inhibitor studies and knockout pheno- (when visual cues are reduced), which are consistent
types (TABLE 2) are not totally concordant. Thus, owing to with the high levels of expression of nNOS in
the loss of sphincter relaxation, nNos knockout mice cerebellum16. Despite pharmacological observations
have dilated bladders and increased urinary frequency, indicating that nNOS might have a role in long-term
and show pyloric stenosis with grossly dilated potentiation (the process by which neurons ‘remember’),
stomachs17, but they still achieve penile erection, and nNos knockout mice seem to have normal memory and
strips of corpus cavernosum relax on electrical-field learning15. A novel nNos knockout that lacks the exon
stimulation18. In the central nervous system (CNS), that encodes the haem-binding domain and therefore
nNOS is widely expressed, but it has been less easy to encodes a global nNos knockout shows hypogonadism
determine its precise roles. A striking feature of the first and infertility19.
nNos knockout mouse line (in which alternative tran- nNOS is highly expressed in skeletal muscle20.
scripts can still be expressed) is an increase in aggression Studies with inhibitors indicate that it might have a role
in regulating muscle contraction, exercise-induced glu-
cose uptake and control of arteriolar tone within the
a b muscle. The nNOS protein is found in the dystrophin
complex, and a role has been proposed in muscular
dystrophy21. In the heart, nNOS and eNOS seem to have
different and sometimes opposing effects22. NO derived
from nNOS has a positive inotropic response, possibly
secondary to activation of sarcoplasmic-reticulum
calcium release, whereas eNOS activity decreases
β-adrenoceptor-induced contractility through effects
on L-type calcium channels. In the airways, nNOS
activity seems to provide important protection against
ovalbumin-induced airway hyperresponsiveness23.
For both eNOS and nNOS, it is clear that there are
sex differences in susceptibility to the effects of loss of
enzyme function24. This might be because expression of
both isoforms is modulated by sex hormones, and
therefore compensatory mechanisms might be more
evident in one or other sex. It is not clear whether such
gender-specific compensation would also occur with
long-term pharmacological inhibition of NOS. Unlike
eNOS, nNOS activity can also cause significant patho-
ApoE ko ApoE/eNos ko ApoE/eNos ko physiology (see below), and so inhibition of this iso-
form has the potential for benefit as well as harm.
Figure 2 | Loss of eNOS causes vascular disease. a | Intact aorta from a normal apolipoprotein E
(ApoE) knockout (ko) mouse and an ApoE/endothelial nitric oxide synthase (eNos) double iNOS. iNOS was first described in macrophages as a
knockout. The double knockout shows enhanced atherosclerosis (white) and an aneurysmal mechanism of macrophage cytotoxicity25. This isoform
dilation of the aorta. The inset on the right shows a higher magnification of the aneurysm. The
arrows show the origin of the left renal artery b | An aorta from a male ApoE/eNos double
is not usually expressed in healthy quiescent cells, but is
knockout shows a dissection that extends beyond the renal arteries. The arrows show the start rapidly transcriptionally induced in multiple cell types
and end of the dissection. Reproduced from REF. 14 © (2001), with permission from Lippincott, in response to stimulation with bacterial endotoxins or
Williams & Wilkins. pro-inflammatory cytokines. Once expressed, it generates

NATURE REVIEWS | DRUG DISCOVERY VOLUME 1 | DECEMBER 2002 | 9 4 1


© 2002 Nature Publishing Group
REVIEWS

large amounts of NO, and its activity is not dependent loss of iNOS is harmful in certain animal models of
on intracellular calcium26. It is becoming clear that inflammation31,32. Expression of iNOS is also impor-
there are species-, tissue- and cell-specific conditions tant for skin wound healing33 and healing of intestinal
for inducing the expression of active iNOS. Although mucosa, and it has been implicated as a key mediator
the protein is reasonably well conserved between of ischaemic preconditioning in the heart (a mecha-
species, the promoter region of the iNOS gene varies nism by which the heart is protected against lethal
greatly27. This difference is of functional importance, ischaemia)34. One possibility is that iNOS is not only
because it is far more difficult to induce expression of involved in the genesis of certain types of inflamma-
functionally active iNOS in human cells in vitro com- tion, but also in the resolution of inflammation. So, it
pared with rodent cells. Furthermore, there are five is probably oversimplistic to assume that eNOS and
copies of the iNOS gene in humans, but it is thought nNOS are physiological isoforms and that iNOS is
that only one is transcribed and translated into a func- involved solely with pathophysiology.
tionally active protein27. Therefore, the extrapolation of
studies in rodents to humans must be undertaken with Overproduction of NO in pathology
caution. However, it is clear that expression of iNOS is Increased generation of NO, either alone or in the
important for killing or host defence against certain presence of other free radicals, such as superoxide, has
protozoa, bacteria, fungi and viruses28. It is also impor- been implicated in pathophysiological changes in virtu-
tant in regulating cytokine production and influencing ally every organ system. Evidence comes from studies
T-helper type 1 (TH1)-cell expansion29. But iNOS is also of NO generation, NOS isoform expression and effects
induced in cells that are not obviously involved in host of NOS inhibitors (usually isoform non-selective).
defence, and in situations in which no live organism is Induction of iNOS is seen in models of septic shock27,
present, and it is in these situations that iNOS activity inflammatory and non-inflammatory pain35,36, arthritis37,
might be harmful and cause tissue damage. The over- inflammatory bowel disease38, asthma39,40 and in the brain
production of NO in response to infection might also after ischaemia or trauma, as well as in various models
be an important part of the tissue damage and destruc- of neurodegeneration or cerebral inflammation41,42. In
tion caused by the infection. the brain, the precise cellular distribution of iNOS
Many of the effects of increased NO generation might vary according to the nature of the insult, and
from iNOS are considered in terms of pathology (see iNOS expression has been described in neurons as well
below), but it seems unlikely that the isoform and the as glial cells, astrocytes and infiltrating neutrophils41–43.
NO it generates in non-immune cells exist solely to There is also evidence from clinical (human) samples
cause harm. Indeed, studies in iNos knockout mice that iNOS is expressed in various pathological tissues
(TABLE 2) have indicated physiological roles; for example, (TABLE 3), but it is interesting to note that in few cases
in osteoclastic bone resorption30. Furthermore, in the has functionally active iNOS definitively been shown
gut, iNOS can be expressed constitutively, and complete to be present. However, the potential importance of
iNOS in contributing to pathophysiology is exempli-
fied by the findings from iNos knockout mice (TABLE 2).
Table 2 | Some phenotypic features of NOS knockout mice iNOS is not the only the only isoform that can
Knockout Phenotype References cause a pathophysiological increase in NO. In particu-
mouse lar, increased nNOS activity is associated with certain
eNos–/– Elevated systemic and pulmonary blood pressure 96 types of neurotoxicity. NMDA (N-methyl-D-aspar-
Increased neointimal thickening after vessel injury 24 tate)-induced neurotoxicity is reduced by NOS
Aneurysm formation 14
Increased leukocyte adhesion 97 inhibitors in some models44, and stroke damage is
Increased cardiac ionotropic response 98 reduced in nNos knockout mice45. NOSs can also gen-
Increased tissue damage in experimental models of 99,100 erate superoxide through NADPH oxidation that is
stroke and global cerebral ischaemia
Reduced neurotransmitter release 101 uncoupled from NO generation3. nNOS seems to be
the isoform that is most likely to generate superoxide
nNos–/– Reduced tissue damage in experimental models of 45,102
stroke and global cerebral ischaemia when arginine or BH4 concentrations are low, or in
Reduced neurotransmitter release 101 the presence of certain arginine-based NOS inhibitors
Altered airway responsiveness 23,103 (see below).
Increased leukocyte adhesion 97
Increased stomach size (pyloric stenosis) 104 Is overproduction of NO from eNOS ever patho-
Gastroparesis, abnormal IJP and NANC relaxation in 105 logical? Although most interest has focused on the
GI smooth muscle cardioprotective effects of eNOS, there are some situa-
Urinary bladder urethral sphincter dysfunction 17
Hypogonadism and infertility 19
tions in which eNOS-derived NO might be harmful.
For example, in hyperoxia-induced retinopathy, eNOS
iNos–/– Increased susceptibility to pathogens 28,106,107
Resistance to sepsis-induced hypotension 106,107,108 deficiency seems to confer benefit46. Furthermore, in
Increased susceptibility to tumours 109 humans, some of the vasodilatation induced by pro-
Impaired osteoclastic bone resorption 30 inflammatory cytokines might be mediated by
Impaired wound healing 33,110 eNOS47, although whether this is protective or harm-
Increased tissue damage in models of lung injury 31
ful is not known. Overall, most data indicate that pro-
eNos, endothelial nitric oxide synthase; GI, gastrointestinal; IJP, inhibitory-junction potential;
iNos, inducible nitric oxide synthase; NANC, non-adrenergic, non-cholinergic; nNos, neuronal nitric longed inhibition of eNOS is likely to do more harm
oxide synthase. than good.

