Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2015 Jan 12;172(6):1397–1414. doi: 10.1111/bph.12980

Extending the translational potential of targeting NO/cGMP-regulated pathways in the CVS

Andreas Papapetropoulos 1, Adrian J Hobbs 2, Stavros Topouzis 3,
PMCID: PMC4369253  PMID: 25302549

Abstract

The discovery of NO as both an endogenous signalling molecule and as a mediator of the cardiovascular effects of organic nitrates was acknowledged in 1998 by the Nobel Prize in Physiology/Medicine. The characterization of its downstream signalling, mediated through stimulation of soluble GC (sGC) and cGMP generation, initiated significant translational interest, but until recently this was almost exclusively embodied by the use of PDE5 inhibitors in erectile dysfunction. Since then, research progress in two areas has contributed to an impressive expansion of the therapeutic targeting of the NO-sGC-cGMP axis: first, an increased understanding of the molecular events operating within this complex pathway and second, a better insight into its dys-regulation and uncoupling in human disease. Already-approved PDE5 inhibitors and novel, first-in-class molecules, which up-regulate the activity of sGC independently of NO and/or of the enzyme's haem prosthetic group, are undergoing clinical evaluation to treat pulmonary hypertension and myocardial failure. These molecules, as well as combinations or second-generation compounds, are also being assessed in additional experimental disease models and in patients in a wide spectrum of novel indications, such as endotoxic shock, diabetic cardiomyopathy and Becker's muscular dystrophy. There is well-founded optimism that the modulation of the NO-sGC-cGMP pathway will sustain the development of an increasing number of successful clinical candidates for years to come.

Linked Articles

This article is part of a themed section on Pharmacology of the Gasotransmitters. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue–6

Tables of Links

TARGETS
GPCRsa Enzymesd
β2-adrenoceptor Arginase
Endothelin receptors COX
Ligand-gated ion channelsb Endothelial NOS (NOS3)
NMDA receptor Inducible NOS (NOS2)
Nuclear hormone receptorsc Neuronal NOS (NOS1)
Glucocorticoid receptor PDE family
PPAR-α PDE2
PPAR-γ PDE5
Soluble GC (sGC)
LIGANDS
Angiotensin II L-arginine
Aspirin LPS
Ataciguat (HMR1766) Methacholine
BAY41-2272 NADPH
BH4 Naproxen
cGMP Nitric oxide (NO)
Cinaciguat (BAY58-2667) Prednisolone
Flunisolide Prostacyclin
Glyceryl trinitrate Riociguat (BAY63-251)
GTP Sildenafil
Hsp90 Tadalafil
Isoprenaline TNF-α
Isosorbide mononitrate YC-1

These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,b,c,dAlexander et al., 2013a,b,c,d).

Introduction

The recent progress in the generation of additional, therapeutic molecules that target the NO transduction pathway is in large part due to a more detailed understanding of the biochemical and mechanistic complexities of the downstream pathways this molecule triggers. That is, the soluble GC (sGC)–cGMP axis. cGMP is a ubiquitous intracellular signalling molecule that affects a wide spectrum of cellular, and thus physiological, processes from cell growth and apoptosis to ion channel gating. Especially in the CVS in which it has been best studied, cGMP regulates many vital homeostatic mechanisms, including endothelial cell permeability, vascular smooth muscle contractility and cardiomyocyte hypertrophy (Francis et al., 2010). Of the two distinct GC systems that generate cGMP, this review exclusively focuses on the contribution of the NO-responsive arm to the detriment of the cGMP pool generated by natriuretic peptide hormones acting on membrane-bound, particulate forms of GC. Whereas there is considerable functional convergence of the two systems downstream, there is overwhelming evidence of spatial compartmentalization that results from the specific cellular co-localization of both the cGMP-generating systems as well as the cGMP-degrading PDEs, exemplified by the ability of PDE2 to selectively interfere with the natriuretic-stimulated cGMP pool, whereas PDE5 targets mainly the cytosolic cGMP pool, in cardiomyocytes (Castro et al., 2006; Piggott et al., 2006; Nausch et al., 2008; Tsai and Kass, 2009; Zhang and Kass, 2011).

This review will highlight the molecules and mechanisms within this pathway whose further study has recently generated successful entries in the medical arsenal, including use in some novel medical indications, thus showing great future promise in contributing to the treatment and elimination of human disease, especially disorders of the CVS.

Basic biology of the NO-sGC-cGMP pathway

Enzymatic generation of NO

Three isoforms of NOS exist, each one with a different pattern of expression (Alderton et al., 2001): neuronal NOS (nNOS or NOS-1), inducible NOS (iNOS or NOS-2) and endothelial NOS (eNOS or NOS-3). nNOS and eNOS are expressed constitutively whereas iNOS is not found in healthy cells but protein expression is induced following tissue injury or infection (Nathan, 1997). NOSs are capable of associating with the cell membrane, with cytosolic proteins or with the cytoskeleton, thus exhibiting dynamic subcellular localization (Oess et al., 2006). NOSs facilitate the five-electron oxidation of the terminal guanidino moiety of the semi-essential amino acid L-arginine, utilizing NADPH and BH4 as electron sources, to generate NO and L-citrulline in the presence of molecular oxygen (Alderton et al., 2001).

The regulation of NO bioavailability is complex and controlled by numerous mechanisms impacting directly NO levels, including NOS expression, substrate provision and chemical inactivation. For example, production of reactive oxygen species can inactivate NO (Münzel et al., 2005), and endogenous asymmetric methylarginines appear to act as NOS inhibitors (Leiper and Nandi, 2011; Caplin and Leiper, 2012). Arginase activity decreases the availability of the NOS substrate, L-arginine (Morris, 2009), uncouples NOS (resulting in generation of cytotoxic superoxide) and is thought to underlie nitrate tolerance (Khong et al., 2012). Modulation of eNOS–caveolin interactions (Garcia-Cardena et al., 1996) acts as an on/off switch for enzyme turnover and, more recently, interactions of NO with somatic haemoglobin (Straub et al., 2012) can reduce NO bioavailability. Furthermore, pharmacological enhancement of NO signalling can also be achieved indirectly. For example, stimulation of the β3-adrenoceptor in the heart has been shown to be coupled to the NO–cGMP pathway, to increase NO bioactivity and to prevent experimental maladaptive myocardial remodelling caused by isoprenaline or angiotensin II, an effect that deserves to be explored further clinically (Belge et al., 2014). Several molecules targeting the above mechanisms have been developed and evaluated preclinically (e.g. a NOS–caveolin disruptive peptide; Bucci et al., 2000); fewer have advanced in clinical trials. The latter include the arginase inhibitor N-hydroxy-nor-arginine, investigated in a phase I trial in coronary disease (Shemyakin et al., 2012; NCT02009527). However, no clinical approval of molecules targeting these mechanisms has yet validated these approaches.

cGMP biosynthesis in response to NO

The major biosensor of the generated NO is the enzyme sGC, which is found as an obligate heterodimer of α (α1 and α2) and β1 subunits; the α1β1 dimer seems to be the prevalent active form in most tissues with the exception of the nervous systems where equal amounts of α11 and α21 are detected. Each sGC subunit consists of (i) an N-terminal regulatory, haem-NO/oxygen (H-NOX) domain; (ii) a central Per-Arnt-Sim domain; (iii) a coiled-coil domain; and (iv) a C-terminal catalytic domain (Derbyshire and Marletta, 2012). There is one haem prosthetic group per heterodimer (Figure 1) that serves as the NO sensor and that is stimulated by nM concentrations of NO leading to an increase in enzymatic activity up to 400-fold (Kamisaki et al., 1986; Tsai and Kass, 2009). The α and β subunits have been proposed to be organized in a parallel fashion and the low basal activity of sGC is thought to result from the inhibitory action exerted by the binding of the catalytic domain to the regulatory domain; this inhibition is relieved upon NO binding. The presence of a reduced (Fe2+, ferrous) haem group is critical in NO sensing by sGC. For example, environmental cues, that increased the presence of reactive species such as superoxide (.O2) and peroxynitrite (ONOO) are translated into changes in the redox status of the haem group and therefore in the ability of sGC to respond to low concentrations of NO (Weber et al., 2001; Stasch and Hobbs, 2009; Figure 1). The implications of this in disease are crucial: it is thought that oxidative stress, a typical trigger for cardiovascular disease, can produce an NO-unresponsive (Fe3+, Ferric) sGC that is rapidly ubiquitinylated and degraded (Evgenov et al., 2006; Stasch and Hobbs, 2009). Furthermore, this sGC ‘uncoupling’ may result from S-nitrozation of vicinal thiols in the β1 subunit in addition to oxidation of the haem group (Stasch et al., 2006; Sayed et al., 2008). Such impairment of sGC activity in cardiovascular disease, coupled to concomitant decreases in NO bioavailability, has been the bedrock on which novel NO and/or haem-independent sGC stimulators and activators have been developed and which will be examined below (Evgenov et al., 2006; Follmann et al., 2013; Gheorghiade et al., 2013).

Figure 1.

Figure 1

Schematic representation of the major targetable components of the NO pathway. Disease-modifying NO can be generated from three main, well-studied sources: (i) cellular conversion from L-arginine; (ii) bacterial-based, enterosalivary bioconversion of food nitrates; and (iii) nitrate drugs such as glyceryl trinitrate, either spontaneously or through cellular conversion. The bioavailability of NO is regulated by its generation by the synthetic NOS enzymes and by the tissue complexation and conversion of NO, for example, to nitrosyl-free radicals. Initially, NO bioactivity is in major part determined by its best-described cellular ‘biosensor’: sGC coupled to reduced haem. sGC ‘stimulators’ such as riociguat, which was recently approved for treatment of two forms of PH, can by themselves activate sGC or synergize with NO. Chemical modification of sGC or oxidation of the haem prosthetic group and dissociation from sGC can occur in pathophysiological situations such as PH and heart ischaemia. Apo-sGC has an impaired ability to respond to NO, thus ‘uncoupling’ the NO pathway. This form of sGC can be ‘resuscitated’ by sGC ‘activators’ such as cinaciguat and ataciguat. PDEs are themselves regulated by and participate in the catabolism of cGMP. PDE5 inhibitors such as sildenafil and tadalafil are approved for erectile dysfunction and treatment of PH. NO pathway modifying drugs are increasingly evaluated in clinical trials in indications as varied as heart failure, traumatic cerebral oedema and forms of skeletal muscle dystrophies.

In addition to its upstream, direct effects on NO availability and sGC function, cellular oxidative stress may also interfere with the NO/cGMP pathway by inducing post-translational activation of the downstream cGMP effector PKG-Iα and thus affect adversely the progress of disease, something that has been experimentally shown to occur in sepsis (Rudyk et al., 2013). Due to this complex, and in some cases antithetical, regulation of NO bioactivity, in such pathological settings a dual-pronged therapeutic approach, that combines upstream restoration of physiological cGMP generation and pharmacological intervention (e.g. anti-oxidants) could be optimal to preserve the physiological function of downstream effectors.

It is important to note that the downstream biochemical pathway of NO is far from limited to cGMP-mediated effects: cGMP-independent changes are undeniably part of the NO signalling repertoire, including NO-triggered protein S-nitrozation (Lima et al., 2010) and effects on mitochondrial respiration and oxygen utilization (Erusalimsky and Moncada, 2007). One should keep in mind, however, that genetic inactivation of sGCβ1 (Friebe et al., 2007) and cGMP-dependent kinase I (PKG1) abolishes the hallmark physiological effect of NO, that is, vasorelaxation (Pfeifer et al., 1998), emphasizing the crucial involvement of cGMP in the effects of NO. It is also clear that the ‘canonical’ (cGMP dependent) NO pathway has provided the major impetus for translational progress and thus constitutes the central focus of the review.

Direct downstream signalling of cGMP

Two main enzyme families are directly regulated and respond to cGMP, to impact the pathophysiology of the CVS: cGMP-dependent PKs (PKGs) and PDEs (Figure 1). In addition, ion channel function is also directly or indirectly (e.g. via PKG-dependent pathways) regulated by cGMP levels, although this phenomenon is largely restricted to sensory transduction (Biel and Michalakis, 2007; Francis et al., 2010). To date, successful translational efforts have, however, focused primarily on the two upstream enzymatic targets: sGC and PDE. The ability of sGC to associate with the plasma membrane (Linder et al., 2005) and the possible compartmentalization of cGMP degrading PDEs (Castro et al., 2006; Nausch et al., 2008; Zhang and Kass, 2011) may further complicate the downstream functions of spatially regulated cGMP levels and the therapeutic targeting of enzymes that regulate its levels in distinct diseases.

The dominant PKG in the CVS is PKG type 1, which consists of two isoforms: α and β (Hofmann et al., 2006; Burley et al., 2007). The binding of cGMP to a regulatory region of the kinase results in a conformational change that ‘unrepresses’ the catalytic activity of the kinase and permits phosphorylation on Ser/Thr residues of client proteins. Pharmacological targeting of PKG is attractive but has not been successful up to now, because selective PKG activators and inhibitors are lacking. In addition, PKG inhibition may result in smooth muscle dysfunction, based on experimental evidence provided by mice with genetic deletion of cGMP kinase I (Pfeifer et al., 1998). Conversely, use of PKG activators to mimic the effects of sGC and pGC turnover is theoretically desirable in cardiovascular disease, but chronic use may be ultimately undesirable, given that gain-of-function genetic mutations in PKG found in humans are causally associated with aortic aneurysms and dissections (Guo et al., 2013).

The second cGMP-responsive system that has been well-studied comprises the PDE family of cyclic nucleotide-hydrolyzing enzymes, which have arguably been the most successful ‘cGMP-based’ therapeutic targets. Of the 11 PDE families (PDE1-11, each consisting of one to four isozymes and their multiple isoforms), PDEs-2, -3, -5, -6 and -11 are regulated by cGMP, of which PDE2, 3 and 5 are expressed in the constituent cells of the CVS, with PDE11 being found in the heart. PDEs exist as dimers, each monomer comprises a characteristic for the isotype N-terminal regulatory domain and a relatively high homology C-terminal catalytic domain that can undergo post-translational prenylation or phosphorylation (Conti and Beavo, 2007; Keravis and Lugnier, 2012). Whereas PDE2 and PDE5 are activated by cGMP binding to their GAF regulatory domain, PDE3 is inhibited by competitive binding of cGMP to its catalytic site. Of these three PDEs, PDE2 and PDE3 can hydrolyse both cGMP and cAMP, while PDE5 is selective for cGMP (Bender and Beavo, 2006; Conti and Beavo, 2007). PDE5, which is highly expressed in the corpus cavernosum and in the lung, is the target of small-molecule inhibitors that have been approved to treat erectile dysfunction and pulmonary arterial hypertension [PAH; World Health Organization (WHO) group I] (Rosen and Kostis, 2003; Croom et al., 2008). Additional preclinical data support a role for PDEs 1, 2, 3 and 10 in pulmonary hypertension, with proof-of-concept studies in cells and tissues from patients with the disease, implying that pharmacological blockade of other PDE isoforms might be beneficial (Phillips et al., 2005; Schermuly et al., 2007; Tian et al., 2011; Bubb et al., 2014). Further consideration of the therapeutic potential of PDE inhibitors, particularly PDE5, is discussed next.

New lead molecules targeting the NO-sGC-cGMP pathway

Innovation in targeting the NO-sGC-cGMP pathway derives from either (i) development of new molecular entities; or (ii) extended clinical applications of already-approved therapeutic molecules. Research that has been conducted in the past 10–15 years has produced novel lead therapeutic molecules that have entered clinical evaluation and, on occasion, are now approved medicines.

Two main categories of novel chemical entities in the early or late clinical arena that target the NO-sGC-cGMP axis are briefly explored below. First, there are established drugs that have been coupled to an NO-donating group to alleviate undesirable side effects of the ‘parent’ molecule. However, far more innovative is the second category, which includes sGC ‘stimulators’ and ‘activators’ and therefore this review will draw attention to their preclinical pharmacology and mode of action.

NO-donating anti-inflammatory drugs

The most clinically advanced, major drug group that has been used as NO-donating, ‘carrier’ scaffold has been the steroidal and non-steroidal anti-inflammatory drugs (NSAIDs), including aspirin. These hybrid molecules are being tested in a wide array of indications, from colon cancer prophylaxis to reduction of vascular complications due to hypercholesterolaemia, not all of which can be thoroughly covered by this review.

The molecules that are perhaps closest to approval are NSAID conjugates whose therapeutic benefit relies (i) on the presumed gastroprotection that released NO would provide to the NSAID moiety, given the increased possibility of ulcer development (del Soldato et al., 1999; Wolfe et al., 1999; Bandarage and Janero, 2001); and (ii) on the counterbalancing of the modest, but significant, effect on blood pressure that certain NSAIDs can cause in some patient populations and that can limit the health benefit of the anti-inflammatory drug (White et al., 2011). NSAIDs are among the most prescribed drugs in the world; however, it is now well established that their use carries the risk of upper gastrointestinal damage, including life-threatening bleeding complications, as side effects of their mode of action. The risk varies with the NSAID used and is especially frequent in certain populations prone to bleeding (Chan et al., 2007). There are approved pharmacological strategies to prophylactically reduce the risk of gastrointestinal events due to NSAID intake, including, for example, co-administration of proton pump inhibitors (Chan et al., 2007; Graham and Chan, 2008).

There is now ample experimental evidence from preclinical models that NO-releasing forms of approved steroidal and NSAIDs, including COX inhibitors such as aspirin and glucocorticoids such as prednisolone and flunisolide, exhibit similar or increased efficacy and a more favourable side effect profile than the parent molecules in several preclinical disease settings (Fiorucci et al., 2002; Paul-Clark et al., 2003; Turesin et al., 2003; Wallace et al., 2004). Such anti-inflammatory drug NO conjugates have been experimentally shown to modulate ovarian (Bratasz et al., 2008) skin (Chaudhary et al., 2013) or intestinal (Williams et al., 2004) solid tumour growth, exert anti-inflammatory activity with reduced symptoms of gastric damage properties (Wallace et al., 2004; Fiorucci et al., 2007) and protect against or accelerate improvement of experimental colitis (Fiorucci et al., 2002; Zwolinska-Wcislo et al., 2011). The increased anti-inflammatory efficacy of at least one of them, the prednisolone derivative NCX-1015, may in part be attributed to glucocorticoid receptor nitration resulting in more robust signalling (Paul-Clark et al., 2003).

A number of NO-conjugated COX inhibitors have also been evaluated in clinical trials. For example, NCX4016 (an aspirin-NO conjugate) has completed clinical testing in preventing colorectal cancer in patients at high risk for developing this disease (ClinicalTrials.gov identifier: NCT00331786) and in improving walking distance in patients with peripheral arterial occlusive disease (NCT01256775); however, no published report of trial outcomes is available at the writing of this review. Another 13 week clinical trial involves a naproxen–NO conjugate (naproxcinod) that is intended to treat ‘hypertensive’ patients (mean arterial pressure >125 mmHg) with osteoarthritis. In these individuals, naproxen induces a small rise (3–8 mmHg) in systolic BP, which increases significantly the risk of cardiac complications in this population. Naproxcinod exhibits a much lower tendency to increase systolic BP than naproxen, sparing the need for anti-hypertensive drugs taken concomitantly by this population (White et al., 2011). However, the FDA has withheld approval until longer term effects of the drug are presented. In sum, none of these molecules has yet progressed to large-scale clinical evaluation, while, for the moment, the clinical use of NO-donating NSAIDs awaits convincing clinical data that for approval (Fiorucci and Distrutti, 2011).

sCG stimulators

Pharmacology and mode of action

Given that reduced NO production is a defining feature of many cardiovascular diseases, including PH, the use of PDE inhibitors is likely to be limited as the efficacy of such molecules is dependent on endogenous cGMP generation. Thus, compounds that activate sGC directly, or that synergize with NO in activating the enzyme, appear a perfect fit as drug candidates in such indications. The initial discovery, by Taiwanese researchers in the mid-1990s, of the first ‘sGC stimulator’, YC-1 (Wu et al., 1995), was paralleled by a wide search performed by a variety of pharmaceutical companies for molecules that could act in dual fashion: they synergize with NO in stimulating sGC and directly stimulate the enzyme in the absence of NO. Both activities are, however, dependent on the presence of a reduced, sGC-bound haem moiety (Hoenicka et al., 1999).

The mechanistic basis of sGC stimulation by these molecules has been extensively studied, but not conclusively elucidated (Follmann et al., 2013), mainly because there are no X-ray data of the full-length crystallized enzyme. Raman spectroscopic studies with sGC stimulators and structural modelling studies (based on the somewhat tenuous similarity to the AC catalytic domain) suggest that molecules such as YC-1 and BAY 41-2272 (i) induce a (indirect) change in the prosthetic haem group geometry that has bound NO, making the enzyme more active and stabilizing the nitrosyl–haem complex; and (ii) photoaffinity labelling of BAY-41-2272 and YC-1 analogues results in labelling of the α-subunit, following binding of the compound to a domain distinct from the catalytic site. However, it is not absolutely clear that the binding itself occurs on the α-subunit. It is possible that the site of binding is in the interface between the sGC subunits and thus elicits an allosteric interaction that results in a more active conformational shift of the enzyme and in the labelling of the α-subunit (reviewed in Derbyshire and Marletta, 2012; Follmann et al., 2013). Alternatively, sGC stimulators have been suggested to relieve an autoinhibitory interaction between the H-NOX domain in the N-terminus, which harbours the haem moiety and the C-terminus catalytic domain (Winger and Marletta, 2005). In a recently published study, Purohit et al. (2014) demonstrated that YC-1 binding to the β1 sGC subunit overcomes the allosteric inhibition by the α1 subunit. In all, the exact binding site of the sGC stimulators has not been assigned with certainty yet, and more structural studies have to be performed to finally understand how sGC stimulators bind to the protein.

Of the many molecules of the sGC stimulator class that have been developed, riociguat (BAY 63-2521) is the one that finished first in the translational race that led to its approval in the past year in the United States, Canada and in the European Union for the treatment of two forms of PH (Conole and Scott, 2013). Many sGC stimulator molecules, including YC-1, were abandoned because of lack of selectivity (YC-1 also inhibited PDEs) and poor pharmacokinetic characteristics (Stasch and Hobbs, 2009). One instructive reason for riociguat's success may be that very early, before full preclinical evaluation, all fellow candidate molecules were evaluated and discarded if they possessed a poor pharmacokinetic profile (Follmann et al., 2013), allowing research to concentrate on candidates that were potent, selective and possessed a favourable bioavailability/pharmacokinetic profile. At the outset, riociguat showed good bioavailability and lack of interaction with the CYP metabolizing system, thus presenting the considerable advantage of future co-administration with other drugs (Follmann et al., 2013). In vitro characterization of the drug showed strong synergy in combination with NO, ability to induce sGC activity in the absence of NO and dependence on a reduced haem prosthetic group. The preclinical evaluation of riociguat in key experimental animal models in vivo displayed, crucially, a long-preserved (several weeks) hypotensive effect in rats made tolerant to organic nitrates, effective inhibition or reversal of pulmonary vasoconstriction and remodelling (muscularization of small pulmonary arteries, hypertrophy of the right ventricle) in the monocrotaline model of PH (Schermuly et al., 2008; Stasch et al., 2011; Lang et al., 2012), and reduction of heart and kidney fibrosis in the Dahl hypertensive rat, resulting in increased survival rates over time (Geschka et al., 2011).

Clinical success of the sGC stimulator, riociguat

There are two clinical areas where considerable progress has been made in the last few years with the sGC stimulators: PH and heart failure, with pulmonary hypertension being the most successfully targeted clinical indication, based on riociguat's approval.

PH is a progressive, debilitating, multifactorial disease and exacts a high socio-economic toll. Most of the approved current treatments target one subgroup: PAH, a life-threatening form of the disease that is characterized by increased pulmonary vascular resistance, excessive remodelling of small vessels and of the pulmonary artery that lead, over time, to right heart failure and death (Baliga et al., 2011; Galiè et al., 2011; Schermuly et al., 2011). Available treatments for PAH include endothelin receptor antagonists, PDE inhibitors, prostacyclin analogues and Ca2+ channel blockers (Baliga et al., 2011; Galiè et al., 2011). The necessity of additional supportive drug therapy to treat concurrent pathophysiologies, which includes oral anticoagulants, digoxin for arrhythmias and diuretics to regulate fluid accumulation and blood pressure (reviewed by Galiè et al., 2011) increases the risk of undesirable drug–drug interactions, especially with the anticoagulants. Approval of any new pharmacological options that are well-tolerated and display minimal drug–drug interactions would be a welcome addition to this therapeutic arsenal.

Among other PH forms, persistent PH of the neonate can be effectively treated with administration of inhaled NO (Roberts et al., 1992; Vosatka et al., 1994), but NO donors are not clinically useful for chronic treatment of PH because of partial patient response, development of severe tolerance over time, short-lived duration of the pulmonary vasodilation and the danger of methaemoglobinaemia with high NO doses (Ichinose et al., 2004; Galiè et al., 2011).

The exact molecular ‘defect’ in the NO-sGC-cGMP axis that may contribute to the development of the various forms of pulmonary hypertension in adults remains debatable and experimental and clinical data seem often contradictory (Giaid and Saleh, 1995; le Cras et al., 1996; Xu et al., 2004). Pharmacological potentiation of the NO pathway (Rossaint et al., 1993; Klinger, 2007; Vermeersch et al., 2007; Geschka et al., 2011) has been the basis for the development of small-molecule inhibitors of PDE5A such as sildenafil and tadalafil, which were introduced in this clinical area in the past decade (Galiè et al., 2009; 2011; Stasch and Hobbs, 2009). The issue, particularly in PAH, is reduced NO bioavailability: PAH is considered an NO-deficient state (Stasch and Evgenov, 2013). Because sGC expression is maintained or even up-regulated in PH, targeting it with a sGC stimulator (which can synergize with NO) seems a particularly beneficial approach.

Clinical trials with the sGC stimulator, riociguat, in two forms of PH were successfully concluded in 2013: the treatment met primary end points in patients diagnosed with PAH and with chronic thromboembolic pulmonary hypertension (CTEPH or WHO group IV). In the phase III trial (PATENT 1 ClinicalTrials.gov) in PAH patients who received riociguat alone or in combination with approved endothelin receptor antagonists or prostanoids for 12 weeks, the 6 min walk distance (6-MWD) increased by 36 m compared with placebo. In addition, there was significant improvement in pulmonary vascular resistance, cardiac output, N-terminal pro-B-type natriuretic peptide (NT-proBNP) plasma levels, time to clinical worsening, WHO functional class, Borg dyspnoea score and quality-of-life assessment. In addition, the benefit was also manifest at 24 weeks (Ghofrani et al., 2013b). The second, 16 week phase III trial (CHEST-1) included patients diagnosed with CTEPH who were either inoperable or showed persistent or recurrent PH despite having undergone pulmonary endarterectomy, a standard surgical option for this group for which no pharmacological options exist. Riociguat increased the 6-MWD by 46 m compared with placebo and produced significant improvement in pulmonary vascular resistance, cardiac output, N-terminal pro-B-type natriuretic peptide level and WHO functional class (Ghofrani et al., 2013a). In both trials, the safety profile of the sGC stimulator was reassuring, a major plus that warrants further evaluation of the molecule in additional indications.