942 | DECEMBER 2002 | VOLUME 1 www.nature.com/reviews/drugdisc


© 2002 Nature Publishing Group
REVIEWS

Table 3 | Some human diseases in which iNOS has been detected inhibitor is the degree of selectivity over other NOS
isoforms, and specificity for NOS over other potential
Disease References
targets. The definition of isoform selectivity has been
Alzheimer’s disease 111
discussed extensively elsewhere3, but is important
Atherosclerosis 112,113 because some of the apparent contradictions in the
Celiac disease 114 literature relate to differing definitions. Some com-
Inflammatory bowel disease 115,116 pounds are apparently selective for one or other iso-
Multiple sclerosis 117 form in vivo, but show no real selectivity at the level of
Arthritis 118,119
the enzyme. For example, 7-nitroindazole (7-NI) is
widely quoted as a selective inhibitor of nNOS, but in
Parkinson’s disease 120
reality it is about equipotent as an inhibitor of all three
Septic shock 121 NOS isozymes (TABLE 4). Its apparent preferential effect
Stroke 122 on NO synthesis in the brain in vivo presumably
iNOS, inducible nitric oxide synthase. relates to a kinetic property of the molecule that allows
selective access or metabolism in certain tissues.
Similarly, there are many studies that have used
In addition to issues relating to isoform specificity of aminoguanidine as a selective inhibitor of iNOS 56,
inhibition, it is important to consider long-term effects although it has at best a tenfold selectivity over eNOS
of NOS inhibition. Much of the data relating to effects and perhaps fivefold selectivity over nNOS, and even
of NOS inhibitors are based on relatively short-term this selectivity might be lost if local concentrations of
studies, and long-term effects of cellular signalling BH4 are elevated47. Furthermore, aminoguanidine was
might be profound and affect several pathways. For initially developed as an inhibitor of advanced glyca-
example, thioredoxin, a redox regulator that is impor- tion end-product formation and also inhibits diamine
tant in cell growth and APOPTOSIS, is activated by NO48, oxidase in a similar concentration range. Because of
and presumably long-term inhibition of NO generation these difficulties, some of the claims of therapeutic
has the potential to increase cellular oxidative stress effects or potential adverse effects of selective inhibi-
through this mechanism. tion of NOS isoforms must be treated with caution.
The industry standard for defining iNOS selectivity
Dual effects of NO in vivo in rodents has been inhibition of endotoxin-
A glance at the NO literature reveals many apparently induced nitrate formation (a stable product of NO)
contradictory papers showing beneficial or harmful with no effect on arterial blood pressure. As arterial
effects of endogenous NO. NO can promote or inhibit blood pressure is well buffered, and inhibition of
apoptosis49, kill tumours or increase the potential for eNOS causes changes in vascular resistance (and pre-
metastasis or vascularization50, increase or protect sumably effects platelets, white cells and atherogenesis)
against damage after stroke51,52 and trigger ischaemic at doses far lower than those that affect blood pressure57,
preconditioning in the heart and mediate late effects of this screen is likely to overestimate the true degree of
preconditioning34,53. Some of these dual effects of NO selectivity for iNOS.
relate to different isoforms of NOS regulating different The selectivity of compounds over other targets is also
processes. For example, protective effects in stroke a potential problem. Several arginine metabolizing or
seem to be mediated by eNOS (vascular), whereas transporter proteins might be affected by substrate-based
harmful effects are due to nNOS activity (neuronal NOS inhibitors, and there is little information available at
toxicity)45,52. Other dual effects might relate simply to present on the effects on enzymes such as dimethylargi-
the amount of NO generated or the background redox nine dimethylaminohydrolase (DDAH), arginine glycine
state of the cell; at low concentrations, NO seems to be amidinotransferase, arginase, argininosuccinate synthase
anti-apoptotic, in part through inhibition of CASPASE or even peptide arginine deiminases.
activity by means of nitrosation, whereas at higher
concentrations, it can indirectly activate caspases54. Studies in humans. NOS inhibitors have been used in
The dual effects of NO present one of the biggest humans to explore mechanisms of physiology and
challenges for the development of potential inhibitors pathophysiology, and as potential treatments. In
— beneficial effects can be offset by harmful effects or patients with septic shock, the isoform non-specific
turn into harmful effects, depending on the underlying inhibitor NG-monomethyl-L-arginine (L-NMMA)
rates of NO synthesis. restores blood pressure and seems to improve haemo-
dynamics58,59 (FIG. 3). However, the largest study so far
Inhibition of NO showed an adverse effect on outcome. A post hoc
It seems clear that blocking NO has the potential to analysis indicated that low doses might have been
APOPTOSIS produce therapeutic benefit. The difficulties lie in beneficial and that the harm occurred when larger
Programmed cell death. achieving isoform specificity, in targeting to specific doses were used (and very high circulating concentra-
cells or tissues and in ensuring that the correct degree tions of L-NMMA were achieved). L-NMMA has also
CASPASES
A family of intracellular
of inhibition is achieved55. As expected, most interest been used to relieve headache 60, block excess NO
cysteine proteases that are has focused on iNOS inhibitors, but with substantial exhalation in asthma and reduce skin inflammation
responsible for apoptosis. interest also in nNOS inhibitors. A key issue for any induced by ultraviolet light (UVB)61.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 1 | DECEMBER 2002 | 9 4 3


© 2002 Nature Publishing Group
REVIEWS

Table 4 | Relative potencies and selectivity of NOS inhibitors


µM)
IC50 (µ Selectivity (fold)
Inhibitor iNOS nNOS eNOS iNOS vs nNOS iNOS vs eNOS nNOS vs eNOS
L-NNA 3.1 0.29 0.35 0.09 0.11 1.2
L-NMMA 6.6 4.9 3.5 0.7 0.5 0.7
7-NI 9.7 8.3 11.8 0.9 1.2 1.4
ARL 17477 0.33 0.07 1.6 0.2 5.0 23.0
Aminoguanidine 31 170 330 5.5 11 1.9
L-NIL 1.6 37 49 23 9 1.3
PBITU 0.047 9 190
(bis-isothiourea)
vinyl L-NIO 60 0.1 12.0 0.002 5 120
1400W 0.23 7.3 1,000 32 >4,000 >130
GW273629 8.0 630 1,000 78 >125 >1.6
GW274150 1.4 145 466 104 333 3.2
AR-C102222 0.04 >100 2 50 >1,000 >50
The absolute potencies might not be directly comparable because of the experimental conditions under which studies were done.
eNOS, endothelial nitric oxide synthase; IC50, half-maximal inhibitory concentration; iNOS, inflammatory nitric oxide synthase;
7-NI, 7-nitroindazole; L-NIL, N(6)-(1-iminoethyl)lysine; L-NIO, N-iminoethyl-L-ornithine; L-NMMA, NG-monomethyl-L-arginine;
G
L-NNA, N -nitro-L-arginine; nNOS, neuronal nitric oxide synthase; PBITU, S,S′-1,3-phenylene-bis(1,2-ethanediyl)bis-isothiourea.
Data adapted from REF. 2.