In addition to the above indication, riociguat is also being tested clinically, and has shown beneficial effects, in proof of concept, pilot or phase II studies in patients with PH secondary to interstitial lung disease and chronic obstructive pulmonary disease (Bonderman et al., 2013; Hoeper et al., 2013; Stasch and Evgenov, 2013). The first report of a phase IIb trial in patients with pulmonary hypertension caused by systolic left ventricular dysfunction, an indication with no approved medication, shows that treatment with riociguat did not meet the primary end point, which was the decrease in mean pulmonary artery pressure at 16 weeks (Bonderman et al., 2013); however, it improved the secondary outcomes cardiac index and systemic and pulmonary resistance. Despite an attempt to decipher possible effects in patient populations after stratification, the study was not powered or designed to answer some critical questions, for example, whether riociguat elicited pulmonary vasodilation (inferred by the calculated drop in pulmonary vascular resistance) or whether variation of the drug dose and duration of treatment in specific patient subpopulations would successfully reach the primary end point. The mitigated results may leave the door open for a more prolonged trial, where long-term ventricular function is monitored and where, given riociguat's safety profile, higher doses are tested. Riociguat is also in early clinical stage evaluation for improvement of flow to the digits in Raynaud's syndrome patients (NCT01926847).

sGC activators

Preclinical pharmacology of sGC activators

Additional screening of a compound library following the discovery of sGC stimulators at Bayer and further examination of hits revealed that a second series of dicarboxylic acids could up-regulate sGC activity in an NO-independent and haem-independent manner, thus inaugurating a quite different molecular class, termed sGC activators. More companies also arrived at similar-acting molecules (Schindler et al., 2006; Costell et al., 2012; Follmann et al., 2013). Most of the second-generation molecules contain only one monocarboxylic acid moiety. An example of an activator that lacks carboxylic acid moieties also exists (HMR176). The mechanistic basis for the mode of action of sGC activators is arguably better understood than that of sGC stimulators. Data from functional, mutational and spectroscopic studies indicate that sGC activators bind in the haem cavity within the H-NOX domain of the β1 subunit, competing with the native ligand (Pellicena et al., 2004; Martin et al., 2010; Follmann et al., 2013). The His105 in the β1 H-NOX domain, which serves as a fifth coordination for the haem iron and is crucial for sGC activation, is displaced from the ‘inactive’ form, causing the rotation of the helix that harbours His105 to a degree that depends on the sGC activator used (Follmann et al., 2013). In this way, this class of compounds activate sGC in the absence of a haem moiety (Pellicena et al., 2004; Follmann et al., 2013). Of the sGC activators, the molecular mechanism of action of BAY 58-2667 (cinaciguat) has been characterized in most detail (Martin et al., 2010). The carboxylic groups of BAY 58-2667 displace the haem propionic acids and interact with Tyr135 and Arg139 of the β1 subunit and sGC activation results from a signal transmission triad composed of His105, Tyr135 and Arg139 (Schmidt et al., 2004).

Cinaciguat, and possibly other sGC activators, can prevent the degradation of sGC subunits that occurs following haem oxidation, apo-sGC formation and subunit ubiquitination in disease conditions. The ability of cinaciguat to closely mimic haem binding rescues sGC from proteasomal degradation by stabilizing the apo-sGC structure and thus possesses a dual mechanism of action (maintenance of sGC levels and sGC activation) in diseases associated with increased oxidative stress (Evgenov et al., 2006; Martin et al., 2010; Follmann et al., 2013).

A more conclusive assessment of the sGC haem redox state in whole cells and in tissues would help improve decision making on which diseases might benefit from the administration of sGC activators (Ahrens et al., 2011). There are two, recently described, methods that may allow this in different contexts in the future, provided that they are validated and confirmed by other laboratories. Fluorescence dequenching can be measured after the attachment of the biarsenical fluorophore FlAsH to the haem moiety (Hoffmann et al., 2011) via energy transfer from this fluorophore to the haem. However, this technique for now is limited to live cells in vitro and has yet to be extended to in vivo applications. In addition, a biochemical determination can be performed by assessing the degree of sGC-Hsp90 complexation: the binding of Hsp90 is limited to the haem-lacking enzyme and Hsp90 is dissociated once sGC has incorporated a haem prosthetic group (Ghosh and Stuehr, 2012). Similar methods, once established, could be very useful in better directing the therapeutic applicability of sGC activators.

This class of NO- and haem-independent sGC activators, therefore, raised the possibility of therapeutic use in situations where sGC is present in its haem-free form. Increased levels of apo-sGC (leading to its ubiquitination and proteasomal degradation) occur during oxidative stress, exemplified by either full-blown, acute inflammatory responses or chronic, low-level inflammation (Stasch et al., 2002; 2006). In these situations, the effect of PDE inhibitors or sGC stimulators is inherently limited (Evgenov et al., 2006) due to a lack of intact NO–sGC signalling. Thus, sGC activators have been extensively characterized in preclinical models of disease to determine if they offer a greater therapeutic potential. For example, drugs modifying the haem-oxidized or haem-free enzyme would target diseased tissue. This proved to be the case with encouraging results observed in models of myocardial infarction, hypertension or congestive heart failure (reviewed by Follmann et al., 2013). Cinaciguat, in a fast ventricular pacing model of congestive heart failure in dogs (Boerrigter et al., 2007), reduced mean arterial, right atrial, pulmonary artery and pulmonary capillary wedge pressure; increased cardiac output and renal blood flow; and preserved glomerular filtration rate and sodium and water excretion, making it a prime therapeutic candidate for cardiovascular indications where sGC is impaired because of oxidative stress. In addition, cinaciguat was shown to antagonize crucial pro-fibrotic mechanisms in vitro (Dunkern et al., 2007), thought to operate in many pathological remodelling processes in chronic cardiovascular diseases. GSK2181236A a sGC activator developed by GlaxoSmithKline, was tested in spontaneously hypertensive stroke-prone rats on a high salt/fat diet, demonstrating organ-protective effects and reducing left ventricular hypertrophy (Costell et al., 2012). Yet another sGC activator, HMR 1766 (ataciguat), was shown to improve ex vivo vascular function and reduce platelet activation (Schäfer et al., 2010). Ataciguat also prevents and reverses pulmonary vascular remodelling and right ventricular hypertrophy in a mouse model of PH (Weissmann et al., 2009). Collectively, these results warranted clinical evaluation in similar indications.

Clinical testing of sGC activators

HMR1766 (ataciguat) has been evaluated in two indications and trials have been completed: in the first, the primary end point was the reduction of pain in patients with neuropathic pain (NCT00799656) and in the second, the primary end point was improvement of intermittent claudication in patients with Fontaine stage II peripheral arterial disease (NCT00443287). The conclusions from these trials are still being awaited.

Cinaciguat has been tested in patients with acute decompensated heart failure, an indication where it seemed to be perfectly poised to succeed because of the strong evidence of NO pathway impairment in this disease and because of the experimentally based ability of the drug to limit fibrosis (reviewed by Tamargo and López-Sendón, 2011; Gheorghiade et al., 2013). Cinaciguat was delivered by i.v. administration at dose rates of 50–150 μg·h−1 and patients were monitored for up to 48 h. The trial, however, was terminated prematurely because of an increased occurrence of hypotension with all three doses (Gheorghiade et al., 2012; Erdmann et al., 2013a), which is an unfavourable occurrence in this patient population; in addition, there was no discernible effect of this treatment on either dyspnoea or on cardiac index and the small patient numbers did not allow stratification (Gheorghiade et al., 2012).

Although some of these clinical results have been disappointing, human genome-wide association studies have identified mutations in the genes encoding α1 (GUCY1A3) and β1 (GUCY1B3) subunits of sGC, and in the sGC-stabilizing protein CCTη, which increase the risk of hypertension, thrombosis and myocardial infarction (Ehret et al., 2011; Erdmann et al., 2013b). Thus, there is strong evidence for a direct involvement of sGC impairment in thromboembolic human disease and in the regulation of blood pressure. Individuals carrying such mutations may be prime candidates for treatment with sGC stimulators or activators, as they are likely to be disease modifying. However, the ethnic divergence in phenotype which is associated with GUCY SNPs suggests that patient stratification to sGC modulating drugs may be necessary.

NOS cofactor supplementation

One particular approach aiming to augment NO production is supplementation of the NOS cofactor tetrahydrobiopterin (BH4). Its bioavailability is reduced in a variety of cardiovascular pathologies, such as in atherosclerosis, at least in part as a result of overproduction of oxygen radicals, and correlates with NOS uncoupling (Förstermann and Li, 2011; Li and Förstermann, 2013). Pharmacological augmentation of BH4, therefore, aims to re-establish a healthy cofactor stoichiometry (Alkaitis and Crabtree, 2012; Starr et al., 2013) and direct eNOS catalytic activity towards producing NO rather than O2. To achieve just that, several clinical trials have been conducted or are in progress in disease conditions that include systolic or systemic hypertension and peripheral artery disease; however, for the moment, results from these trials either do not reveal statistically significant changes or are still not reported (Alkaitis and Crabtree, 2012; Cunnington et al., 2012). Characteristically, supplementation of BH4 in patients with coronary artery disease, although it produced increased levels of BH4 in saphenous vein (but not in internal mammary artery), resulted in the presence of the oxidation product BH2, which lacks NOS cofactor properties, and failed to either reduce superoxide levels or improve vascular function (Cunnington et al., 2012). These results demonstrate that, while supplementation of NOS cofactor(s) is based on a sound therapeutic rationale, the establishment of a favourable target BH4 : BH2 ratio is hard to achieve. Therefore, a fundamentally different approach targeting BH4 may be more useful, such as indirectly increasing its recycling and preservation. Indeed, in atherosclerotic patients, supplementation with 5-methyl-tetrahydrofolate (which prevents peroxynitrite-driven oxidation of BH4) has been shown to reduce peroxynitrite-mediated BH4 oxidation, to ameliorate the BH4/total biopterin ratio and to increase NOS coupling, thus preserving in vivo and ex vivo vascular endothelial function (Antoniades et al., 2006).

Repositioning of existing medicines and combination approaches

New molecular entities and modes of action have unquestionably boosted excitement in the NO field, and have advanced understanding of the physiology and pathology of sGC–cGMP signalling. However, significant translational progress has also been made with older, approved drugs. Quite a few of these have been, or are currently being, evaluated in indications that are either poorly served by available medications, or where an improvement of the currently obtainable therapeutic effect is desired.

One such example is the small (six patient), pilot clinical trial with a combination of the tried-and-tested organic nitrate, isosorbide mononitrate (ISMN), and the PDE5 inhibitor, sildenafil, in achieving better regulation of the blood pressure in patients afflicted with ‘resistant’ hypertension (Oliver et al., 2010). Monotherapy with either drug alone effectively reduced brachial systolic and diastolic blood pressure, and central systolic and diastolic arterial pressure. Combination of sildenafil and ISMN elicited significantly stronger reduction of brachial systolic blood pressure and central arterial systolic pressure, compared with either drug alone. Reduction of central arterial pressure with the combination reached a maximum of 26/18 mmHg (systolic blood pressure/diastolic blood pressure) compared with placebo (Oliver et al., 2010), thus opening the way for a study involving more patients and evaluation of longer administration of this combination in this challenging patient population.

Sildenafil also showed improvement in non-ischaemic, non-failing diabetic cardiomyopathy (i.e. at a relatively early stage) in a small, 3 month trial in 59 diabetic patients (NCT00692237), improving left ventricle contraction and preventing cardiac remodelling through, presumably, direct intramyocardial effects, independent of endothelial vasodilatation (Giannetta et al., 2012). Longer term results are expected in the next 48 months.

More impressively, in a 1 year prospective trial in 45 patients with stable, systolic heart failure, sildenafil, at 6 months and 1 year, improved left ventricle ejection fraction and elicited reverse remodelling of left atrial volume index and left ventricle mass index. These structural and functional ameliorations by sildenafil were coupled with improved exercise performance, ventilation efficiency and quality of life, thus making sildenafil the first PDE5 inhibitor that demonstrably elicits structural and functional changes in the human heart (Guazzi et al., 2011). A year later, the same group (Guazzi et al., 2012) reported that sildenafil succeeded, in a group of patients with heart failure that presented oscillatory breathing during exercise (attributed to pulmonary vasoconstriction), to almost eliminate (in ∼90% of the patients at 6 and 12 months) oscillatory breathing, a sign of poor prognosis for the progress of the disease, as well as to improve functional performance. These results were accompanied by reductions of pulmonary vascular resistance and pulmonary arterial pressure. Unfortunately, in the longer term follow-up RELAX study (Effectiveness of Sildenafil at Improving Health Outcomes and Exercise Ability in People With Diastolic Heart Failure), treatment of HFpEF patients with sildenafil failed to produce a significant change in exercise capacity, its primary outcome measure (Redfield et al., 2013), despite the positive outcome achieved in systolic heart failure patients (Guazzi et al., 2011).

Sildenafil was also tested in a 12 week clinical trial (NCT00517933) in patients with idiopathic pulmonary fibrosis (Zisman et al., 2010). Although the primary end point (increase in the 6 min walk distance by more than 20%) was not met, secondary symptomatic end points such as oxygenation, dyspnoea and quality of life score were improved by sildenafil (Zisman et al., 2010), raising the possibility of an expanded clinical investigation in the future.

Yet another approved PDE5 inhibitor, tadalafil, was the second molecule of its class to be approved for PAH in 2009 (Rosenzweig, 2010). Furthermore, in 2012, in a small pilot study, tadalafil proved effective in normalizing blood flow to the muscles of patients with Becker's muscular dystrophy (BMD). This genetic disease is linked to mutations in the gene encoding the skeletal muscle protein dystrophin, which induces defective sarcolemmal targeting of proteins, among which nNOSμ, and progressive muscle damage and wasting (Bushby et al., 2010a,b). There is no pharmacological treatment directed to this disease, which is associated with cardiomyopathy and results in loss of ambulation. The investigators tested a small patient group (and a matched cohort control, n = 10 each) for restoration of the exercise-induced attenuation of reflex sympathetic vasoconstriction. This is a physiological reflex that optimizes perfusion to the exercising muscles. This reflex was absent in 9/10 men carrying the disease and tellingly correlated with missing sarcolemmal nNOSμ. Tadalafil, given once, normalized this adaptive blood flow in response to sympathetic vasoconstriction in all participant patients (Martin et al., 2012) and can therefore benefit people with BMD by preventing muscle damage due to pathological vasoconstriction during exercise. In addition, in a promising preclinical study in a related indication sildenafil reversed cardiac dysfunction in the mdx mouse model of Duchenne muscular dystrophy (Adamo et al., 2010).

It can safely be said that the expectation, broadly shared by the research community, that PDE5 inhibitors would be clinically useful in treating heart failure (Zhang and Kass, 2011) or other diseases with a critical cardiac and/or vascular dysfunction (Kukreja et al., 2011) is slowly but steadily being fulfilled, despite the occasional hiccup. The clinical success and failures of PDE5 inhibitors reveal both the potential and the limitations of their therapeutic utility. More diversified trials may be expected to near completion in the next 2–3 years, firmly positioning PDE5 inhibitors in the therapeutic arena for years to come.

Thinking ‘outside the box’: re-examination of existing work and targeting novel therapeutic areas

Innovative rethinking of the role of the NO pathway in disease can open new opportunities, described briefly in the sections below. This is particularly true of the role of NO in sepsis, which points towards ‘a window of opportunity’ for sGC activators. In addition, dietary supplementation with inorganic nitrates offers an elegant example of how one can clinically improve cardiovascular disease by administering a simple, cheap and effective molecule. The therapeutic advantage of inhibition of the NO pathway has received relatively little attention, compared to efforts to increase NO activity; however, there are situations where this could provide therapeutic benefit. Lastly, the involvement of NO in energy expenditure is a topic with immense translational potential in atherometabolic diseases.

Time-sensitive apo-sGC stabilization in sepsis?

After the recent withdrawal of recombinant activated protein C from the market, there are no other specifically approved medications for sepsis, a largely (>50%) lethal indication (Ranieri et al., 2012). The hypothesis that boosting NO signalling may be of therapeutic interest in this life-threatening disorder is a novel concept, and directly opposite to the initial notion that iNOS inhibitors, which reduce the excessive NO production associated with systemic expression of this NOS isozyme in sepsis, would be the better approach (a thesis that failed to be substantiated in clinical evaluation; López et al., 2004). The anti-inflammatory properties of NO are well documented and augmenting NO signalling shows positive preliminary results in animal models of endotoxaemia (Da et al., 2007) and alleviates some symptoms in humans presented with adult respiratory distress syndrome (Taylor et al., 2004). Furthermore, nitrite generates NO selectively in hypoxic conditions (Lundberg et al., 2008) and can rescue mice subjected to LPS- or TNF-α-elicited shock (Cauwels et al., 2009), an effect mediated by cGMP produced by sGCα11 (Buys et al., 2009). However, initial experimental tests in endotoxin-exposed subjects that received inhaled NO have not yielded positive results (Hållström et al., 2008). A recent study in mice (Vandendriessche et al., 2013), though, has re-addressed this issue and has generated some very interesting observations, namely that a beneficial effect may critically depend on a combination of optimal timing and of apo-sGC stabilization. Mice that received an LPS injection were treated with sildenafil, the sGC stimulator BAY 41-2272 or the sGC activator cinaciguat, 3 or 8 h post-LPS challenge. Mortality was prevented only by cinaciguat, and only when it was given at the late, 8 h, time point after LPS. The effect of late treatment with cinaciguat correlated with stabilized body temperature and reduced cardiomyocyte apoptosis (Vandendriessche et al., 2013). This preclinical work demonstrates that ‘reactivation/preservation’ of apo-sGC is crucial in endotoxaemic shock and that the response critically depends on the time of treatment, when ‘rescued’ function of haem-free sGC is optimally amenable to impact the course of the disease. It is therefore of particular importance in future clinical trials in sepsis and systemic inflammatory response syndrome to correctly estimate this target window of apo-sGC responsiveness. It should be stressed that in sepsis, distinguishing between the effects of NO in the macrocirculation and in the microcirculation is important, and generation of NO selectively in the microcirculation may provide critical cytoprotective and tissue-protective effects. Indeed, treatment with nitrite, which is converted to NO selectively in hypoxic/acidic conditions, characteristic of the septic microvasculature, provides therapeutic benefit in preclinical murine models based on challenge by LPS or TNF-α, alleviating telltale symptoms of sepsis, such as organ damage and progressive hypothermia (Cauwels and Brouckaert, 2011).

Inorganic nitrite and nitrate

Although organic nitrates have been used for the treatment of angina and heart failure for more than 150 years, the physiological importance and pharmacodynamic properties of inorganic nitrite (NO2) and nitrate (NO3) have only recently been established (Lundberg et al., 2008; 2009,<). Initially considered to be simply inactive oxidation products of NO, it is now clear that these molecules can be reduced, preferentially under conditions of hypoxia and acidosis, to bioactive NO. This ‘non-canonical’ route of NO generation (Figure 1) is dependent on reduction of nitrate to nitrite by anaerobic bacteria that colonize the tongue, concentration of nitrite in the saliva, followed by absorption through the gut wall and entry into the systemic circulation (Lundberg et al., 2008; Kapil et al., 2010b). Production of NO from nitrite is then catalysed by ‘nitrite reductase’ enzymes, including xanthine oxidoreductase (Millar et al., 1998; Zhang et al., 1998; Webb et al., 2008a) and globins (Doyle et al., 1981; Basu et al., 2007; Tiso et al., 2011). In preclinical models, augmentation of this ‘nitrate-nitrite-NO’ pathway lowers systemic blood pressure, protects against ischaemia–reperfusion (I/R) injury and ameliorates pulmonary hypertension (Hunter et al., 2004; Webb et al., 2004; 2008b; Hendgen-Cotta et al., 2008; Casey et al., 2009; Zuckerbraun et al., 2010; Baliga et al., 2012). Such positive observations and the comparative ease of pharmacological and/or dietary manipulation of nitrite/nitrate levels has led to rapid translation of this phenomenon to healthy volunteers and patients with cardiovascular disease. Inorganic nitrite and nitrate have both been shown to lower systemic blood pressure in healthy volunteers (Cosby et al., 2003; Larsen et al., 2006; Webb et al., 2008b; Kapil et al., 2010a) and dietary nitrate supplementation reduces blood pressure in hypertensive patients (Ghosh et al., 2013) with a significantly increased potency, suggesting the beneficial effects of modulating nitrate-nitrite-NO signalling for therapeutic benefit are enhanced in disease. Further clinical evaluation has been conducted in patients presenting with acute myocardial infarction undergoing percutaneous coronary intervention. In a randomized, placebo-controlled, double-blind phase II evaluation, a 5 min i.v. administration of sodium nitrite prior to angioplasty did not reduce infarct size (the primary end point), although a subgroup of patients with diabetes did show some improvement (Siddiqi et al., 2013; NCT01388504 and ISRCTN57596739). This lack of efficacy is disappointing, given the preclinical observations, but may be dose related as the 70 μmol NaNO2 administrated was insufficient to significantly increase circulating NO2 concentrations, at least to levels shown to be required for blood pressure effects in healthy volunteers and hypertensive patients. Thus, further studies with higher doses of nitrite (and/or nitrate) and using different routes of administration (e.g. intracoronary) are warranted. Several further studies, primarily to assess the pharmacokinetic and safety profile of inorganic nitrite or nitrate, are also underway or nearing completion in patients with cardiovascular disease (e.g. cerebral vasospasm, sickle cell, peripheral arterial disease). Nitrite, at least in part via bioconversion to NO, can also provide tissue and organ protection following ischaemia (Rassaf et al., 2014), whether the experimental ischaemic insult is established in heart, kidney, brain or liver. In addition, nitrite also offers protection from experimental hypoxia-induced pulmonary hypertension (Rassaf et al., 2014). Based on these data, the beneficial effects of inhaled nitrite are currently being investigated in a phase I clinical trial, determining the changes in pulmonary vascular resistance in patients with pulmonary hypertension that undergo right heart catheterization (NCT01431313). In sum, raising plasma nitrite levels by pharmacological or dietary means represents a novel and inexpensive strategy to augment sGC–cGMP signalling for therapeutic gain.

Therapeutic potential of NOS inhibitors

High NO concentrations can compromise the blood–brain barrier and lead to brain oedema. The expression of iNOS and the levels of NO peak about 24–48 h after traumatic brain injury in humans (Clark et al., 1996). The improved pathology in mice subjected to cryogenic cerebral trauma that have been subjected to genetic (Jones et al., 2004) or pharmacological (Rinecker et al., 2003) ablation of iNOS indicates a deleterious role for NO in the recovery in this disease setting. VAS203 (6R,S)-4-amino-5,6,7,8-tetrahydro-L-biopterin) is an allosteric NOS inhibitor which, in a preclinical mouse model of intracranial oedema formation subsequent to cerebral trauma, showed improvements in short-term (24 h) oedema formation and in long-term functional preservation (Terpolilli et al., 2009). VAS203 is being tested in the clinic (NOSTRA: NO-Synthase inhibition in TRAumatic brain injury), in a European multicentre trial that is ongoing. Preliminary phase IIa results (according to a communiqué of the company) seem promising; however, the end of the trial has to be awaited to conclude on the efficacy of this molecule. Nonetheless, these findings are welcome because to date, iNOS inhibitors have failed to make the positive clinical impact predicted by animal models, particularly in the setting of sepsis.

NO production by NOS isoforms is regulated through protein–protein interactions. In particular, nNOS has been found to exist in a ternary complex with the synaptic scaffolding protein PSD95 and the NMDA receptor. Activation of this complex by glutamate following stroke and excessive NO production contributes to neuronal excitotoxicity and brain damage, making nNOS–PSD95 uncoupling a therapeutic approach to limit neurotoxicity (Cao et al., 2005). Tat-NR2B9c is a chimeric peptide that consists of the HIV-1 Tat protein transduction domain to facilitate cell penetration fused to a sequence that binds to the PDZ domains of PSD95 disrupting downstream neurotoxic signalling pathways, without blocking NMDA receptor activity. It was demonstrated that i.v. administration of Tat-NR2B9c 1 h after middle cerebral artery occlusion in non-human primates led to a reduction in infarct volume by 70% after 30 days. An improved, dimeric version of this peptide (NA-1) was generated and first tested in mice with favourable results (Bach et al., 2012). NA-1 was subsequently tested for its ability to improve the outcome of iatrogenic strokes occurring during aneurysm repair and assessed in a phase II trial (ENACT, NCT00728182). Although no differences in infarct volumes were observed between the saline and NA-1 groups, patients who received NA-1 exhibited significantly fewer new brain lesions than those receiving saline (Hill et al., 2012). This landmark study provides a proof of concept that neuroprotection is feasible in humans; however, the efficacy of NA-1 in community-onset stroke needs to be further established in more extended studies.

NO is also involved in nociceptive processing in the brain and contributes to cerebral artery vasodilatation, which is a symptomatic epiphenomenon of migraine (Hoffmann and Goadsby, 2012). iNOS seems to play a role in the pathogenesis of the disease, and for this reason iNOS inhibitors have also been in various stages of preclinical and clinical development to treat migraine, of which GW274150 is the most advanced. This molecule has been clinically tested both as a prophylactic treatment and as a treatment in acute migraine (NCT00242866 and NCT00319137). Results from both trials show that at doses that are predicted to inhibit iNOS by 80–90%, GW724150 was ineffective in reducing pain (Høye et al., 2009; Palmer et al., 2009; Høivik et al., 2010). Taken together, therefore, these data suggest that iNOS inhibition is unlikely to provide therapeutic relief in this indication.

iNOS inhibitors have also been, or are being, tested in additional indications. Elevated NO biosynthesis has been linked with increased angiogenesis, bone resorption and destruction of connective tissue in rheumatoid arthritis (Farrell et al., 1992; Stefanovic-Racic et al., 1993; Sakurai et al., 1995), manifestations that correlate with the pathogenesis and progress of the disease. GW274150 has been under clinical evaluation for use in this indication (NCT00370435 and NCT00379990). Final evaluation of 28 day treatment with GW274150 in reducing synovial thickness and vascularity in a rheumatoid arthritis in an early phase trial showed only a non-statistically significant trend (Seymour et al., 2012). The results of additional concurrent trials are being awaited. Last, GW274150 has also been tested in the treatment of mild asthma (NCT00273013). The conclusions of the study were negative, as GW274150 did not inhibit early or late asthmatic challenges to allergen or to methacholine-induced responses (Singh et al., 2007).

These mixed clinical results suggest that it may be important in the future to focus testing selective NOS inhibitors in a subset of carefully chosen clinical indications.

Regulation of fat phenotype and energy expenditure by NO

Our understanding of the molecular mechanisms that determine adipose tissue phenotype and of the respective pathophysiological roles of white and brown fat has made impressive progress lately (Bartelt and Heeren, 2014). Hence, ways to pharmacologically control and modulate fat phenotype can have a potentially enormous impact on various pathologies, including atherometabolic diseases. Pharmacological inhibition of NO activity in vitro or eNOS genetic inactivation in vivo results in decreased mitochondrial biogenesis, which is ascribed to altered cGMP generation; these interventions also interfere with non-shivering thermogenesis by brown fat and with energy expenditure (Nisoli et al., 2003). Conversely, eNOS transgenic mice (overexpressing eNOS under the pre-proendothelin promoter) on high fat diet display increased systemic metabolic rate (not attributed to hyperthyroidism) and adipose cell hypertrophy, while their adipose tissue shows signs of ‘browning’, with higher mitochondrial activity and elevated PPAR-α and PPAR-γ expression (Sansbury et al., 2012). In addition to NO-dependent pathways, natriuretic peptide signalling can also trigger a brown fat thermogenic programme in white adipocytes (Bordicchia et al., 2012). Collectively, these data clearly show anti-obesity effects of cGMP-mediated signalling and raise the possibility that increased NO bioactivity may help control some crucial features of the metabolic syndrome. Importantly, in the study by Sansbury et al., eNOS overexpression did not affect blood glucose handling. These exciting results point to a novel biochemical pathway that can be effectively targeted, even with currently available medications, to control clinical features of metabolic disorder associated with obesity.

Summary

A promising future for molecules targeting the NO-sGC-cGMP pathway in cardiovascular diseases

The collective research effort to better understand the biochemical and mechanistic complexity of the NO-sGC-cGMP pathway, combined with the progress in elucidating its regulation and involvement in pathophysiology (Figure 1), have successfully guided the translational development of medicines to address important human therapeutic needs. The extraordinary robustness of the field is mainly due to three factors: (i) the existence of already-approved molecules with good safety profile that continue to be pillars of therapy; (ii) the successful, steady repositioning of approved molecules in new therapeutic niches; and (iii) the continuous development of a significant number of lead molecules that employ a novel mechanism of action or target molecular components of the system (Table 1) that had received poor attention before (e.g. riociguat and NA-1 respectively). It can be predicted that an increasing number of new therapeutic candidates that target the NO-sGC-cGMP pathway will be seeking clinical assessment and approval in the next few years to benefit the treatment of therapeutically challenging, or even intractable, human pathologies (Figure 1).

Table 1.