Substrate analogues. The first described inhibitors of Although these compounds have some selectivity for
NOS were analogues of L-arginine62. These compounds iNOS over eNOS, their development as drugs was lim-
bind at the arginine-binding site and are competitive ited by toxic effects, including actions on Na+/K+
with arginine. However, some of them are also metabo- ATPase64. Crystal structures of the eNOS haem domain
lized by NOS, and active enzyme and NADPH are with S-alkyl-isothioureas show that the thiourea group
required for inhibition to proceed from weak binding to occupies the same position as the guanidine group of
potent inhibition. The first highly selective iNOS L-arginine. Binding studies with more bulky ligands led
inhibitors described were the bis-isothioureas63. to the suggestion that an isoform-selective inhibitor
would have three characteristics64. First, there should
120
be a structural scaffold that provides a guanidino,
amidino or ureido group that donates hydrogen
SVR 666
110 CO 10.7 bonds to the glutamate residue in the NOS active site.
HR 119 There should also be a small hydrophobic group, such
100
614 as an alkyl or thienyl group, as part of the scaffold to
607 10.9
10.3 573 120
provide further non-polar interaction with the pro-
90
95 575 10.1 tein opposite to the glutamate residue. Second, it
536 122
Mean arterial pressure (mm Hg)

9.7 10.2 should have an isoform-selectivity-conferring func-


110 511
80 9.5
122 tional group bearing hydrogen-bonding capability that
135 519 can reach into the substrate-access channel remote from
487 10.2
70 10.1 123
the active site. Such a group could take advantage of the
123 amino-acid differences in this channel between iso-
367
60 12.2 3
forms. Third, it should have a linker between the scaf-
122 2
fold and the functional group of appropriate length and
391 flexibility to reach isoform-specific regions.
50 8.3 1
105 Despite the high degree of similarity between the
0.30
40
arginine-binding sites of NOS isoforms65, highly selec-
0.16 0.10
0.02 tive iNOS inhibitors have been developed that compete
30
with arginine and presumably bind to this site. For
example, the compound 1400W (FIG. 4) is ~10,000-fold
–5 –4 –3 –2 –1 0 10 20 30 40 50 60 70
selective for iNOS over eNOS and at least 30-fold selec-
Time before first L-NMMA bolus (hr) Time after first L-NMMA bolus (min)
tive for iNOS compared with nNOS66. Once again, this
Figure 3 | Effects of L-NMMA in a patient with severe septic shock. The patient was treated seems to be a ‘mechanism-based’ inhibitor, the action
initially with noradrenaline to try to maintain blood pressure (yellow columns show dose in µg kg–1 of which is dependent on NADPH and develops
min–1). When this approach failed, he was treated with three bolus doses of L-NMMA (1 = 0.3 mg
slowly. Interestingly, whereas the effects of 1400W on
kg–1; 2 = 0.3 mg kg–1 and 3 = 1 mg kg–1). The L-NMMA restored blood pressure, and this was due
to an increase in systemic vascular resistance (SVR), with no significant change in cardiac output iNOS develop slowly and reverse only slowly, the
(CO) or heart rate (HR). L-NMMA, NG-monomethyl-L-arginine. Redrawn from REF. 58 © (1991), with effects on eNOS and nNOS are rapidly reversible by
permission from Elsevier Science. arginine. Although this compound has toxicity that

944 | DECEMBER 2002 | VOLUME 1 www.nature.com/reviews/drugdisc


© 2002 Nature Publishing Group
REVIEWS

prevents its administration to humans, it has been use- (L-NIL; FIG. 4), and this compound has ~30-fold selectiv-
ful to define some of the potential for selective iNOS ity for iNOS over the other two isoforms. A PRODRUG of
70
inhibition in animal models. Other compounds in this L-NIL has been developed that again exerts beneficial
series include GW273629 and GW274150. These com- effects in carageenan and arthritis models and is reported
pounds have been shown to reduce experimental post- to decrease exhaled NO in asthmatic humans. Whether
operative ileus and have some beneficial effects in the degree of isoform selectivity achieved will be suffi-
models of gastrointestinal damage67. cient remains to be determined. One thing that has
An alternative chemical approach has been to become clear from the substrate-based inhibitor studies is
develop spiroquinazolone derivatives68. The Astra- that achieving selectivity for iNOS over nNOS is more
Zeneca compound AR-C102222 has a half-maximal difficult than achieving selectivity for iNOS over eNOS.
inhibitory concentration (IC50) of ~0.04 µM, and is
reported to be highly selective for iNOS. In vivo, it is Dimerization inhibitors. An alternative approach is to
effective in reducing inflammation in the standard block enzyme dimerization71. Pyrimidineimidazole-
carageenan paw oedema model and decreases adjuvant- based compounds have been described (FIG. 4) that
induced arthritis (including joint destruction) in rats. coordinate the haem in the iNOS monomer, disrupt
A NOS inhibitor that has been used experimentally one of the helices and displace a crucial arginine-binding
for many years is L-N(6)-(1-iminoethyl)lysine69,70 residue from the active site71,72. Essentially, the effect is

a Substrate-based inhibitors

NH NH NH NH
NH2
CO2H CO2H
N N N N N NH2
N CO2H
H H H H
NH2 NH2 H

L-NMMA ADMA L-NIL 1400W

O
F
H N NH NH
N O NH2
S S CO2H
N N
N N N CO2H
H O H
NH2
F NH2

AR-C102222 GW273629 GW274150

b Dimerization inhibitors

R
N N Cl
O

N O N
N
H N
Cl
N O
N
Cl O
X N N
N
Cl Cl

Pyrimidineimidazole-based Clotrimidazole Miconazole

c Cofactor inhibitors d Isothioureas

O
N S S
PRODRUG N HN NH
A pharmacologically inactive N O
NH2 H2N
compound that is converted to
H2N N N
the active form of the drug by
endogenous enzymes or Aminopteridine 1,4-PBITU
metabolism. It is generally
designed to overcome problems
associated with stability, toxicity, Figure 4 | Inhibitors of NOS. a | Substrate-based inhibitors. b | Dimerization inhibitors. c | Cofactor inhibitors. d | Isothioureas.
lack of specificity or limited ADMA, asymmetric dimethylarginine; L-NIL, L-N(6)-(1-iminoethyl)lysine; L-NMMA, NG-monomethyl-L-arginine; 1,4-PBITU,
(oral) bioavailability. S,S′-1,4-phenylene-bis(1,2-ethanediyl)bis-isothiourea.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 1 | DECEMBER 2002 | 9 4 5