Summary of the therapeutically amenable molecular targets within the NO-cGMP-sGC axis discussed in this review

Target Molecules Function References
Arginase N-hydroxy-nor-arginine Arginase inhibition; L-Arg preservation Shemyakin et al., 2012; NCT02009527
NOS Caveolin-derived peptide Disruption of NOS–caveolin interaction Bucci et al., 2000
BH4 Cofactor supplementation; enhancement of NOS coupling Alkaitis and Crabtree, 2012; Cunnington et al., 2012; Antoniades et al., 2006
VAS203 (6R,S)-4-amino-5,6,7,8-tetrahydro-L-biopterin NOS inhibition Terpolilli et al., 2009
NA-1 peptide NO-PSD95-(NMDAR) complex uncoupling Bach et al., 2012; Hill et al., 2012
GW274150 iNOS inhibition Høye et al., 2009; Singh et al., 2007
sGC NO anti-inflammatory drug conjugates NO release–anti-inflammatory action NCT00331786; NCT01256775; White et al., 2011
sGC ‘stimulators’ NO-independent, haem-dependent sGC activation Follmann et al., 2013; Ghofrani et al., 2013a,b
sGC ‘activators’ NO- and haem-independent sGC activation Follmann et al., 2013; Vandendriessche et al., 2013
Nitrate–nitrite Bioconversion to NO Lundberg et al., 2008; Cosby et al., 2003; Kapil et al., 2010a
PDE5 PDE inhibitors Enhancement of cGMP signalling Keravis & Lugnier, 2012

Targets and molecules that have been validated preclinically or clinically are shown.

Acknowledgments

A. J. H., A. P. and S. T. receive support from COST Action BM1005: ENOG: European network on gasotransmitters (http://www.gasotransmitters.eu). A. P. and S. T. are also supported by EU FP7 REGPOT CT-2011-285950 – ‘SEE-DRUG’: ‘ESTABLISHMENT OF A CENTRE OF EXCELLENCE FOR STRUCTURE-BASED DRUG TARGET CHARACTERIZATION: STRENGTHENING THE RESEARCH CAPACITY OF SOUTH-EASTERN EUROPE’ (http://www.seedrug.upatras.gr).

Glossary

sGC

soluble GC

Conflict of interest

A. J. H. has acted as a consultant/advisory board member for Bayer AG, Novartis, Merck and Palatin Technologies.

References

  1. Adamo CM, Dai DF, Percival JM, Minami E, Willis MS, Patrucco E, et al. Sildenafil reverses cardiac dysfunction in the mdx mouse model of Duchenne muscular dystrophy. Proc Natl Acad Sci U S A. 2010;107:19079–19083. doi: 10.1073/pnas.1013077107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ahrens I, Habersberger J, Baumlin N, Qian H, Smith BK, Stasch JP, et al. Measuring oxidative burden and predicting pharmacological response in coronary artery disease patients with a novel direct activator of haem-free/oxidised sGC. Atherosclerosis. 2011;218:431–434. doi: 10.1016/j.atherosclerosis.2011.06.042. [DOI] [PubMed] [Google Scholar]
  3. Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J. 2001;357:593–615. doi: 10.1042/0264-6021:3570593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol. 2013a;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Ligand-gated ion channels. Br J Pharmacol. 2013b;170:1582–1606. doi: 10.1111/bph.12446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14:Nuclear hormone receptors. Br J Pharmacol. 2013c;170:1652–1675. doi: 10.1111/bph.12448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Enzymes. Br J Pharmacol. 2013d;170:1797–1867. doi: 10.1111/bph.12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Alkaitis MS, Crabtree MJ. Recoupling the cardiac nitric oxide synthases: tetrahydrobiopterin synthesis and recycling. Curr Heart Fail Rep. 2012;9:200–210. doi: 10.1007/s11897-012-0097-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Antoniades C, Shirodaria C, Warrick N, Cai S, de Bono J, Lee J, et al. 5-methyltetrahydrofolate rapidly improves endothelial function and decreases superoxide production in human vessels: effects on vascular tetrahydrobiopterin availability and endothelial nitric oxide synthase coupling. Circulation. 2006;14:1193–1201. doi: 10.1161/CIRCULATIONAHA.106.612325. [DOI] [PubMed] [Google Scholar]
  10. Bach A, Clausen BH, Møller M, Vestergaard B, Chi CN, Round A, et al. A high-affinity, dimeric inhibitor of PSD-95 bivalently interacts with PDZ1-2 and protects against ischemic brain damage. Proc Natl Acad Sci U S A. 2012;109:3317–3322. doi: 10.1073/pnas.1113761109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Baliga RS, MacAllister RJ, Hobbs AJ. New perspectives for the treatment of pulmonary hypertension. Br J Pharmacol. 2011;163:125–140. doi: 10.1111/j.1476-5381.2010.01164.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Baliga RS, Milsom AB, Ghosh SM, Trinder SL, Macallister RJ, Ahluwalia A, et al. Dietary nitrate ameliorates pulmonary hypertension: cytoprotective role for endothelial nitric oxide synthase and xanthine oxidoreductase. Circulation. 2012;125:2922–2932. doi: 10.1161/CIRCULATIONAHA.112.100586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bandarage UK, Janero DR. Nitric oxide-releasing nonsteroidal anti-inflammatory drugs: novel gastrointestinal-sparing drugs. Mini Rev Med Chem. 2001;1:57–70. doi: 10.2174/1389557013407160. [DOI] [PubMed] [Google Scholar]
  14. Bartelt A, Heeren J. Adipose tissue browning and metabolic health. Nat Rev Endocrinol. 2014;10:24–36. doi: 10.1038/nrendo.2013.204. [DOI] [PubMed] [Google Scholar]
  15. Basu S, Grubina R, Huang J, Conradie J, Huang Z, Jeffers A, et al. Catalytic generation of N2O3 by the concerted nitrite reductase and anhydrase activity of hemoglobin. Nat Chem Biol. 2007;3:785–794. doi: 10.1038/nchembio.2007.46. [DOI] [PubMed] [Google Scholar]
  16. Belge C, Hammond J, Dubois-Deruy E, Manoury B, Hamelet J, Beauloye C, et al. Enhanced expression of β3-adrenoceptors in cardiac myocytes attenuates neurohormone-induced hypertrophic remodeling through nitric oxide synthase. Circulation. 2014;129:451–462. doi: 10.1161/CIRCULATIONAHA.113.004940. [DOI] [PubMed] [Google Scholar]
  17. Bender AT, Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev. 2006;58:488–520. doi: 10.1124/pr.58.3.5. [DOI] [PubMed] [Google Scholar]
  18. Biel M, Michalakis S. Function and dysfunction of CNG channels: insights from channelopathies and mouse models. Mol Neurobiol. 2007;35:266–277. doi: 10.1007/s12035-007-0025-y. [DOI] [PubMed] [Google Scholar]
  19. Boerrigter G, Costello-Boerrigter LC, Cataliotti A, Lapp H, Stasch JP, Burnett JC., Jr Targeting heme-oxidized soluble guanylate cyclase in experimental heart failure. Hypertension. 2007;49:1128–1133. doi: 10.1161/HYPERTENSIONAHA.106.083832. [DOI] [PubMed] [Google Scholar]
  20. Bonderman D, Ghio S, Felix SB, Ghofrani HA, Michelakis E, Mitrovic V, et al. Riociguat for patients with pulmonary hypertension caused by systolic left ventricular dysfunction: a phase IIb double-blind, randomized, placebo-controlled, dose-ranging hemodynamic study. Circulation. 2013;128:502–511. doi: 10.1161/CIRCULATIONAHA.113.001458. [DOI] [PubMed] [Google Scholar]
  21. Bordicchia M, Liu D, Amri EZ, Ailhaud G, Dessì-Fulgheri P, Zhang C, et al. Cardiac natriuretic peptides act via p38 MAPK to induce the brown fat thermogenic program in mouse and human adipocytes. J Clin Invest. 2012;122:1022–1036. doi: 10.1172/JCI59701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Bratasz A, Selvendiran K, Wasowicz T, Bobko A, Khramtsov VV, Ignarro LJ, et al. NCX-4040, a nitric oxide-releasing aspirin, sensitizes drug-resistant human ovarian xenograft tumors to cisplatin by depletion of cellular thiols. J Transl Med. 2008;26:9. doi: 10.1186/1479-5876-6-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Bubb KJ, Trinder SL, Baliga RS, Patel J, Clapp LH, MacAllister RJ, et al. Inhibition of phosphodiesterase 2 augments cGMP and cAMP signaling to ameliorate pulmonary hypertension. Circulation. 2014;130:496–507. doi: 10.1161/CIRCULATIONAHA.114.009751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bucci M, Gratton J-P, Rudic RD, Acevedo L, Roviezzo F, Cirino G, et al. In vivo delivery of the caveolin-1 scaffolding domain inhibits nitric oxide synthesis and reduces inflammation. Nat Med. 2000;6:1362–1367. doi: 10.1038/82176. [DOI] [PubMed] [Google Scholar]
  25. Burley DS, Ferdinandy P, Baxter GF. Cyclic GMP and protein kinase-G in myocardial ischaemia-reperfusion: opportunities and obstacles for survival signaling. Br J Pharmacol. 2007;152:855–869. doi: 10.1038/sj.bjp.0707409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, Cripe L, et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management. Lancet Neurol. 2010a;9:77–93. doi: 10.1016/S1474-4422(09)70271-6. [DOI] [PubMed] [Google Scholar]
  27. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, Cripe L, et al. Care Considerations Working Group, Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care. Lancet Neurol. 2010b;9:177–189. doi: 10.1016/S1474-4422(09)70272-8. [DOI] [PubMed] [Google Scholar]
  28. Buys ES, Cauwels A, Raher MJ, Passeri JJ, Hobai I. sGC(alpha)1(beta)1 attenuates cardiac dysfunction and mortality in murine inflammatory shock models. J Physiol. 2009;297:654–663. doi: 10.1152/ajpheart.00367.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Cao J, Viholainen JI, Dart C, Warwick HK, Leyland ML, Courtney MJ. The PSD95-nNOS interface: a target for inhibition of excitotoxic p38 stress-activated protein kinase activation and cell death. J Cell Biol. 2005;168:117–126. doi: 10.1083/jcb.200407024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Caplin B, Leiper J. Endogenous nitric oxide synthase inhibitors in the biology of disease: markers, mediators, and regulators? Arterioscler Thromb Vasc Biol. 2012;32:1343–1353. doi: 10.1161/ATVBAHA.112.247726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Casey DB, Badejo AM, Jr, Dhaliwal JS, Murthy SN, Hyman AL, Nossaman BD, et al. Pulmonary vasodilator responses to sodium nitrite are mediated by an allopurinol-sensitive mechanism in the rat. Am J Physiol Heart Circ Physiol. 2009;96:H524–H533. doi: 10.1152/ajpheart.00543.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Castro LR, Verde I, Cooper DM, Fischmeister R. Cyclic guanosine monophosphate compartmentation in rat cardiac myocytes. Circulation. 2006;113:2221–2228. doi: 10.1161/CIRCULATIONAHA.105.599241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Cauwels A, Brouckaert P. Nitrite regulation of shock. Cardiovasc Res. 2011;89:553–559. doi: 10.1093/cvr/cvq317. [DOI] [PubMed] [Google Scholar]
  34. Cauwels A, Buys ES, Thoonen R, Geary L, Delanghe J. Nitrite protects against morbidity and mortality associated with TNF- or LPS-induced shock in a soluble guanylate cyclase-dependent manner. J Exp Med. 2009;206:2915–2924. doi: 10.1084/jem.20091236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Chan FK, Wong VW, Suen BY, Wu JC, Ching JY, Hung LC, et al. Combination of a cyclo-oxygenase-2 inhibitor and a proton-pump inhibitor for prevention of recurrent ulcer bleeding in patients at very high risk: a double-blind, randomised trial. Lancet. 2007;369:1621–1626. doi: 10.1016/S0140-6736(07)60749-1. [DOI] [PubMed] [Google Scholar]
  36. Chaudhary SC, Singh T, Kapur P, Weng Z, Arumugam A, Elmets CA, et al. Nitric oxide-releasing sulindac is a novel skin cancer chemopreventive agent for UVB-induced photocarcinogenesis. Toxicol Appl Pharmacol. 2013;268:249–255. doi: 10.1016/j.taap.2012.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Clark RS, Kochanek PM, Obrist WD, Wong HR, Billiar TR, Wisniewski SR, et al. Cerebrospinal fluid and plasma nitrite and nitrate concentrations after head injury in humans. Crit Care Med. 1996;24:1243–1251. doi: 10.1097/00003246-199607000-00030. [DOI] [PubMed] [Google Scholar]
  38. Conole D, Scott LJ. Riociguat: first global approval. Drugs. 2013;73:1967–1975. doi: 10.1007/s40265-013-0149-5. [DOI] [PubMed] [Google Scholar]
  39. Conti M, Beavo J. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu Rev Biochem. 2007;76:481–511. doi: 10.1146/annurev.biochem.76.060305.150444. [DOI] [PubMed] [Google Scholar]
  40. Cosby K, Partovi KS, Crawford JH, Patel RP, Reiter CD, Martyr S, et al. Nitrite reduction to nitric oxide by deoxyhemoglobin vasodilates the human circulation. Nat Med. 2003;9:1498–1505. doi: 10.1038/nm954. [DOI] [PubMed] [Google Scholar]
  41. Costell MH, Ancellin N, Bernard RE, Zhao S, Upson JJ, Morgan LA, et al. Comparison of soluble guanylate cyclase stimulators and activators in models of cardiovascular disease associated with oxidative stress. Front Pharmacol. 2012;3:1–14. doi: 10.3389/fphar.2012.00128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. le Cras TD, Xue C, Rengasamy A, Johns RA. Chronic hypoxia upregulates endothelial and inducible NO synthase gene and protein expression in rat lung. Am J Physiol. 1996;270:164–170. doi: 10.1152/ajplung.1996.270.1.L164. [DOI] [PubMed] [Google Scholar]
  43. Croom KF, Curran MP, Abman SH, Channick RN, Heresi GA, Rubin LJ, et al. Sildenafil: a review of its use in pulmonary arterial hypertension. Drugs. 2008;68:383–397. doi: 10.2165/00003495-200868030-00009. [DOI] [PubMed] [Google Scholar]
  44. Cunnington C, Van Assche T, Shirodaria C, Kylintireas I, Lindsay AC, Lee JM, et al. Systemic and vascular oxidation limits the efficacy of oral tetrahydrobiopterin treatment in patients with coronary artery disease. Circulation. 2012;125:1356–1366. doi: 10.1161/CIRCULATIONAHA.111.038919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Da J, Chen L, Hedenstierna G. Nitric oxide up-regulates the glucocorticoid receptor and blunts the inflammatory reaction in porcine endotoxin sepsis. Crit Care Med. 2007;35:26–32. doi: 10.1097/01.CCM.0000250319.91575.BB. [DOI] [PubMed] [Google Scholar]
  46. Derbyshire E, Marletta MA. Structure and regulation of soluble guanylate cyclase. Annu Rev Biochem. 2012;81:533–559. doi: 10.1146/annurev-biochem-050410-100030. [DOI] [PubMed] [Google Scholar]
  47. Doyle MP, Pickering RA, DeWeert TM, Hoekstra JW, Pater D. Kinetics and mechanism of the oxidation of human deoxyhemoglobin by nitrites. J Biol Chem. 1981;256:12393–12398. [PubMed] [Google Scholar]
  48. Dunkern TR, Feurstein D, Rossi GA, Sabatini F, Hatzelmann A. Inhibition of TGF-beta induced lung fibroblast to myofibroblast conversion by phosphodiesterase inhibiting drugs and activators of soluble guanylyl cyclase. Eur J Pharmacol. 2007;572:12–22. doi: 10.1016/j.ejphar.2007.06.036. [DOI] [PubMed] [Google Scholar]
  49. Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, et al. Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 2011;478:103–109. doi: 10.1038/nature10405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Erdmann E, Semigran MJ, Nieminen MS, Gheorghiade M, Agrawal R, Mitrovic V, et al. Cinaciguat, a soluble guanylate cyclase activator, unloads the heart but also causes hypotension in acute decompensated heart failure. Eur Heart J. 2013a;34:57–67. doi: 10.1093/eurheartj/ehs196. [DOI] [PubMed] [Google Scholar]
  51. Erdmann J, Stark K, Esslinger UB, Rumpf PM, Koesling D, Wit C, et al. Dysfunctional nitric oxide signalling increases risk of myocardial infarction. Nature. 2013b;504:432–436. doi: 10.1038/nature12722. [DOI] [PubMed] [Google Scholar]
  52. Erusalimsky JD, Moncada S. Nitric oxide and mitochondrial signaling: from physiology to pathophysiology. Arterioscler Thromb Vasc Biol. 2007;227:2524–2531. doi: 10.1161/ATVBAHA.107.151167. [DOI] [PubMed] [Google Scholar]
  53. Evgenov OV, Pacher P, Schmidt PM, Haskó G, Schmidt HH, Stasch JP. NO-independent stimulators and activators of soluble guanylate cyclase: discovery and therapeutic potential. Nat Rev Drug Discov. 2006;5:755–768. doi: 10.1038/nrd2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Farrell AJ, Blake DR, Palmer RMJ, Moncada S. Increased concentrations of nitrite in synovial fluid and serum samples suggest increased nitric oxide synthesis in rheumatic diseases. Ann Rheum Dis. 1992;51:1219–1222. doi: 10.1136/ard.51.11.1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Fiorucci S, Distrutti E. COXIBs, CINODs and H2S-releasing NSAIDs: current perspectives in the development of safer non steroidal anti-inflammatory drugs. Curr Med Chem. 2011;18:3494–3505. doi: 10.2174/092986711796642508. [DOI] [PubMed] [Google Scholar]
  56. Fiorucci S, Antonelli E, Distrutti E, Del Soldato P, Flower RJ, Clark MJ, et al. NCX-1015, a nitric-oxide derivative of prednisolone, enhances regulatory T cells in the lamina propria and protects against 2,4,6-trinitrobenzene sulfonic acid-induced colitis in mice. Proc Natl Acad Sci U S A. 2002;99:15770–15775. doi: 10.1073/pnas.232583599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Fiorucci S, Santucci L, Distrutti E. NSAIDs, coxibs, CINOD and H2S-releasing NSAIDs: what lies beyond the horizon. Dig Liver Dis. 2007;39:1043–1051. doi: 10.1016/j.dld.2007.09.001. [DOI] [PubMed] [Google Scholar]
  58. Follmann M, Griebenow N, Hahn MG, Hartung I, Mais F-J, Mittendorf J, et al. The chemistry and biology of soluble guanylate cyclase stimulators and activators. Angew Chem Int Ed Engl. 2013;52:9442–9462. doi: 10.1002/anie.201302588. [DOI] [PubMed] [Google Scholar]
  59. Förstermann U, Li H. Therapeutic effect of enhancing endothelial nitric oxide synthase (eNOS) expression and preventing eNOS uncoupling. Br J Pharmacol. 2011;164:213–223. doi: 10.1111/j.1476-5381.2010.01196.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Francis SH, Busch JL, Corbin JD, Sibley D. cGMP-dependent protein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action. Pharmacol Rev. 2010;62:525–563. doi: 10.1124/pr.110.002907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Friebe A, Mergia E, Dangel O, Lange A, Koesling D. Fatal gastrointestinal obstruction and hypertension in mice lacking nitric oxide-sensitive guanylyl cyclase. Proc Natl Acad Sci U S A. 2007;104:7699–7704. doi: 10.1073/pnas.0609778104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Galiè N, Brundage BH, Ghofrani HA, Oudiz RJ, Simonneau G, Safdar Z, et al. Tadalafil therapy for pulmonary arterial hypertension. Circulation. 2009;119:2894–2903. doi: 10.1161/CIRCULATIONAHA.108.839274. [DOI] [PubMed] [Google Scholar]