© 2002 Nature Publishing Group
REVIEWS

to block the formation of both the arginine- and the inhibits nNOS and seems to offer protection in an
BH4-binding sites and produce an inhibitor–monomer animal model of cerebral ischaemia78. However, this
complex. These effects are probably similar to those pro- compound shows only modest selectivity for nNOS
duced by the antifungal imidazoles clotrimidazole and over iNOS, and so some of the effects might also have
miconazole (FIG. 4), although these compounds also pre- been due to inhibition of iNOS. As more selective
vent calmodulin binding to iNOS73,74. Once again, the inhibitors of nNOS emerge, the full potential for thera-
problem is to achieve selectivity for iNOS over nNOS. peutic and unwanted effects should become clearer.
The dimerization inhibitors show some specific affinity Vinyl N-iminoethyl-L-ornithine (L-NIO)79 has substan-
for iNOS and nNOS monomers and have weak affinity tial selectivity for nNOS over iNOS and eNOS (TABLE 4),
for eNOS monomers, but the molecular basis for this and should be useful to define the potential for thera-
effect is not clear. One possibility is that part of the effect peutic manipulation of nNOS, as should inhibitors of
that is seen in vitro is dependent on the stability of the nNOS based on the pterin (BH4) site75.
dimers, and because eNOS dimers are more stable, some
of the selectivity for certain compounds is apparent Transcriptional regulation. Inhibition of transcription of
rather than real. This class of compound is also known to iNOS by glucocorticoids accounts for some of the actions
inhibit cytochrome P450 enzymes, which could present a of these established medicines80. Whether more selective
problem in terms of unwanted effects. Dimerization inhibition of iNOS transcription can be achieved is not
inhibitors have been shown to have potentially beneficial clear. There is interest in transcriptional upregulation of
effects in the usual screens of endotoxin-induced nitrate eNOS, but this is outside the scope of this article.
formation and adjuvant arthritis, and also in experimen-
tal allergic encephalomyelitis as a model of demyelinat- Alternative molecular targets
ing disease. The newer compounds seem to be highly Reducing harmful overproduction of NO clearly has
selective over eNOS and nNOS for time courses longer therapeutic potential. Inhibition of iNOS would seem to
than the half-life of dimers, but it remains to be deter- have use as an anti-inflammatory strategy in a wide
mined whether the apparent isoform selectivity can be range of disease states. Inhibition of nNOS also has
maintained chronically in vivo. potential in CNS disorders associated with neuro-
degeneration and neurotoxicity, as well as in pain models.
Cofactor blockers. Inhibitors that affect flavoproteins or However, inhibition of these isoforms might also
calmodulin binding have been developed, but it seems cause harm. Inhibition of iNOS might be expected to
unlikely that such compounds will have the necessary increase susceptibility to infections, inhibit wound heal-
selectivity or specificity to be useful as approaches for ing and exacerbate tissue damage in certain situations.
drug discovery. A series of inhibitors has been made Inhibition of nNOS might alter gastrointestinal and
that targets the BH4-binding site75. Differences in the genitourinary function, cause spasm of sphincters and
BH4-binding site between NOS isoforms have been affect higher cerebral functions. For prolonged use, a high
identified, and antipterin compounds have been devel- degree of isoform selectivity is essential to avoid the
oped that show tenfold selectivity for nNOS over the unwanted effects of inhibiting eNOS, which include
other two isoforms75. hypertension, enhanced white-cell and platelet activation,
and increased atherogenesis. It is also clear that complete
Protein–protein interactions. Various endogenous inhibition of NOS (even if isoform specific) is probably
peptides and proteins that bind to NOS isoforms and undesirable, and that partial inhibition and/or tissue or
modify activity have been identified. For example, the cell-specific inhibition might be the ultimate goals.
89-amino-acid protein termed protein inhibitor of Interestingly, the first described NOS inhibitor,
NOS (PIN) binds to the amino-terminal extension of L-NMMA, is a naturally occurring amino acid that is
nNOS and inhibits activity76. Other inhibitory domains generated endogenously81. Arginine residues on proteins
on eNOS and nNOS have been identified, but whether are methylated by the action of protein arginine
these can be targets for selective inhibition remains to be methyltransferases (PRMTs). Three methylarginines
determined. In vivo delivery of the caveolin-1 scaffold- are generated: L-NMMA, asymmetric dimethylarginine
ing domain inhibits eNOS activity, showing that, in (ADMA) and symmetric dimethylarginine (SDMA)82.
principle, this approach works. The asymmetrically methylated arginine residues
(ADMA and L-NMMA) inhibit all three isoforms of
nNOS inhibitors. There is a rationale for assuming that NOS, whereas SDMA is not an NOS inhibitor.
nNOS inhibitors might be of value in stroke, in certain Normally, the concentration of ADMA and L-NMMA
types of neurodegeneration and as analgesics (partic- seems to be kept low by the action of DDAH83,84, which
ularly, but not exclusively, in the setting of inflammation). catalyses their conversion to citrulline, and dimethyl-
Although the peripheral effects of nNOS inhibition can amine or monomethylamine, respectively (FIG. 5). This
include substantial gastrointestinal and genitourinary pathway represents an endogenous mechanism for the
effects (see above), the existence of variants of nNOS regulation of NO production by competitive inhibition.
raises the possibility of further tissue specificity. Various Indeed, pharmacological inhibition of DDAH causes a
dipeptide amines and peptidomimetic inhibitors of rise in ADMA levels sufficient to block NO generation85,
nNOS have been described, but some interfere with and this raises the possibility of a novel approach to
other arginine pathways77. The compound ARL 17477 blocking overproduction of NO.

946 | DECEMBER 2002 | VOLUME 1 www.nature.com/reviews/drugdisc


© 2002 Nature Publishing Group
REVIEWS

CIT
a

R
R
R Me
R R
(Me)2 R

R R
R
Me NO•
Me

Me
(Me)2
Competitive
Proteolysis R (Me)2 inhibition
R
Me
(Me)2 R
R
R

DDAH DDAH b
Me Me Me
Me Me

HN NH HN NH HN N H2N O

NH NH NH NH Me Me
Renal
clearance + NH

H2N COO– H2N COO– H2N COO– H2N COO–

SDMA L-NMMA ADMA Citrulline


Figure 5 | The DDAH/NOS pathway. a | Arginine residues (R) within proteins can be post-translationally methylated by a family of
protein arginine methyltransferases (PRMTs). Proteolysis of arginine-methylated proteins releases free methylarginine into the
cytosol. In mammalian cells, three forms of methylarginine are found: symmetric dimethylarginine (SDMA), NG-monomethyl-L-
arginine (L-NMMA) and asymmetric dimethylarginine (ADMA). Of these, only the asymmetrically methylated forms (L-NMMA and
ADMA) are inhibitors of nitric oxide synthase (NOS). These residues also differ in their routes of clearance; whereas SDMA is cleared
by the kidney without further metabolism, L-NMMA and ADMA are hydrolysed to citrulline (CIT) and methylamines by the action of
dimethylarginine dimethylaminohydrolase (DDAH). Accumulation of asymmetrically methylated arginine residues leads to inhibition
of nitric oxide (NO) synthesis, as the residues compete with arginine for the active site of NOS. NO can inhibit DDAH activity by
S-nitrosation91. b | Crystal structure of DDAH with substrate (Protein Data Bank code 1h70).

Two isoforms of DDAH have been identified84, one reduced in patients with Alzheimer’s disease87. More
with a predominantly neuronal distribution (DDAH1) generalized inhibition of DDAH might allow L-NMMA
and the other with a more widespread and vascular dis- and ADMA levels to rise sufficiently to inhibit partially
tribution (DDAH2). Isoform-selective inhibition of the excess NO generation seen in septic shock (or
DDAH might provide a different profile of NOS inhibi- other types of inflammation), a condition in which
tion to direct inhibition of NOS isozymes. Clearly, inhi- low doses of L-NMMA might be beneficial, whereas
bition of DDAH would not provide isoform-selective larger doses are detrimental. The ADMA/L-NMMA
inhibition of NOS, as ADMA and L-NMMA block all binding site of DDAH is sufficiently different to that of
three isoforms. It is also unlikely that endogenous NOSs for a selective inhibitor to be developed88.
ADMA or L-NMMA would ever increase to levels suffi- The notion of preferentially inhibiting excess NO
cient to inhibit NOS by more than 30%85. However, inhi- generation while leaving physiological NO production
bition of DDAH might provide tissue-specific partial untouched also underlies the approach of targeting
inhibition of NOS. It is conceivable that this could be of enzymes that allow regeneration of arginine within
value to limit excess production of NO without the dan- cells. Arginosuccinate-synthase activity seems to be
ger inherent in using direct NOS inhibitors of causing required for high-output NO synthesis, at least in some
near total inhibition of NO. Specifically, inhibition of situations, and therefore might provide a target for drug
DDAH1 would be expected to be of value in neuro- action89. An alternative approach that could emerge is to
degeneration and possibly in pain. Consistent with this, target individual proteins on which NO acts to inhibit
DDAH1 expression is increased in models of neuronal specific adverse effects of NO while leaving other NO
injury86, and ADMA levels have been reported to be signalling pathways intact.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 1 | DECEMBER 2002 | 9 4 7


© 2002 Nature Publishing Group
REVIEWS

Into the clinic? skin diseases and other indications, and it will be inter-
The identification of NO as a biological mediator has esting to see which are chosen. Selective inhibitors of
led to vast numbers of publications — in the order of nNOS are also in development, and will help to answer
3,000 per year at present. There can be little doubt that important questions about potential therapeutic appli-
NOS activity is of considerable importance physiologi- cations of nNOS blockade. In summary, the current
cally, or that overproduction of NO or increased NOS position seems to be as follows. First, partial inhibition
activity has the potential to cause harm. However, part of NO generation is probably preferable to complete
of the problem in turning scientific discovery into useful inhibition, whichever isoform is targeted. Second, the
therapeutics has been the sheer range of processes in desired and undesired effects of selective iNOS inhibi-
which NO has been implicated, and the opposing effects tion might change with time depending on the stage of
of NO even within a single disease. A good example is inflammation — initiation, chronic or resolving. Third,
provided in the field of cancer biology. High-output NO the range of effects (desired and undesired) of iNOS or
generation from macrophages or from tumour cells nNOS inhibitors is likely to be broad, and the idea that
themselves can be tumoricidal and prevent metastasis. iNOS inhibitors will be ‘glucocorticoids without the side
However, iNOS is expressed constitutively in some effects’ seems misplaced. Fourth, the full range of dis-
tumour cells, where it promotes tumour growth, neo- eases for which NOS inhibition might be beneficial is
vascularization and invasiveness through induction of unlikely to become clear until, and unless, one or more
mutations to the tumour-suppressor gene TP53 and compounds have been in clinical use for some years and
upregulation of VEGF expression28. Furthermore, in clinical researchers have had a chance to explore their
other tumours, it is eNOS expression that correlates therapeutic effect. And finally, novel approaches to
with tumour malignancy and vascularity90. How can achieving isoform, cell or tissue selectivity of NOS inhi-
selective benefit be gained without a welter of unwanted bition might lead to more specific therapies.
effects, and which is the single therapeutic target at
which the pharmaceutical industry should aim? Note added in proof
The field of NO therapeutics is now entering a cru- Long-term treatment with N ω-nitro-L-arginine methyl
cial and exciting stage. Selective inhibitors of iNOS have ester enhances atherosclerosis in eNos knockout mice,
been identified and will enter clinical trials shortly. indicating that some substrate-based NOS inhibitors
Companies are considering, or have considered, asthma, might have further detrimental effects independent of
arthritis, inflammatory bowel disease, inflammatory eNOS inhibition123.

1. Hanafy, K. A., Krumenacker, J. S. & Murad, F. 13. Cayatte, A. J., Palacino, J. J., Horten, K. & Cohen, R. A. 25. Hibbs, J. B. Jr, Taintor, R. R., Vavrin, Z. & Rachlin, E. M. Nitric
NO, nitrotyrosine, and cyclic GMP in signal transduction. Chronic inhibition of nitric oxide production accelerates oxide: a cytotoxic activated macrophage effector molecule.
Med. Sci. Monit. 7, 801–819 (2001). neointima formation and impairs endothelial function in Biochem. Biophys. Res. Commun. 157, 87–94 (1988).
2. Ignarro, L. J., Napoli, C. & Loscalzo, J. Nitric oxide donors hypercholesterolemic rabbits. Arterioscler. Thromb. 14, 26. Marletta, M. A. Nitric oxide synthase: aspects concerning
and cardiovascular agents modulating the bioactivity of nitric 753–759 (1994). structure and catalysis. Cell 78, 927–930 (1994).
oxide. Circ. Res. 90, 21–28 (2002). 14. Kuhlencordt, P. J. et al. Accelerated atherosclerosis, aortic 27. Vallance, P. & Charles, I. Nitric oxide in sepsis: of mice and
3. Alderton, W. K., Cooper, C. E. & Knowles, R. G. Nitric oxide aneurysm formation, and ischaemic heart disease in men. Sepsis 1, 93–100 (1998).
synthases: structure, function and inhibition. Biochem. J. apolipoprotein E/endothelial nitric oxide synthase double- 28. Bogdan, C. Nitric oxide and the immune response. Nature
357, 593–615 (2001). knockout mice. Circulation 104, 448–454 (2001). Immunol. 2, 907–916 (2001).
A comprehensive review of the biochemistry and 15. Rand, M. J. Nitrergic transmission: nitric oxide as a mediator The pleiotropic effects of NO are nowhere more
some structural aspects of NOSs. of non-adrenergic, non-cholinergic neuro-effector evident than in the immune system. This review
4. Cabe, T. J., Fulton, D., Roman, L. J. & Sessa, W. C. transmission. Clin. Exp. Pharmacol. Physiol. 19, 147–169 outlines these effects (some of which seem to oppose
Enhanced electron flux and reduced calmodulin (1992). each other), describes some of the controversy about
dissociation may explain ‘calcium-independent’ eNOS 16. Baranano, D. E. & Snyder, S. H. Neural roles for heme iNOS in humans and discusses the problems that face
activation by phosphorylation. J. Biol. Chem. 275, oxygenase: contrasts to nitric oxide synthase. Proc. Natl NOS inhibitors.
6123–6128 (2000). Acad. Sci. USA 98, 10996–11002 (2001). 29. Niedbala, W., Wei, X. Q., Piedrafita, D., Xu, D. & Liew, F. Y.
5. Lowenstein, C. J. Purification and assessment of proteins 17. Burnett, A. L. et al. Urinary bladder-urethral sphincter Effects of nitric oxide on the induction and differentiation of
associated with nitric oxide synthase. Methods Enzymol. dysfunction in mice with targeted disruption of neuronal TH1 cells. Eur. J. Immunol. 29, 2498–2505 (1999).
353, 233–240 (2002). nitric oxide synthase models idiopathic voiding disorders in 30. van’t Hof, R. J. et al. Requirement of the inducible nitric oxide
6. Ignarro, L. J., Cirino, G., Casini, A. & Napoli, C. Nitric oxide humans. Nature Med. 3, 571–574 (1997). synthase pathway for IL-1-induced osteoclastic bone
as a signaling molecule in the vascular system: an overview. 18. Burnett, A. L. et al. Nitric oxide-dependent penile erection in resorption. Proc. Natl Acad. Sci. USA 97, 7993–7998 (2000).
J. Cardiovasc. Pharmacol. 34, 879–886 (1999). mice lacking neuronal nitric oxide synthase. Mol. Med. 2, 31. Kenyon, N. J. et al. Susceptibility to ozone-induced acute
7. Davis, K. L., Martin, E., Turko, I. V. & Murad, F. Novel effects 288–296 (1996). lung injury in iNOS-deficient mice. Am. J. Physiol. 282,
of nitric oxide. Annu. Rev. Pharmacol. Toxicol. 41, 203–236 19. Gyurko, R., Leupen, S. & Huang, P. L. Deletion of exon 6 of L540–L545 (2002).
(2001). the neuronal nitric oxide synthase gene in mice results in 32. Arnett, H. A. et al. The protective role of nitric oxide in a
8. Ischiropoulos, H. & Gow, A. J. Nitric oxide chemistry and hypogonadism and infertility. Endocrinology 143, neurotoxicant-induced demyelinating model. J. Immunol.
cellular signaling. J. Cell. Physiol. 187, 277–282 (2001). 2767–2774 (2002). 168, 427–433 (2002).
9. O’Donnell, V. B. et al. Nitration of unsaturated fatty acids by 20. Kobzik, L., Reid, M. B., Bredt, D. S. & Stamler, J. S. Nitric 33. Kane, A. J. et al. Inducible nitric oxide synthase (iNOS)
nitric oxide-derived reactive species. Methods Enzymol. oxide in skeletal muscle. Nature 8, 546–548 (1994). activity promotes ischaemic skin flap survival. Br. J.
301, 454–470 (1999). 21. Brenman, J. E., Chao, D. S., Xia, H., Aldape, K. & Bredt, D. S. Pharmacol. 132, 1631–1638 (2001).
10. Stamler, J. S., Lamas, S. & Fang, F. C. Nitrosylation, the Nitric oxide synthase complexed with dystrophin and absent 34. Nandagopal, K., Dawson, T. M. & Dawson, V. L. Critical role
prototypic redox-based signaling mechanism. Cell 106, from skeletal muscle sarcolemma in Duchenne muscular for nitric oxide signaling in cardiac and neuronal ischaemic
675–683 (2001). dystrophy. Cell 8, 743–752 (1995). preconditioning and tolerance. J. Pharmacol. Exp Ther. 297,
The recognition that NO might affect many proteins 22. Barouch, L. A. et al. Nitric oxide regulates the heart by 474–478 (2002).
through nitrosation reactions increases the range of spatial confinement of nitric oxide synthase isoforms. Nature 35. Luo, Z. D. & Cizkova, D. The role of nitric oxide in
effects of NO and the possibility that these will vary 416, 337–339 (2002). nociception. Curr. Rev. Pain 4, 459–466 (2000).
with the redox state of the cell. This clearly has 23. De Sanctis, G. T. et al. Contribution of nitric oxide synthases 36. Saban, M. R., Nguyen, N. B., Hammond, T. G. & Saban, R.
implications for understanding the long-term effects 1, 2, and 3 to airway hyperresponsiveness and inflammation Gene expression profiling of mouse bladder inflammatory
of inhibitors. in a murine model of asthma. J. Exp. Med. 189, 1621–1629 responses to LPS, substance P, and antigen-stimulation.
11. Brown, G. C. Regulation of mitochondrial respiration by nitric (1999). Am. J. Pathol. 160, 2095–2110 (2002).
oxide inhibition of cytochrome c oxidase. Biochim. Biophys. 24. Moroi, M. et al. Interaction of genetic deficiency of 37. van’t Hof, R. J. & Ralston, S. H. Nitric oxide and bone.
Acta 1504, 46–57 (2001). endothelial nitric oxide, gender, and pregnancy in vascular Immunology 103, 255–261 (2001).
12. Napoli, C. & Ignarro, L. J. Nitric oxide and atherosclerosis. response to injury in mice. J. Clin. Invest. 101, 1225–1232 38. Kubes, P. & McCafferty, D. M. Nitric oxide and intestinal
Nitric Oxide 5, 88–97 (2001). (1998). inflammation. Am. J. Med. 109, 150–158 (2000).

948 | DECEMBER 2002 | VOLUME 1 www.nature.com/reviews/drugdisc


© 2002 Nature Publishing Group
REVIEWS

39. Sanders, S. P. Nitric oxide in asthma: pathogenic, A structural analysis of the binding of NOS inhibitors. localization in the rat brain and its coincident up-regulation
therapeutic or diagnostic? Am. J. Respir. Cell Mol. Biol. 21, This paper begins to define some of the potential with neuronal NO synthase (nNOS) in axotomized
147–149 (1999). structural requirements for isoform-selective motoneurons. Eur. J. Neurosci. 11, 2160–2166 (1999).
40. Silkoff, P. E., Robbins, R. A., Gaston, B., Lundberg, J. O. & inhibition of NOS. 87. Abe, T., Toghi, H., Murata, T., Isobe, C. & Sato, C. Reduction
Townley, R. G. Endogenous nitric oxide in allergic airway 65. Li, H. et al. Crystal structures of zinc-free and bound heme is asymmetrical dimethylarginine, an endogenous nitric
disease. J. Allergy Clin. Immunol. 105, 438–448 (2000). domain of human inducible nitric oxide synthase: oxide synthase inhibitor, in the cerebrospinal fluid during
41. Iravani, M. M., Kashefi, K., Mander, P., Rose, S. & Jenner, P. implications for dimer stability and comparison with aging and in patients with Alzheimer’s disease. Neurosci.
Involvement of inducible nitric oxide synthase in endothelial nitric oxide synthase. J. Biol. Chem. 274, Lett. 312, 177–179 (2001).
inflammation-induced dopaminergic neurodegeneration. 21276–21284 (1999). 88. Murray-Rust, J. et al. Structural insights into the hydrolysis of
Neuroscience 110, 49–58 (2002). 66. Garvey, E. P. et al. 1400W is a slow, tight binding, and highly cellular nitric oxide synthase inhibitors by dimethylarginine
42. Wada, K., Chatzipanteli, K., Kraydieh, S., Bustol, R. & selective inhibitor of inducible nitric oxide synthase in vitro dimethylaminohydrolase. Nature Struct. Biol. 8, 679–683
Dietrich, W. D. Inducible nitric oxide synthase expression and in vivo. J. Biol. Chem. 272, 4959–4963 (1997). (2001).
after traumatic brain injury and neuroprotection with 67. Evans, S. M. & Whittle, B. J. Interactive roles of superoxide Blocking DDAH leads to an accumulation of
aminoguanidine treatment in rats. Neurosurgery 43, and inducible nitric oxide synthase in rat intestinal injury naturally occurring NOS inhibitors. This paper
1427–1436 (1998). provoked by non-steroidal anti-inflammatory drugs. Eur. J. provides structural insight into enzyme function
43. Heneka, M. T. & Feinstein, D. L. Expression and function of Pharmacol. 429, 287–296 (2001). and possibilities for inhibition of DDAH as a route
inducible nitric oxide synthase in neurons. J. Neuroimmunol. 68. Beaton, H., Hamley, P., Nicholls, D. J., Tinker, A. C. & to affect NO generation.
114, 8–18 (2001). Wallace, A. V. 3,4-Dihydro-1-isoquinolinamines: a novel 89. Xie, L., Hattori, Y., Tume, N. & Gross, S. S. The preferred
44. Izumi, Y., Benz, A. M., Clifford, D. B. & Zorumski, C. F. class of nitric oxide synthase inhibitors with a range of source of arginine for high-output nitric oxide synthesis in
Nitric oxide inhibitors attenuate N-methyl-D-aspartate isoform selectivity and potency. Biorganic Med. Chem. Lett. blood vessels. Semin. Perinatol. 24, 42–45 (2000).
excitotoxicity in rat hippocampal slices. Neurosci. Lett. 11, 1023–1026 (2001). 90. Iwata, S. et al. Endothelial nitric oxide synthase expression in
135, 227–230 (1992). This paper describes a new class of selective iNOS tumor vasculature is correlated with malignancy in human
45. Huang, Z. et al. Effects of cerebral ischaemia in mice inhibitors. A member of this class is apparently close supratentorial astrocytic tumors. Neurosurgery 45, 24–28
deficient in neuronal nitric oxide synthase. Science 265, to entering clinical trials. (1999).
1883–1885 (1994). 69. Moore, W. M. et al. L-N6-(1-iminoethyl)lysine: a selective 91. Leiper, J., Murray-Rust, J., MacDonald, N. & Vallance, P.
The original paper that identified the neurotoxic inhibitor of inducible nitric oxide synthase. J. Med. Chem. S-nitrosylation of dimethylarginine dimethylaminohydrolase
effects of nNOS and proposed a protective role for 37, 3886–3888 (1994). regulates enzyme activity: further interactions between nitric
eNOS in stroke. 70. Moore, W. M. et al. Synthesis and biological characterization oxide synthase and ADMA pathways. Proc. Natl Acad. Sci.
46. Ando, A. et al. Nitric oxide is proangiogenic in the retina and of L-N 6-(1-iminoethyl)lysine 5-tetrazole-amide, a prodrug of a USA 99, 13527–13532 (2002).
choroid. J. Cell. Physiol. 191, 116–124 (2002). selective iNOS inhibitor. J. Med. Chem. 45, 1686–1689 92. Drapier, J. C. & Hibbs, J. B. Jr. Aconitases: a class of
47. Bhagat, K., Hingorani, A. D., Palacios, M., Charles, I. G. & (2002). metalloproteins highly sensitive to nitric oxide synthesis.
Vallance P. Cytokine-induced venodilatation in humans in 71. McMillan, K. et al. Allosteric inhibitors of inducible nitric oxide Methods Enzymol. 269, 26–36 (1996).
vivo: eNOS masquerading as iNOS. Cardiovasc. Res. 41, synthase dimerization discovered via combinatorial 93. Sun, J., Xin, C., Eu, J. P., Stamler, J. S. & Meissner, G.
754–764 (1999). chemistry. Proc. Natl Acad. Sci. USA 97, 1506–1511 Cysteine-3635 is responsible for skeletal muscle ryanodine
48. Haendeler, J. et al. Redox regulatory and anti-apoptotic (2000). receptor modulation by NO. Proc. Natl Acad. Sci. USA 98,
functions of thioredoxin depend on S-nitrosylation at 72. Blasko, E. et al. Mechanistic studies with potent and 11158–11162 (2001).
cysteine 69. Nature Cell Biol. 4, 743–749 (2002). selective inducible nitric oxide synthase dimerization 94. Guittet, O. et al. Differential sensitivity of the tyrosyl radical of
49. Moncada, S. & Erusalimsky, J. D. Does nitric oxide modulate inhibitors. J. Biol. Chem. 277, 295–302 (2002). mouse ribonucleotide reductase to nitric oxide and
mitochondrial energy generation and apoptosis? Nature 73. Wolff, D. J., Datto, G. A. & Samatovicz, R. A. The dual mode peroxynitrite. J. Biol. Chem. 273, 22136–22144 (1998).
Rev. Mol. Cell Biol. 3, 214–220 (2002). of inhibition of calmodulin-dependent nitric oxide synthase 95. Goodwin, D. C. et al. Nitric oxide trapping of tyrosyl radicals
50. Lala, P. K. & Chakraborty, C. Role of nitric oxide in by antifungal imidazole agents. J. Biol. Chem. 268, generated during prostaglandin endoperoxide synthase
carcinogenesis and tumour progression. Lancet Oncol. 9430–9436 (1993). turnover. Detection of the radical derivative of tyrosine 385.
2, 149–156 (2001). 74. Bogle, R. G. & Vallance, P. Functional effects of econazole J. Biol. Chem. 273, 8903–8909 (1998).
51. Huang, P. L. Mouse models of nitric oxide synthase on inducible nitric oxide synthase: production of a 96. Huang, P. L. et al. Hypertension in mice lacking the gene for
deficiency. J. Am. Soc. Nephrol. 11, S120–S123 (2000). calmodulin-dependent enzyme. Br. J. Pharmacol. 117, endothelial nitric oxide synthase. Nature 377, 239–242
52. Dawson, V. L. & Dawson, T. M. Nitric oxide in 1053–1058 (1996). (1995).
neurodegeneration. Prog. Brain Res. 118, 215–229 (1998). 75. Matter, H. et al. Structural requirements for inhibition of the 97. Lefer, D. J. et al. Leukocyte–endothelial cell interactions in
53. Dawn, B. & Bolli, R. Role of nitric oxide in myocardial neuronal nitric oxide synthase (NOS-1): 3D-QSAR analysis nitric oxide synthase-deficient mice. Am. J. Physiol. 276,
preconditioning. Ann. NY Acad. Sci. 962, 18–41 (2002). of 4-oxo and 4-amino-pteridine-based inhibitors. J. Med. 1943–1950 (1999).
54. Kim, P. K., Kwon, Y. G., Chung, H. T. & Kim, Y. M. Ann. NY Chem. 45, 2923–2941 (2002). 98. Gyurko, R., Kuhlencordt, P., Fishman, M. C. & Huang, P. L.
Acad. Sci. 962, 42–52 (2002). 76. Jaffrey, S. R. & Snyder, S. H. PIN: an associated protein Modulation of mouse cardiac function in vivo by eNOS and
55. Kubes, P. Inducible nitric oxide synthase: a little bit of good inhibitor of neuronal nitric oxide synthase. Science 274, ANP. Am. J. Physiol. 278, H971–H981 (2000).
in all of us. Gut 47, 6–9 (2000). 774–777 (1996). 99. Huang, Z. et al. Enlarged infarcts in endothelial nitric oxide
56. Corbett, J. A. & McDaniel, M. L. The use of aminoguanidine, 77. Huang, H., Martasek, P., Roman, L. J. & Silverman, R. B. synthase knockout mice are attenuated by nitro-L-arginine.
a selective iNOS inhibitor, to evaluate the role of nitric oxide Synthesis and evaluation of peptidomimetics as selective J. Cereb. Blood Flow Metab. 16, 981–987 (1996).
in the development of autoimmune diabetes. Methods 10, inhibitors and active site probes of nitric oxide synthases. 100. Lo, E. H. et al. Temporal correlation mapping analysis of the
21–30 (1996). J. Med. Chem. 43, 2938–2945 (2000). hemodynamic penumbra in mutant mice deficient in
57. Stamler, J. S., Loh, E., Roddy, M. A., Currie, K. E. & 78. O’Neill, M. J. et al. ARL 17477, a selective nitric oxide endothelial nitric oxide synthase gene expression. Stroke 27,
Creager, M. A. Nitric oxide regulates basal systemic and synthase inhibitor, with neuroprotective effects in animal 1381–1385 (1996).
pulmonary vascular resistance in healthy humans. models of global and focal cerebral ischaemia. Brain Res. 101. Kano, T., Shimizu-Sasamata, M., Huang, P. L.,
Circulation 89, 2035–2040 (1994). 871, 234–244 (2000). Moskowitz, M. A. & Lo, E. H. Effects of nitric oxide synthase
58. Petros, A., Bennett, D. & Vallance, P. Effects of nitric oxide 79. Babu, B. R. & Griffith, O. W. N5-(1-imino-3-butenyl)-L- gene knockout on neurotransmitter release in vivo.
synthase inhibitors on hypotension in patients with septic ornithine, a neuronal isoform selective mechanism-based Neuroscience 86, 695–699 (1998).
shock. Lancet 338, 1557–1558 (1991). inactivator of nitric oxide synthase. J. Biol. Chem. 273, 102. Panahian, N. et al. Attenuated hippocampal damage after
The first administration of an NOS inhibitor to 8882–8889 (1998). global cerebral ischemia in mice mutant in neuronal nitric
patients. This paper shows that the haemodynamic 80. Radomski, M. W., Palmer, R. M. & Moncada, S. oxide synthase. Neuroscience 72, 343–354 (1996).
effects of septic shock can be reversed by Glucocorticoids inhibit the expression of an inducible, but 103. Kakuyama, M., Ahluwalia, A., Rodrigo, J. & Vallance, P.
blocking NOS. not the constitutive, nitric oxide synthase in vascular Cholinergic contraction is altered in nNOS knockouts:
59. Petros, A. et al. Effects of a nitric oxide synthase inhibitor in endothelial cells. Proc. Natl Acad. Sci. USA 87, co-operative modulation of neural bronchoconstriction by
humans with septic shock. Cardiovasc. Res. 28, 34–39 10043–10047 (1990). nNOS and COX. Am. J. Respir. Crit. Care Med. 160,
(1994). 81. Vallance, P., Leone, A., Calver, A., Collier, J. & Moncada, S. 2072–2078 (1999).
60. Ashina, M., Lassen, L. H., Bendtsen, L., Jensen, R. & Accumulation of an endogenous inhibitor of nitric oxide 104. Huang, P. L., Dawson, T. M., Bredt, D. S., Snyder, S. H. &
Olesen, J. Effect of inhibition of nitric oxide synthase on synthesis in chronic renal failure. Lancet 339, 572–575 Fishman, M. C. Targeted disruption of the neuronal nitric
chronic tension-type headache: a randomised crossover (1992). oxide synthase gene. Cell 75, 1273–1286 (1993).
trial. Lancet 353, 287–289 (1999). 82. Leiper, J. & Vallance, P. Biological significance of The original paper that describes the striking
61. Warren, J. B. Nitric oxide and human skin blood flow endogenous methylarginines that inhibit nitric oxide gastrointestinal phenotype of nNos knockout mice.
responses to acetylcholine and ultraviolet light. FASEB J. synthases. Cardiovasc. Res. 43, 542–548 (1999). 105. Mashimoto, H., He, X. D., Huang, P. L., Fishman, M. C. &
8, 247–251 (1994). 83. Ogawa, T., Kimoto, M. & Sasaoka, K. Purification and Goyal, R. K. Neuronal constitutive nitric oxide synthase is
62. Rees, D. D., Palmer, R. M., Schulz, R., Hodson, H. F. & properties of a new enzyme, NG,NG-dimethylarginine involved in murine enteric inhibitory neurotransmission.
Moncada, S. Characterization of three inhibitors of dimethylaminohydrolase, from rat kidney. J. Biol. Chem. J. Clin. Invest. 98, 8–13 (1996).
endothelial nitric oxide synthase in vitro and in vivo. Br. J. 264, 10205–10209 (1989). 106. MacMicking, J. D. et al. Altered responses to bacterial
Pharmacol. 101, 746–752 (1990). 84. Leiper, J. M. et al. Identification of two human infection and endotoxic shock in mice lacking inducible nitric
63. Garvey, E. P. et al. Potent and selective inhibition of dimethylarginine dimethylaminohydrolases with distinct oxide synthase. Cell 81, 541–650 (1995).
human nitric oxide synthases. Inhibition of non-amino tissue distributions and homology with microbial arginine 107. Wei, X. Q. et al. Altered immune responses in mice lacking
acid isothioureas. J. Biol. Chem. 269, 26669–26676 deiminases. Biochem. J. 343, 209–214 (1999) inducible nitric oxide synthase. Nature 375, 408–411 (1995).
(1994). 85. MacAllister, R. J. et al. Regulation of nitric oxide synthesis by References 106 and 107 describe the effects of loss of
64. Raman, C. S. et al. Implications for isoform-selective dimethylarginine dimethylaminohydrolase. Br. J. Pharmacol. iNOS. There are some interesting differences between
inhibitor design derived from the binding mode of bulky 119, 1533–1540 (1996). the phenotypes described.
isothioureas to the heme domain of endothelial nitric 86. Nakagomi, S., Kiryu-Seo, S., Kimoto, M., Emson, P. C. & 108. Gross, S. S., Kilbourn, R. G. & Griffith, O. W. NO in septic
oxide synthase. J. Biol. Chem. 276, 26486–26491 Kiyama, H. Dimethylarginine dimethylaminohydrolase shock: good, bad or ugly? Learning from iNOS knockouts.
(2001). (DDAH) as a nerve-injury-associated molecule: mRNA Trends Microbiol. 4, 47–49 (1996).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 1 | DECEMBER 2002 | 9 4 9


© 2002 Nature Publishing Group
REVIEWS

109. Scott, D. J. et al. Lack of inducible nitric oxide synthase 116. Perner, A. et al. Expression of nitric oxide synthases and Acknowledgements
promotes intestinal tumorigenesis in the Apc(Min/+) mouse. effects of L-arginine and L-NMMA on nitric oxide production Work in the laboratory of P. V. is generously supported by grants
Gastroenterology 121, 889–899 (2001). and fluid transport in collagenous colitis. Gut 49, 387–394 from the British Heart Foundation, the Wellcome Trust and the
110. Yamasaki, K. et al. Reversal of impaired wound repair (2001). Medical Research Council. The authors are grateful to R. Knowles,
in iNOS-deficient mice by topical adenoviral-mediated 117. Liu, J. S., Zhao, M. L., Brosnan, C. F. & Lee, S. C. N. Boughton-Smith, and C. S. Raman for information provided,
iNOS gene transfer. J. Clin. Invest. 101, 967–971 Expression of inducible nitric oxide synthase and and to A. Hobbs for critical reading of the manuscript. Figure 3 was
(1998). nitrotyrosine in multiple sclerosis lesions. Am. J. Pathol. prepared by S. Rossiter. From the work of the authors, University
111. Heneka, M. T. et al. Neuronal and glial coexpression of 158, 2057–2066 (2001). College London (UCL) holds patents in relation to DDAH as a target
argininosuccinate synthetase and inducible nitric oxide 118. Vuolteenaho, K., Moilanen, T., Al-Saffar, N., Knowles, R. G. for drug action.
synthase in Alzheimer disease. J. Neuropathol. Exp. Neurol. & Moilanen, E. Regulation of the nitric oxide production
60, 906–916 (2001). resulting from the glucocorticoid-insensitive expression of
112. Easki, T. et al. Expression of inducible nitric oxide iNOS in human osteoarthritic cartilage. Osteoarthritis Online links
synthase and Fas/Fas ligand correlates with the Cartilage 9, 597–605 (2001).
incidence of apoptotic cell death in atheromatous 119. Borderie, D. et al. Nitric oxide synthase is expressed in DATABASES
plaques of human coronary arteries. Nitric Oxide 4, the lymphomononuclear cells of synovial fluid in patients The following terms in this article are linked online to:
561–571 (2000). with rheumatoid arthritis. J. Rheumatol. 26, 2083–2088 LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink/
113. Behr-Roussel, D., Rupin, A., Sansilvestri-Morel, P., (1999). aconitase | ApoE | arginase | arginine deiminases | arginine glycine
Fabiani, J. N. & Verbeuren, T. J. Histochemical evidence for 120. Knott, C., Stern, G. & Wilkin, G. P. Inflammatory regulators amidinotransferase | argininosuccinate synthase | calmodulin |
inducible nitric oxide synthase in advanced but non-ruptured in Parkinson’s disease: iNOS, lipocortin-1, and cyclooxy- caveolin-1 | DDAH | diamine oxidase | dystrophin | eNOS | eNos |
human atherosclerotic carotid arteries. Histochem. J. 32, genases-1 and-2. Mol. Cell. Neurosci. 16, 724–39 (2000). iNOS | iNos | nNOS | nNos | PIN | soluble guanylyl cyclase |
41–51 (2000). 121. Annane, D. et al. Compartmentalised inducible nitric-oxide thioredoxin | TP53 | VEGF
114. Murray, I. A., Daniels, I., Coupland, K., Smith, J. A. & Long, synthase activity in septic shock. Lancet 355, 1143–1148 OMIM: http://www.ncbi.nlm.nih.gov/Omim/
R. G. Increased activity and expression of iNOS in human (2001). Alzheimer’s disease | Celiac disease | multiple sclerosis |
duodenal enterocytes from patients with celiac disease. 122. Forster, C., Clark, H. B., Ross, M. E. & Iadecola, C. Inducible Parkinson’s disease
Am. J. Physiol. Gastrointest. Liver. Physiol. 283, 19–26 nitric oxide synthase expression in human cerebral infarcts.
(2002). Acta. Neuropathol. 97, 215–220 (1999). FURTHER INFORMATION
115. Dijkstra, G. et al. Increased expression of inducible nitric 123. Suda, O. et al. Long-term treatment with Nω-nitro-L-arginine Encyclopedia of Life Sciences: http://www.els.net/
oxide synthase in circulating monocytes from patients with methyl ester (L-NAME) causes arteriosclerotic coronary nitric oxide: role in human disease | nitric oxide: synthesis and action
active inflammatory bowel disease. Scand. J. Gastroenterol. lesions in endothelial nitric oxide synthase-deficient mice. Protein Data Bank: http://www.rcsb.org/pdb/
37, 546–554 (2002). Circulation 106, 1729–1735 (2002). Access to this interactive links box is free online.

950 | DECEMBER 2002 | VOLUME 1 www.nature.com/reviews/drugdisc


© 2002 Nature Publishing Group

También podría gustarte