  63. Galiè N, Hoeper MM, Humbert M, Torbicki A, Vachiery JL, Barbera JA, et al. Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2011;34:1219–1263. doi: 10.1183/09031936.00139009. [DOI] [PubMed] [Google Scholar]
  64. Garcia-Cardena G, Fan R, Stern DF, Liu J, Sessa WC. Endothelial nitric oxide synthase is regulated by tyrosine phosphorylation and interacts with caveolin-1. J Biol Chem. 1996;271:27237–27240. doi: 10.1074/jbc.271.44.27237. [DOI] [PubMed] [Google Scholar]
  65. Geschka S, Kretschmer A, Sharkovska Y, Evgenov OV, Lawrenz B, Hucke A, et al. Soluble guanylate cyclase stimulation prevents fibrotic tissue remodeling and improves survival in salt-sensitive Dahl rats. PLoS ONE. 2011;6:e21853. doi: 10.1371/journal.pone.0021853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Gheorghiade M, Greene SJ, Filippatos G, Erdmann E, Ferrari R, Levy PD, et al. Cinaciguat, a soluble guanylate cyclase activator: results from the randomized, controlled, phase IIb COMPOSE programme in acute heart failure syndromes. Eur J Heart Fail. 2012;14:1056–1066. doi: 10.1093/eurjhf/hfs093. [DOI] [PubMed] [Google Scholar]
  67. Gheorghiade M, Marti CN, Sabbah HN, Roessig L, Greene SJ, Bohm M, et al. Soluble guanylate cyclase: a potential therapeutic target for heart failure. Heart Fail Rev. 2013;18:123–134. doi: 10.1007/s10741-012-9323-1. [DOI] [PubMed] [Google Scholar]
  68. Ghofrani HA, D'Armini AM, Grimminger F, Hoeper MM, Jansa P, Kim NH, et al. Riociguat for the treatment of chronic thromboembolic pulmonary hypertension. N Engl J Med. 2013a;369:319–329. doi: 10.1056/NEJMoa1209657. [DOI] [PubMed] [Google Scholar]
  69. Ghofrani HA, Galiè N, Grimminger F, Grünig E, Humbert M, Jing ZC, et al. Riociguat for the treatment of pulmonary arterial hypertension. N Engl J Med. 2013b;369:330–340. doi: 10.1056/NEJMoa1209655. [DOI] [PubMed] [Google Scholar]
  70. Ghosh A, Stuehr DJ. Soluble guanylyl cyclase requires heat shock protein 90 for heme insertion during maturation of the NO-active enzyme. Proc Natl Acad Sci U S A. 2012;109:1298–3003. doi: 10.1073/pnas.1205854109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Ghosh SM, Kapil V, Fuentes-Calvo I, Bubb KJ, Pearl V, Milsom AB, et al. Enhanced vasodilator activity of nitrite in hypertension: critical role for erythrocytic xanthine oxidoreductase and translational potential. Hypertension. 2013;61:1091–1102. doi: 10.1161/HYPERTENSIONAHA.111.00933. [DOI] [PubMed] [Google Scholar]
  72. Giaid A, Saleh D. Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med. 1995;333:214–221. doi: 10.1056/NEJM199507273330403. [DOI] [PubMed] [Google Scholar]
  73. Giannetta E, Isidori AM, Galea N, Carbone I, Mandosi E, Vizza CD, et al. Chronic inhibition of cGMP phosphodiesterase 5A improves diabetic cardiomyopathy: a randomized, controlled clinical trial using magnetic resonance imaging with myocardial tagging. Circulation. 2012;125:2323–2333. doi: 10.1161/CIRCULATIONAHA.111.063412. [DOI] [PubMed] [Google Scholar]
  74. Graham DY, Chan FKL. NSAIDs, risks, and gastroprotective strategies: current status and future. Gastroenterology. 2008;134:1240–1246. doi: 10.1053/j.gastro.2008.02.007. [DOI] [PubMed] [Google Scholar]
  75. Guazzi M, Vicenzi M, Arena R, Guazzi MD. PDE5 inhibition with sildenafil improves left ventricular diastolic function, cardiac geometry, and clinical status in patients with stable systolic heart failure: results of a 1-year, prospective, randomized, placebo-controlled study. Circ Heart Fail. 2011;4:8–17. doi: 10.1161/CIRCHEARTFAILURE.110.944694. [DOI] [PubMed] [Google Scholar]
  76. Guazzi M, Vicenzi M, Arena R. Phosphodiesterase 5 inhibition with sildenafil reverses exercise oscillatory breathing in chronic heart failure: a long-term cardiopulmonary exercise testing placebo-controlled study. Eur J Heart Fail. 2012;14:82–90. doi: 10.1093/eurjhf/hfr147. [DOI] [PubMed] [Google Scholar]
  77. Guo DC, Regalado E, Casteel DE, Santos-Cortez RL, Gong L, Kim JJ, et al. Recurrent gain-of-function mutation in PRKG1 causes thoracic aortic aneurysms and acute aortic dissections. Am J Hum Genet. 2013;93:398–404. doi: 10.1016/j.ajhg.2013.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Hållström L, Berghäll E, Frostell C, Sollevi A, Soop AL. Nitric oxide inhalation and glucocorticoids as combined treatment in human experimental endotoxemia. Crit Care Med. 2008;36:3043–3047. doi: 10.1097/CCM.0b013e318186f5b2. [DOI] [PubMed] [Google Scholar]
  79. Hendgen-Cotta UB, Merx MW, Shiva S, Schmitz J, Becher S, Klare JP, et al. Nitrite reductase activity of myoglobin regulates respiration and cellular viability in myocardial ischemia-reperfusion injury. Proc Natl Acad Sci U S A. 2008;105:10256–10261. doi: 10.1073/pnas.0801336105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Hill MD, Martin RH, Mikulis D, Wong JH, Silver FL, Terbrugge KG, et al. Safety and efficacy of NA-1 in patients with iatrogenic stroke after endovascular aneurysm repair (ENACT): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2012;11:942–950. doi: 10.1016/S1474-4422(12)70225-9. [DOI] [PubMed] [Google Scholar]
  81. Hoenicka M, Becker EM, Apeler H, Sirichoke T, Schröder H, Gerzer R, et al. Purified soluble guanylyl cyclase expressed in a baculovirus/Sf9 system: stimulation by YC-1, nitric oxide, and carbon monoxide. J Mol Med (Berl) 1999;77:14–23. doi: 10.1007/s001090050292. [DOI] [PubMed] [Google Scholar]
  82. Hoeper MM, Halank M, Wilkens H, Günther A, Weimann G, Gebert I, et al. Riociguat for interstitial lung disease and pulmonary hypertension: a pilot trial. Eur Respir J. 2013;41:853–860. doi: 10.1183/09031936.00213911. [DOI] [PubMed] [Google Scholar]
  83. Hoffmann J, Goadsby PJ. New agents for acute treatment of migraine: CGRP receptor antagonists, iNOS inhibitors. Curr Treat Options Neurol. 2012;14:50–59. doi: 10.1007/s11940-011-0155-4. [DOI] [PubMed] [Google Scholar]
  84. Hoffmann LS, Schmidt PM, Keim Y, Hoffmann C, Schmidt HHHW, Stasch J-P. Fluorescence dequenching makes haem-free soluble guanylate cyclase detectable in living cells. PLoS ONE. 2011;6:e23596. doi: 10.1371/journal.pone.0023596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Hofmann F, Feil R, Kleppisch T, Schlossmann J. Function of cGMP-dependent protein kinases as revealed by gene deletion. Physiol Rev. 2006;86:1–23. doi: 10.1152/physrev.00015.2005. [DOI] [PubMed] [Google Scholar]
  86. Høivik HO, Laurijssens BE, Harnisch LO, Twomey CK, Dixon RM, Kirkham AJ, et al. Lack of efficacy of the selective iNOS inhibitor GW274150 in prophylaxis of migraine headache. Cephalalgia. 2010;30:1458–1467. doi: 10.1177/0333102410370875. [DOI] [PubMed] [Google Scholar]
  87. Høye KLB, Harnisch LO, Twomey CK, Dixon RM, Kirkham AJ, Williams PM, et al. Efficacy and tolerability of the iNOS inhibitor GW274150 administered up to 120 mg daily for 12 weeks in the prophylactic treatment of migraine. Cephalalgia. 2009;29:132. [Google Scholar]
  88. Hunter CJ, Dejam A, Blood AB, Shields H, Kim-Shapiro DB, Machado RF, et al. Inhaled nebulized nitrite is a hypoxia-sensitive NO-dependent selective pulmonary vasodilator. Nat Med. 2004;10:1122–1127. doi: 10.1038/nm1109. [DOI] [PubMed] [Google Scholar]
  89. Ichinose F, Roberts JD, Jr, Zapol WM. Inhaled nitric oxide: a selective pulmonary vasodilator: current uses and therapeutic potential. Circulation. 2004;109:3106–3111. doi: 10.1161/01.CIR.0000134595.80170.62. [DOI] [PubMed] [Google Scholar]
  90. Jones NC, Constantin D, Gibson CL, Prior MJ, Morris PG, Marsden CA, et al. A detrimental role for nitric oxide synthase-2 in the pathology resulting from acute cerebral injury. J Neuropathol Exp Neurol. 2004;63:708–720. doi: 10.1093/jnen/63.7.708. [DOI] [PubMed] [Google Scholar]
  91. Kamisaki Y, Saheki S, Nakane M, Palmieri JA, Kuno T, Chang BY, et al. Soluble guanylate cyclase from rat lung exists as a heterodimer. J Biol Chem. 1986;261:7236–7241. [PubMed] [Google Scholar]
  92. Kapil V, Milsom AB, Okorie M, Maleki-Toyserkani S, Akram F, Rehman F, et al. Inorganic nitrate supplementation lowers blood pressure in humans: role for nitrite-derived NO. Hypertension. 2010a;56:274–281. doi: 10.1161/HYPERTENSIONAHA.110.153536. [DOI] [PubMed] [Google Scholar]
  93. Kapil V, Webb AJ, Ahluwalia A. Inorganic nitrate and the cardiovascular system. Heart. 2010b;96:1703–1709. doi: 10.1136/hrt.2009.180372. [DOI] [PubMed] [Google Scholar]
  94. Keravis T, Lugnier C. Cyclic nucleotide phosphodiesterase (PDE) isozymes as targets of the intracellular signalling network: benefits of PDE inhibitors in various diseases and perspectives for future therapeutic developments. Br J Pharmacol. 2012;165:1288–1305. doi: 10.1111/j.1476-5381.2011.01729.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Khong SM, Andrews KL, Huynh NN, Venardos K, Aprico A, Michell DL, et al. Arginase II inhibition prevents nitrate tolerance. Br J Pharmacol. 2012;166:2015–2023. doi: 10.1111/j.1476-5381.2012.01876.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Klinger JR. The nitric oxide/cGMP signaling pathway in pulmonary hypertension. Clin Chest Med. 2007;28:143–167. doi: 10.1016/j.ccm.2006.12.002. [DOI] [PubMed] [Google Scholar]
  97. Kukreja RC, Salloum FN, Das A, Koka S, Ockaili RA, Xi L. Emerging new uses of phosphodiesterase-5 inhibitors in cardiovascular diseases. Exp Clin Cardiol. 2011;16:e30–e35. [PMC free article] [PubMed] [Google Scholar]
  98. Lang M, Kojonazarov B, Tian X, Kalymbetov A, Weissmann N, Grimminger F, et al. The soluble guanylate cyclase stimulator riociguat ameliorates pulmonary hypertension induced by hypoxia and SU5416 in rats. PLoS ONE. 2012;7:e43433. doi: 10.1371/journal.pone.0043433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Larsen FJ, Ekblom B, Sahlin K, Lundberg JO, Weitzberg E. Effects of dietary nitrate on blood pressure in healthy volunteers. N Engl J Med. 2006;355:2792–2793. doi: 10.1056/NEJMc062800. [DOI] [PubMed] [Google Scholar]
  100. Leiper J, Nandi M. The therapeutic potential of targeting endogenous inhibitors of nitric oxide synthesis. Nat Rev Drug Discov. 2011;10:277–291. doi: 10.1038/nrd3358. [DOI] [PubMed] [Google Scholar]
  101. Li H, Förstermann U. Uncoupling of endothelial NO synthase in atherosclerosis and vascular disease. Curr Opin Pharmacol. 2013;13:161–167. doi: 10.1016/j.coph.2013.01.006. [DOI] [PubMed] [Google Scholar]
  102. Lima B, Forrester MT, Hess DT, Stamler JS. S-nitrosylation in cardiovascular signaling. Circ Res. 2010;106:633–646. doi: 10.1161/CIRCRESAHA.109.207381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Linder AE, McCluskey LP, Cole KR, III, Lanning KM, Webb RC. Dynamic association of nitric oxide downstream signaling molecules with endothelial caveolin-1 in rat aorta. J Pharmacol Exp Ther. 2005;314:9–15. doi: 10.1124/jpet.105.083634. [DOI] [PubMed] [Google Scholar]
  104. López A, Lorente JA, Steingrub J, Bakker J, McLuckie A, Willatts S, et al. Multiple-center, randomized, placebo-controlled, double-blind study of the nitric oxide synthase inhibitor 546C88: effect on survival in patients with septic shock. Crit Care Med. 2004;32:21–30. doi: 10.1097/01.CCM.0000105581.01815.C6. [DOI] [PubMed] [Google Scholar]
  105. Lundberg JO, Weitzberg E, Gladwin MT. The nitrate–nitrite–nitric oxide pathway in physiology and therapeutics. Nat Rev Drug Discov. 2008;7:156–167. doi: 10.1038/nrd2466. [DOI] [PubMed] [Google Scholar]
  106. Lundberg JO, Gladwin MT, Ahluwalia A, Benjamin N, Bryan NS, Butler A, et al. Nitrate and nitrite in biology, nutrition and therapeutics. Nat Chem Biol. 2009;5:865–869. doi: 10.1038/nchembio.260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Martin EA, Barresi R, Byrne BJ, Tsimerinov EI, Scott BL, Walker AE, et al. Tadalafil alleviates muscle ischemia in patients with Becker muscular dystrophy. Sci Transl Med. 2012;4:155–162. doi: 10.1126/scitranslmed.3004327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Martin F, Baskaran P, Ma X, Dunten PW, Schaefer M, Stasch JP, et al. Structure of cinaciguat (BAY 58-2667) bound to Nostoc H-NOX domain reveals insights into heme-mimetic activation of the soluble guanylyl cyclase. J Biol Chem. 2010;285:22651–22657. doi: 10.1074/jbc.M110.111559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Millar TM, Stevens CR, Benjamin N, Eisenthal R, Harrison R, Blake DR. Xanthine oxidoreductase catalyses the reduction of nitrates and nitrite to nitric oxide under hypoxic conditions. FEBS Lett. 1998;427:225–228. doi: 10.1016/s0014-5793(98)00430-x. [DOI] [PubMed] [Google Scholar]
  110. Morris SM., Jr Recent advances in arginine metabolism: roles and regulation of the arginases. Br J Pharmacol. 2009;157:922–930. doi: 10.1111/j.1476-5381.2009.00278.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Münzel T, Daiber A, Ullrich V, Mülsch A. Vascular consequences of endothelial nitric oxide synthase uncoupling for the activity and expression of the soluble guanylyl cyclase and the cGMP-dependent protein kinase. Arterioscler Thromb Vasc Biol. 2005;25:1551–1557. doi: 10.1161/01.ATV.0000168896.64927.bb. [DOI] [PubMed] [Google Scholar]
  112. Nathan C. Inducible nitric oxide synthase: what difference does it make? J Clin Invest. 1997;100:2417–2423. doi: 10.1172/JCI119782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Nausch LW, Ledoux J, Bonev AD, Nelson MT, Dostmann WR. Differential patterning of cGMP in vascular smooth muscle cells revealed by single GFP-linked biosensors. Proc Natl Acad Sci U S A. 2008;105:365–370. doi: 10.1073/pnas.0710387105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C, Sciorati C, et al. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science. 2003;299:896–899. doi: 10.1126/science.1079368. [DOI] [PubMed] [Google Scholar]
  115. Oess S, Icking A, Fulton D, Govers R, Müller-Esterl W. Subcellular targeting and trafficking of nitric oxide synthases. Biochem J. 2006;396:401–409. doi: 10.1042/BJ20060321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Oliver JJ, Hughes VE, Dear JW, Webb DJ. Clinical potential of combined organic nitrate and phosphodiesterase type 5 inhibitor in treatment-resistant hypertension. Hypertension. 2010;56:62–67. doi: 10.1161/HYPERTENSIONAHA.109.147686. [DOI] [PubMed] [Google Scholar]
  117. Palmer JE, Guillard FL, Laurijssens BE, Wentz AL, Dixon RM, Williams PM, et al. A randomised, single-blind, placebo-controlled, adaptive clinical trial of GW274150, a selective iNOS inhibitor, in the treatment of acute migraine. Cephalalgia. 2009;29:124. [Google Scholar]
  118. Paul-Clark MJ, Roviezzo F, Flower RJ, Cirino G, Soldato PD, Adcock IM, et al. Glucocorticoid receptor nitration leads to enhanced anti-inflammatory effects of novel steroid ligands. J Immunol. 2003;171:3245–3252. doi: 10.4049/jimmunol.171.6.3245. [DOI] [PubMed] [Google Scholar]
  119. Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. NC-IUPHAR. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledgebase of drug targets and their ligands. Nucl Acids Res. 2014;42:D1098–D1106. doi: 10.1093/nar/gkt1143. (Database Issue) [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Pellicena P, Karow DS, Boon EM, Marletta MA, Kuriyan J. Crystal structure of an oxygen-binding heme domain related to soluble guanylate cyclases. Proc Natl Acad Sci U S A. 2004;101:12854–12859. doi: 10.1073/pnas.0405188101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Pfeifer A, Klatt P, Massberg S, Ny L, Sausbier M, Hirneiss C, et al. Defective smooth muscle regulation in cGMP kinase I-deficient mice. EMBO J. 1998;17:3045–3051. doi: 10.1093/emboj/17.11.3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Phillips PG, Long L, Wilkins MR, Morrell NW. cAMP phosphodiesterase inhibitors potentiate effects of prostacyclin analogs in hypoxic pulmonary vascular remodelling. Am J Physiol Lung Cell Mol Physiol. 2005;288:L103–L115. doi: 10.1152/ajplung.00095.2004. [DOI] [PubMed] [Google Scholar]
  123. Piggott LA, Hassell KA, Berkova Z, Morris AP, Silberbach M, Rich TC. Natriuretic peptides and nitric oxide stimulate cGMP synthesis in different cellular compartments. J Gen Physiol. 2006;128:3–14. doi: 10.1085/jgp.200509403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Purohit R, Fritz BG, The J, Issaian A, Weichsel A, David CL, et al. YC-1 binding to the beta subunit of the soluble guanylyl cyclase overcomes allosteric inhibition by the alpha subunit. Biochemistry. 2014;53:101–114. doi: 10.1021/bi4015133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Ranieri VM, Thompson BT, Barie PS, Dhainaut J-F, Douglas IS. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med. 2012;366:2055–2064. doi: 10.1056/NEJMoa1202290. [DOI] [PubMed] [Google Scholar]
  126. Rassaf T, Ferdinandy P, Schulz R. Nitrite in organ protection. Br J Pharmacol. 2014;171:1–11. doi: 10.1111/bph.12291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Redfield MM, Chen HH, Borlaug BA, Semigran MJ, Lee KL, Lewis G, et al. Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. JAMA. 2013;309:1268–1277. doi: 10.1001/jama.2013.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Rinecker M, Plesnila N, Baethmann A, Stoffel M. Secondary growth of a cortical necrosis: effect of NOS inhibition by aminoguanidine post insult. Acta Neurochir (Wien) 2003;145:977–981. doi: 10.1007/s00701-003-0131-1. [DOI] [PubMed] [Google Scholar]
  129. Roberts JD, Polaner DM, Lang P, Zapol WM. Inhaled nitric oxide in persistent pulmonary hypertension of the newborn. Lancet. 1992;340:818–819. doi: 10.1016/0140-6736(92)92686-a. [DOI] [PubMed] [Google Scholar]
  130. Rosen RC, Kostis JB. Overview of phosphodiesterase 5 inhibition in erectile dysfunction. Am J Cardiol. 2003;92:9M–18M. doi: 10.1016/s0002-9149(03)00824-5. [DOI] [PubMed] [Google Scholar]
  131. Rosenzweig EB. Tadalafil for the treatment of pulmonary arterial hypertension. Expert Opin Pharmacother. 2010;11:127–132. doi: 10.1517/14656560903413542. [DOI] [PubMed] [Google Scholar]
  132. Rossaint R, Falke KJ, Lopez F, Slama K, Pison U, Zapol WM. Inhaled nitric oxide for the adult respiratory distress syndrome. N Engl J Med. 1993;328:399–405. doi: 10.1056/NEJM199302113280605. [DOI] [PubMed] [Google Scholar]
  133. Rudyk O, Phinikaridou A, Prysyazhna O, Burgoyne JR, Botnar RM, Eaton P. Protein kinase G oxidation is a major cause of injury during sepsis. Proc Natl Acad Sci U S A. 2013;110:9909–9913. doi: 10.1073/pnas.1301026110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Sakurai H, Kohsaka W, Liu MF, Higashiyama H, Hirata Y, Kanno K, et al. Nitric oxide production and inducible nitric oxide synthase expression in inflammatory arthritides. J Clin Invest. 1995;96:2357–2363. doi: 10.1172/JCI118292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Sansbury BE, Cummins TD, Tang Y, Hellmann J, Holden CR, Harbeson MA, et al. Overexpression of endothelial nitric oxide synthase prevents diet-induced obesity and regulates adipocyte phenotype. Circ Res. 2012;111:1176–1189. doi: 10.1161/CIRCRESAHA.112.266395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Sayed N, Kim DD, Fioramonti X, Iwahashi T, Duran WN, Beuve A. Nitroglycerin-induced S-nitrosylation and desensitization of soluble guanylyl cyclase contribute to nitrate tolerance. Circ Res. 2008;103:606–614. doi: 10.1161/CIRCRESAHA.108.175133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Schäfer A, Fraccarollo D, Werner L, Bauersachs J. Guanylyl cyclase activator ataciguat improves vascular function and reduces platelet activation in heart failure. Pharmacol Res. 2010;62:432–438. doi: 10.1016/j.phrs.2010.06.008. [DOI] [PubMed] [Google Scholar]
  138. Schermuly RT, Pullamsetti SS, Kwapiszewska G, Dumitrascu R, Tian X, Weissmann N, et al. Phosphodiesterase 1 upregulation in pulmonary arterial hypertension: target for reverse-remodeling therapy. Circulation. 2007;115:2331–2339. doi: 10.1161/CIRCULATIONAHA.106.676809. [DOI] [PubMed] [Google Scholar]
  139. Schermuly RT, Stasch JP, Pullamsetti SS, Middendorff R, Müller D, Schlüter KD, et al. Expression and function of soluble guanylate cyclase in pulmonary arterial hypertension. Eur Respir J. 2008;32:881–891. doi: 10.1183/09031936.00114407. [DOI] [PubMed] [Google Scholar]
  140. Schermuly RT, Ghofrani HA, Wilkins MR, Grimminger F. Mechanisms of disease: pulmonary arterial hypertension. Nat Rev Cardiol. 2011;8:443–455. doi: 10.1038/nrcardio.2011.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Schindler U, Strobel H, Schönafinger K, Linz W, Löhn M, Martorana PA, et al. Biochemistry and pharmacology of novel anthranilic acid derivatives activating heme-oxidized soluble guanylyl cyclase. Mol Pharmacol. 2006;69:1260–1268. doi: 10.1124/mol.105.018747. [DOI] [PubMed] [Google Scholar]
  142. Schmidt PM, Schramm M, Schröder H, Wunder F, Stasch JP. Identification of residues crucially involved in the binding of the heme moiety of soluble guanylate cyclase. J Biol Chem. 2004;279:3025–3032. doi: 10.1074/jbc.M310141200. [DOI] [PubMed] [Google Scholar]
  143. Seymour M, Pétavy F, Chiesa F, Perry H, Lukey PT, Binks M, et al. Ultrasonographic measures of synovitis in an early phase clinical trial: a double-blind, randomised, placebo and comparator controlled phase IIa trial of GW274150 (a selective inducible nitric oxide synthase inhibitor) in rheumatoid arthritis. Clin Exp Rheumatol. 2012;30:254–261. [PubMed] [Google Scholar]
  144. Shemyakin A, Kövamees O, Rafnsson A, Böhm F, Svenarud P, Settergren M, et al. Arginase inhibition improves endothelial function in patients with coronary artery disease and type 2 diabetes mellitus. Circulation. 2012;126:2943–2950. doi: 10.1161/CIRCULATIONAHA.112.140335. [DOI] [PubMed] [Google Scholar]
  145. Siddiqi N, Bruce M, Neil CJ, Jagpal B, Maclennon G, Cotton SC, et al. Protocol: does sodium nitrite administration reduce ischaemia-reperfusion injury in patients presenting with acute ST segment elevation myocardial infarction? Nitrites in acute myocardial infarction (NIAMI) J Transl Med. 2013;11:116. doi: 10.1186/1479-5876-11-116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Singh D, Richards D, Knowles RG, Schwartz S, Woodcock A, Langley S, et al. Selective inducible nitric oxide synthase inhibition has no effect on allergen challenge in asthma. Am J Respir Crit Care Med. 2007;176:988–993. doi: 10.1164/rccm.200704-588OC. [DOI] [PubMed] [Google Scholar]
  147. del Soldato P, Sorrentino R, Pinto A. NO-aspirins: a class of new antiinflammatory and antithrombotic agents. Trends Pharmacol Sci. 1999;20:319–323. doi: 10.1016/s0165-6147(99)01353-x. [DOI] [PubMed] [Google Scholar]
  148. Starr A, Hussein D, Nandi M. The regulation of vascular tetrahydrobiopterin bioavailability. Vascul Pharmacol. 2013;58:219–230. doi: 10.1016/j.vph.2012.08.002. [DOI] [PubMed] [Google Scholar]
  149. Stasch J-P, Schmidt P, Alonso-Alija C, Apeler H, Dembowsky K. NO- and haem-independent activation of soluble guanylyl cyclase: molecular basis and cardiovascular implications of a new pharmacological principle. Br J Pharmacol. 2002;136:773–783. doi: 10.1038/sj.bjp.0704778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Stasch JP, Evgenov OV. Soluble guanylate cyclase stimulators in pulmonary hypertension. Handb Exp Pharmacol. 2013;218:279–313. doi: 10.1007/978-3-642-38664-0_12. [DOI] [PubMed] [Google Scholar]
  151. Stasch JP, Hobbs AJ. NO-independent, haem-dependent soluble guanylate cyclase stimulators. Handb Exp Pharmacol. 2009;191:277–308. doi: 10.1007/978-3-540-68964-5_13. [DOI] [PubMed] [Google Scholar]
  152. Stasch JP, Schmidt PM, Nedvetsky PI, Nedvetskaya TY, Arun Kumar HS, Meurer S, et al. Targeting the heme-oxidized nitric oxide receptor for selective vasodilatation of diseased blood vessels. J Clin Invest. 2006;116:2552–2561. doi: 10.1172/JCI28371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Stasch JP, Pacher P, Evgenov OV. Soluble guanylate cyclase as an emerging therapeutic target in cardiopulmonary disease. Circulation. 2011;123:2263–2273. doi: 10.1161/CIRCULATIONAHA.110.981738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Stefanovic-Racic MS, Stadler J, Evans CH. Nitric oxide and arthritis. Arthritis Rheum. 1993;36:1036–1044. doi: 10.1002/art.1780360803. [DOI] [PubMed] [Google Scholar]
  155. Straub AC, Lohman AW, Billaud M, Johnstone SR, Dwyer ST, Lee MY, et al. Endothelial cell expression of haemoglobin α regulates nitric oxide signalling. Nature. 2012;491:473–477. doi: 10.1038/nature11626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Tamargo J, López-Sendón J. Novel therapeutic targets for the treatment of heart failure. Nat Rev Drug Discov. 2011;10:536–555. doi: 10.1038/nrd3431. [DOI] [PubMed] [Google Scholar]
  157. Taylor RW, Zimmerman JL, Dellinger RP, Straube RC, Criner GJ, Davis K, Jr, et al. Low-dose inhaled nitric oxide in patients with acute lung injury: a randomized controlled trial. J Am Med Assoc. 2004;291:1603–1609. doi: 10.1001/jama.291.13.1603. [DOI] [PubMed] [Google Scholar]
  158. Terpolilli NA, Zweckberger K, Trabold R, Schilling L, Schinzel R, Tegtmeier F, et al. The novel nitric oxide synthase inhibitor 4-amino-tetrahydro-L-biopterine prevents brain edema formation and intracranial hypertension following traumatic brain injury in mice. J Neurotrauma. 2009;26:1963–1975. doi: 10.1089/neu.2008.0853. [DOI] [PubMed] [Google Scholar]
  159. Tian X, Vroom C, Ghofrani AH, Weissmann N, Bieniek E, Grimminger F. Phosphodiesterase 10A upregulation contributes to pulmonary vascular remodeling. PLoS ONE. 2011;6:e18136. doi: 10.1371/journal.pone.0018136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Tiso M, Tejero J, Basu S, Azarov I, Wang X, Simplaceanu V, et al. Human neuroglobin functions as a redox-regulated nitrite reductase. J Biol Chem. 2011;286:18277–18289. doi: 10.1074/jbc.M110.159541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Tsai EJ, Kass DA. Cyclic GMP signaling in cardiovascular pathophysiology and therapeutics. Pharmacol Ther. 2009;122:216–238. doi: 10.1016/j.pharmthera.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Turesin F, Del Soldato P, Wallace JL. Enhanced anti-inflammatory potency of a nitric oxide-releasing derivative of prednisolone in the rat. Br J Pharmacol. 2003;139:966–972. doi: 10.1038/sj.bjp.0705324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Vandendriessche B, Rogge E, Goossens V, Vandenabeele P, Stasch JP, Brouckaert P, et al. The soluble guanylate cyclase activator BAY 58-2667 protects against morbidity and mortality in endotoxic shock by recoupling organ systems. PLoS ONE. 2013;8:e72155. doi: 10.1371/journal.pone.0072155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Vermeersch P, Buys E, Pokreisz P, Marsboom G, Ichinose F, Sips P. Soluble guanylate cyclase-alpha1 deficiency selectively inhibits the pulmonary vasodilator response to nitric oxide and increases the pulmonary vascular remodeling response to chronic hypoxia. Circulation. 2007;116:936–943. doi: 10.1161/CIRCULATIONAHA.106.677245. [DOI] [PubMed] [Google Scholar]
  165. Vosatka RJ, Kashyap S, Trifiletti RR. Arginine deficiency accompanies persistent pulmonary hypertension of the newborn. Biol Neonate. 1994;66:65–70. doi: 10.1159/000244091. [DOI] [PubMed] [Google Scholar]
  166. Wallace JL, Rizzo G, Cirino G, Del Soldato P, Fiorucci S. Enhanced anti-inflammatory potency of a nitric oxide-releasing derivative of flunisolide: role of nuclear factor-kappaB. J Pharmacol Exp Ther. 2004;310:1096–1102. doi: 10.1124/jpet.104.067850. [DOI] [PubMed] [Google Scholar]
  167. Webb A, Bond R, McLean P, Uppal R, Benjamin N, Ahluwalia A. Reduction of nitrite to nitric oxide during ischemia protects against myocardial ischemia-reperfusion damage. Proc Natl Acad Sci U S A. 2004;101:13683–13688. doi: 10.1073/pnas.0402927101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Webb AJ, Milsom AB, Rathod KS, Chu WL, Qureshi S, Lovell MJ, et al. Mechanisms underlying erythrocyte and endothelial nitrite reduction to nitric oxide in hypoxia: role for xanthine oxidoreductase and endothelial nitric oxide synthase. Circ Res. 2008a;103:957–964. doi: 10.1161/CIRCRESAHA.108.175810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Webb AJ, Patel N, Loukogeorgakis S, Okorie M, Aboud Z, Misra S, et al. Acute blood pressure lowering, vasoprotective, and antiplatelet properties of dietary nitrate via bioconversion to nitrite. Hypertension. 2008b;51:784–790. doi: 10.1161/HYPERTENSIONAHA.107.103523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Weber M, Lauer N, Mulsch A, Kojda G. The effect of peroxynitrite on the catalytic activity of soluble guanylyl cyclase. Free Radic Biol Med. 2001;31:1360–1367. doi: 10.1016/s0891-5849(01)00706-7. [DOI] [PubMed] [Google Scholar]
  171. Weissmann N, Hackemack S, Dahal BK, Pullamsetti SS, Savai R, Mittal M, et al. The soluble guanylate cyclase activator HMR1766 reverses hypoxia-induced experimental pulmonary hypertension in mice. Am J Physiol Lung Cell Mol Physiol. 2009;297:658–665. doi: 10.1152/ajplung.00189.2009. [DOI] [PubMed] [Google Scholar]
  172. White WB, Schnitzer TJ, Bakris GL, Frayssinet H, Duquesroix B, Weber M. Effects of naproxcinod on blood pressure in patients with osteoarthritis. Am J Cardiol. 2011;107:1338–1345. doi: 10.1016/j.amjcard.2010.12.046. [DOI] [PubMed] [Google Scholar]
  173. Williams JL, Kashfi K, Ouyang N, del Soldato P, Kopelovich L, Rigas B. NO-donating aspirin inhibits intestinal carcinogenesis in Min (APC(Min/+)) mice. Biochem Biophys Res Commun. 2004;313:784–788. doi: 10.1016/j.bbrc.2003.12.015. [DOI] [PubMed] [Google Scholar]
  174. Winger JA, Marletta MA. Expression and characterization of the catalytic domains of soluble guanylate cyclase: interaction with the heme domain. Biochemistry. 2005;44:4083–4090. doi: 10.1021/bi047601d. [DOI] [PubMed] [Google Scholar]
  175. Wolfe MM, Lichtenstein DR, Singh G. Gastrointestinal toxicity of nonsteroidal antiinflammatory drugs. N Engl J Med. 1999;340:1888–1899. doi: 10.1056/NEJM199906173402407. [DOI] [PubMed] [Google Scholar]
  176. Wu CC, Ko FN, Kuo SC, Lee FY, Teng CM. YC-1 inhibited human platelet aggregation through NO-independent activation of soluble guanylate cyclase. Br J Pharmacol. 1995;116:1973–1978. doi: 10.1111/j.1476-5381.1995.tb16400.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Xu W, Kaneko FT, Zheng S, Comhair SA, Janocha AJ, Goggans T, et al. Increased arginase II and decreased NO synthesis in endothelial cells of patients with pulmonary arterial hypertension. FASEB J. 2004;18:1746–1748. doi: 10.1096/fj.04-2317fje. [DOI] [PubMed] [Google Scholar]
  178. Zhang M, Kass DA. Phosphodiesterases and cardiac cGMP: evolving roles and controversies. Trends Pharmacol Sci. 2011;32:360–365. doi: 10.1016/j.tips.2011.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Zhang Z, Naughton D, Winyard PG, Benjamin N, Blake DR, Symons MC. Generation of nitric oxide by a nitrite reductase activity of xanthine oxidase: a potential pathway for nitric oxide formation in the absence of nitric oxide synthase activity. Biochem Biophys Res Commun. 1998;249:767–772. doi: 10.1006/bbrc.1998.9226. [DOI] [PubMed] [Google Scholar]
  180. Zisman DA, Schwarz M, Anstrom KJ, Collard HR, Flaherty KR, Hunninghake GW, et al. A controlled trial of sildenafil in advanced idiopathic pulmonary fibrosis. N Engl J Med. 2010;363:620–628. doi: 10.1056/NEJMoa1002110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Zuckerbraun BS, Shiva S, Ifedigbo E, Mathier MA, Mollen KP, Rao J, et al. Nitrite potently inhibits hypoxic and inflammatory pulmonary arterial hypertension and smooth muscle proliferation via xanthine oxidoreductase-dependent nitric oxide generation. Circulation. 2010;121:98–109. doi: 10.1161/CIRCULATIONAHA.109.891077. [DOI] [PubMed] [Google Scholar]
  182. Zwolinska-Wcislo M, Brzozowski T, Ptak-Belowska A, Targosz A, Urbanczyk K, Kwiecien S, et al. Nitric oxide-releasing aspirin but not conventional aspirin improves healing of experimental colitis. World J Gastroenterol. 2011;17:4076–4089. doi: 10.3748/wjg.v17.i36.4076. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES