Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2017 Mar 23;175(2):223–231. doi: 10.1111/bph.13749

Targeting phosphodiesterase 5 as a therapeutic option against myocardial ischaemia/reperfusion injury and for treating heart failure

Sevil Korkmaz‐Icöz 1,, Tamás Radovits 2, Gábor Szabó 1
PMCID: PMC5758391  PMID: 28213937

Abstract

Phosphodiesterase type 5 (PDE5) selectively hydrolyses the second messenger cGMP into 5′‐GMP, thereby regulating its intracellular concentrations. Dysregulation of the cGMP‐dependent pathway plays a significant role in various cardiovascular diseases. Therefore, its modulation by drugs, such as PDE5 inhibitors, may represent an effective therapeutic approach. There are currently four PDE5 inhibitors available for the treatment of erectile dysfunction: sildenafil, vardenafil, tadalafil and avanafil. Sildenafil and tadalafil have also received Food and Drug Administration approval for the treatment of pulmonary arterial hypertension. This review summarizes the pharmacological aspects and clinical potential of PDE5 inhibition for the treatment of myocardial ischaemia/reperfusion injury and heart failure.

Linked Articles

This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc


Abbreviations

Bcl 2

B‐cell lymphoma 2

HF

heart failure

IR

ischemia/reperfusion

KCa

calcium‐activated potassium channel

LAD

left anterior descending artery

PGC‐1α

PPAR gamma coactivator 1‐alpha

SIRT1

sirtuin 1

Tables of Links

TARGETS
Other protein targets a Enzymes e
FABP4 Acetyl CoA carboxylase
TNF‐α Adenylate cyclase
GPCRs b Akt (PKB)
GLP‐1 receptor ERK1
Nuclear hormone receptors c ERK2
PPARγ FASN
Transporters d Hormone sensitive lipase (HSL)
GLUT4 PDE5A
PKA
SIRT1

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 (Southan et al., 2016), and are permanently archived in the Concise Guide to PHARMACOLOGY 2015/16 (a,b,c,d,eAlexander et al., 2015a,b,c,d,e).

PDE5

PDEs are a superfamily of enzymes that catalyse the breakdown of the intracellular second messengers cAMP and cGMP into their corresponding inactive 5′‐AMP and 5′‐GMP forms (Bender and Beavo, 2006). In total, 11 PDE families (PDE1–PDE11) (Omori and Kotera, 2007) have been identified, differing in amino acid sequence, affinity and turnover rates for cAMP and cGMP, cellular expression and intracellular localization. Of all the PDEs, PDE5, the main cGMP‐metabolizing enzyme, has been extensively investigated. The three major PDE5 isoforms described and identified are A1, A2 and A3 (Lin et al., 2000a). They share similar cGMP‐catalytic activities but differ at their N‐terminal regions (Lin et al., 2000b). They also showed similar sensitivity to inhibition by sildenafil, although the PDE5A1 isoform seems more resistant than PDE5A2 or PDE5A3 (Lin, 2004). There are no obvious functional differences in PDE5A1, PDE5A2 and PDE5A3. PDE5 was originally identified and purified from rat platelets (Coquil et al., 1980) and lungs (Francis et al., 1980). Although less widely distributed compared with many other PDEs, PDE5 is highly expressed in vascular and bronchial smooth muscle, in renal tubules and in platelets. Its expression in cardiac tissue has been more controversial. Previous studies, aimed at quantifying mRNA (using either RT‐PCR or northern blot) and proteins (using immunohistochemical staining), have demonstrated the expression of PDE5 in heart tissue (Loughney et al., 1998; Wallis et al., 1999), whereas no PDE5 enzymatic activity and/or no immunoreactive bands were observed in human ventricular tissue (Wallis et al., 1999). However, this concept was challenged by a study demonstrating an abundant expression of PDE5 in isolated canine ventricular cardiomyocytes, as assessed by immunohistochemistry (Senzaki et al., 2001), and also in murine ventricular cardiomyocytes, as shown by RT‐PCR, western blot and immunohistochemical assay (Das et al., 2005). PDE5A is generally considered to be a cytosolic protein (Lin, 2004). The sub‐cellular localization of PDE5A, myocyte z‐bands, was not observed when cells were isolated from failing myocytes, suggesting its intracellular localization is altered in these cells (Senzaki et al., 2001). Nevertheless, PDE5 is highly expressed in both experimental and human heart disease (Nagendran et al., 2007; Pokreisz et al., 2009), making it a promising therapeutic target for cardiovascular disease.

PDE5 inhibitors available for patient use

There are currently four commercially available oral PDE5 inhibitors: sildenafil (commonly known as Viagra; Pfizer Inc, Berlin, Germany), vardenafil (Levitra; Bayer HealthCare Pharmaceuticals, Berlin, Germany), tadalafil (Cialis; Eli Lilly, Bad Homburg vor der Höhe, Germany) and avanafil (Stendra; Vivus, Campbell, CA, USA). They all display similar pharmacological properties and structure (Figure 1). Sildenafil, discovered in 1989, was originally developed for the treatment of coronary artery disease (Chrysant and Chrysant, 2012). Although sildenafil did not prove effective for the treatment of coronary heart disease, due to the risk of coronary steal or hypotension, it revealed the interesting side effect of penile erection. These drugs are already in clinical use for the treatment of erectile dysfunction (Boolell et al., 1996). In addition to this, sildenafil (under the trade name Revatio®) (Croom and Curran, 2008) and tadalafil (Adcirca®) (Henrie et al., 2015) were approved by the Food and Drug Administration as vasodilators for the treatment of pulmonary arterial hypertension.

Figure 1.

Figure 1

Molecular structures of the four clinically approved PDE5 inhibitors: sildenafil, vardenafil, tadalafil and avanafil.

Mechanism of action of PDE5 inhibition

All of the aforementioned, specific PDE5 inhibitors share the same mechanisms of action involving an inhibition of the PDE5 isoenzyme, which prevents cGMP breakdown, thereby increasing its intracellular concentration. Increased cGMP, in turn, activates PKG, stimulates the phosphorylation of proteins regulating smooth muscle tone and reduces cytosolic calcium concentrations. Local regulation of vascular tone and blood flow is primarily dependent upon the release of endothelial NOS‐derived NO. After diffusing from the endothelial cell into subjacent vascular smooth muscle cells, NO activates guanylate cyclase, the enzyme that catalyses the production of cGMP from GTP. As cGMP is degraded by PDE5, inhibiting this enzyme prevents cGMP breakdown, leading to smooth muscle relaxation and consequently vasodilatation. In the myocardium, the cardioprotective mechanisms of PDE5 inhibition include the activation of cGMP‐dependent PKG, which involves the activation of ERK and inhibition of glycogen synthase kinase 3β (GSK3β; Das et al., 2008), and additionally includes the activation of PKC and the opening of mitochondrial ATP‐sensitive potassium channels (KATP channels) (Kukreja et al., 2005), leading to cytoprotection.

Myocardial protection with PDE5 inhibitors

Cardiac PDE5 expression was reported to be up‐regulated in hypertrophic, dilated and ischaemic cardiomyopathy (Nagendran et al., 2007; Pokreisz et al., 2009; Vandeput et al., 2009) and in conditions such as congestive heart failure (HF) (Lu et al., 2010). Therefore, in certain disorders, the pharmacological inhibition of PDE5 could significantly increase intracellular cGMP levels in the myocardium. This restoration of the cardioprotective cGMP–PKG pathway could then exert cardioprotective properties (Kukreja et al., 2012). Therefore, enhanced cGMP signalling by PDE5 inhibition may attenuate myocardial ischaemia/reperfusion (IR) injury. Moreover, recent preclinical studies suggest PDE5 inhibition has direct myocardial effects, which may contribute to its therapeutic effects during heart transplantation and in HF.

Myocardial ischaemia/reperfusion injury

Percutaneous coronary interventions, stenting, coronary bypass surgery, heart transplantation and acute myocardial infarction are associated with myocardial IR injury. Ischaemia is a condition suffered by tissues/organs due to deprived blood flow, followed by an inadequate oxygen and nutrient supply. Paradoxically, the reperfusion of previously ischaemic myocardium acutely causes additional tissue damage. The underlying mechanisms of IR injury are complex and multifactorial. They include intracellular calcium overload or redistribution, the production of ROS, energy depletion, rapid restoration of physiological pH levels and inflammation‐inducing organ dysfunction (Hearse and Bolli, 1992). In the clinical situation, there are four basic types of reperfusion injury: (i) myocardial stunning (Ambrosio and Tritto, 2001), which can be found after reperfusion of a globally ischaemic myocardium or in the setting of regional IR; (ii) reperfusion arrhythmias (Goldberg et al., 1983), including ventricular arrhythmias such as ventricular tachycardia and fibrillation; (iii) lethal, irreversible reperfusion injury (Piper et al., 2003); and (iv) vascular injury (endothelial and microvascular dysfunction including the no‐reflow phenomenon) (Kloner, 1993).

Regional myocardial ischaemia/reperfusion injury

Experimental in vivo studies

There continues to be a great interest in exploring sildenafil's role in myocardial protection against IR injury in animal models. In 2002, it was demonstrated that a treatment of rabbits with sildenafil (0.7 mg·kg−1, iv), 30 min and 24 h prior to ischaemia (30 min) and 3 h of reperfusion, was potent at protecting the myocardium from injury in both the acute and delayed phase, inducing a significant reduction in infarct size compared with the vehicle treatment (Ockaili et al., 2002). This cardioprotective effect is mediated through the opening of mitochondrial KATP channels (Ockaili et al., 2002) and the activation of calcium‐activated potassium (KCa) channels (Wang et al., 2008). This preconditioning‐like effect in the heart has also been observed in other species such as rats (Das et al., 2002), mice (Salloum et al., 2003) and dogs (Reffelmann and Kloner, 2003). Moreover, post conditioning, that is, administration of sildenafil or vardenafil at reperfusion, also induced significant cardioprotection, similar to the preconditioning‐like effects (Salloum et al., 2007). Furthermore, a previous study by Nagy et al. (2004) showed, in dogs, that orally administered sildenafil decreases the incidence of severe life‐threatening ventricular arrhythmias, which arise early after coronary artery occlusion.

Clinical studies

Compared to the extensive in vivo experimental studies, there is a lack of clinical trials examining the protective effects of PDE5 inhibitors against regional IR injury in the myocardium. In a population‐based study, Anderson et al. (2016) showed that in type 2 diabetic patients with a history of acute myocardial infarction or who have suffered acute myocardial infarction during the study period, the use of PDE5 inhibitors (sildenafil, vardenafil or tadalafil) was associated with a lower frequency of acute myocardial infarction and lower mortality.

Global myocardial ischaemia/reperfusion injury

Experimental in vivo studies

Although most in vivo preclinical studies investigating the effects of PDE5 inhibitors were performed in experimental models of myocardial IR, a limited number of studies have been conducted in an in vivo model of global IR injury. In our laboratory, the effects of vardenafil on global IR injury were investigated using our well‐established rat model of heterotopic heart transplantation (Szabo et al., 2002; Loganathan et al., 2015; Hegedus et al., 2016). Our results show that a preconditioning of donor rats with vardenafil (10 μg·kg−1, i.v.), 1 h before explantation, restores systolic and diastolic left ventricular function close to the level of hearts not subjected to IR injury after transplantation (Loganathan et al., 2008). Also, in a clinically relevant large animal model of cardiopulmonary bypass with hypothermic cardiac arrest, we have showed that the application of vardenafil improves myocardial and endothelial functions (Szabo et al., 2009).

Clinical studies

In contrast to preclinical studies, there have been no clinical investigations on the direct effects of PDE5 inhibitors on graft function after transplantation. The clinical studies with PDE5 inhibitors that have been done include patients with pulmonary artery hypertension and/or right ventricular functional recovery after heart transplantation or hypertension. Data from a cohort of high‐risk heart transplant recipients showed that sildenafil has a therapeutic role in the management of acute right ventricular dysfunction in heart transplant recipients with pulmonary hypertension, and can reduce the requirement for inotropes (De Santo et al., 2008). An interesting study published in 2003 (Schofield et al., 2003) demonstrated that 50 mg of sildenafil is well tolerated in hypertensive heart transplant recipients and does not induce hypotension despite the continuation of background antihypertensive medication.

PDE inhibitors in treating heart failure

HF is a condition in which the cardiac contractile performance is insufficient to provide the oxygen requirements of the body, which results in the hypoperfusion of peripheral organs, even at rest. The two main types of HF are acute HF, which develops suddenly, initially with symptoms that are severe, and chronic HF, a long‐term condition that is associated with the heart undergoing adaptive responses (e.g. hypertrophy). Hence, the treatment of HF is a challenging task.

Acute heart failure

Acute decompensated HF is commonly due to left ventricular systolic or diastolic dysfunction, with or without additional cardiac pathologies, such as coronary artery disease or valve abnormalities. The main principles in the treatment of acute HF remain the restoration of oxygenation and the improvement of haemodynamics and organ perfusion. Even though few studies have evaluated the effects of PDE5 inhibitors on acute HF, a previous study by Salloum showed that tadalafil prevents acute HF with reduced ejection fraction in mice (Salloum et al., 2014). However, the potential adverse effects of sildenafil, vardenafil and tadalafil include hypotension (Kloner, 2004), which can be a concern in certain populations of at risk patients. Therefore, these drugs could contribute to adverse clinical outcomes in a number of patients with acute decompensated HF.

Chronic heart failure

Experimental in vivo studies

Volume overload‐induced heart failure

A recent study was the first to show that chronic PDE5 inhibition with sildenafil can attenuate left ventricular remodelling and HF following left ventricular volume overload caused by mechanically‐induced chronic mitral regurgitation (Kim et al., 2012). In this study the cardioprotective effects of sildenafil were associated with its anti‐apoptotic and anti‐inflammatory effects. Sildenafil has been shown to increase both endothelial and inducible NOS proteins, and NO is thought to induce a preconditioning effect in the cardiomyocytes (Das et al., 2005). The preliminary results of Das et al. (2005) suggest that the elevatation of cGMP induced by sildenafil may activate PKG, which contributes to its anti‐apoptotic effects in mouse isolated cardiomyocytes. Additionally, sildenafil treatment has been shown to increase the Bcl‐2 to Bax ratio and, as it is well known that Bcl‐2 is a key anti‐apoptotic protein, this may be one of the factors responsible for the attenuated cardiomyocyte apoptosis (Reed, 1994; Das et al., 2005). Furthermore, sildenafil has been found to reduce the expression of IL‐6, IL‐18, inducible NOS and cyclin‐dependent kinase inhibitor 2a, which further substantiates that it has an inhibitory effect on cardiac inflammation (Kim et al., 2012).

Pressure overload‐induced heart failure

Sustained pressure overload induces pathological cardiac hypertrophy and HF. In a chronic pressure overload mouse model of transverse aortic constriction, Takimoto et al. (2005) showed that blocking the intrinsic catabolism of cGMP by the administration of sildenafil, p.o., suppresses chamber and myocyte hypertrophy and improves in vivo heart function. Furthermore, sildenafil reversed the pre‐established hypertrophy induced by pressure overload, and restored the chamber function to normal (Takimoto et al., 2005).

Congestive heart failure in diabetes mellitus

We have shown that a chronic treatment with vardenafil improves contractile function, without affecting the mean arterial pressure, in a rat model of streptozotocin‐induced diabetic cardiomyopathy (Radovits et al., 2009). Furthermore, vardenafil did not induce a direct positive inotropic effect (no modulating effect on myocardial contractility was observed) in non‐diabetic control rats. Accordingly, the improved cardiac function in the diabetic treated group is a specific phenomenon; this direct cardiac effect of vardenafil reflects a reversal of diabetic cardiomyopathy rather than the consequence of non‐specific effects (Radovits et al., 2009). Recently, Matyas et al. (2017) showed that the treatment of type 2 diabetic rats with vardenafil prevented the development of HF with preserved ejection fraction.

NO production has been shown to activate sirtuin (SIRT)1, a histone deacetylase that binds and activates the PPARγ coactivator (PGC)‐1α, a major regulator of cellular energy metabolism. Chronic treatment with tadalafil has been demonstrated to improve mitochondrial respiratory function via activation of NO‐induced SIRT1‐PGC‐1α signalling (Koka et al., 2014), which attenuates oxidative stress and improves mitochondrial integrity (Koka et al., 2013). Moreover, tadalafil therapy reduces circulating levels of two key pro‐inflammatory cytokines, TNF‐α and IL‐1β, while also improving fasting glucose levels and reducing infarct size following IR injury in obese, diabetic mice (Varma et al., 2012). Additionally, in a model of diabetic cardiomyopathy, chronic sildenafil treatment improves cardiac kinetics and circulating markers through an anti‐remodelling mechanism that is independent of vascular, endothelial or metabolic factors (Giannetta et al., 2012).

Doxorubicin‐induced heart failure

Doxorubicin is associated with both morphological and functional cardiac changes, which are similar to those of dilated cardiomyopathy. In vivo pretreatment of mice with sildenafil has been shown to attenuate doxorubicin‐induced cardiotoxicity (Fisher et al., 2005). Additionally, Koka et al. (2010) also concluded that tadalafil, a long‐acting PDE5 inhibitor, improves left ventricular function and prevents cardiomyocyte apoptosis during doxorubicin‐induced cardiomyopathy in mice. They suggested that the mechanisms of tadalafil involved the up‐regulation of cGMP/PKG activity and manganese superoxide dismutase levels, and that these effects of tadalafil did not interfer with the chemotherapeutic benefits of doxorubicin.

Postinfraction heart failure

Congestive HF, secondary to acute myocardial infarction, continues to be a major complication. Chronic treatment with sildenafil has been shown to attenuate ischaemic cardiomyopathy in mice by limiting necrosis within the first 24 h, and by reducing apoptosis and preserving left ventricular function at 7 and 28 days; it improves survival at 28 days after left anterior descending coronary artery (LAD) ligation, possibly through an NO‐dependent pathway (Salloum et al., 2008). Additionally, Salloum et al. (2014) have shown that chronic treatment with tadalafil, starting immediately following myocardial infarction, attenuates the progression of ischaemic HF in mice. They suggested that tadalafil induces this protective effect by reducing necrosis, apoptosis and myocardial fibrosis, thereby blunting adverse left ventricular remodelling.

Clinical studies

In a randomized, double‐blind, placebo‐controlled study, chronic sildenafil treatment was shown to improve exercise capacity and quality of life (Lewis et al., 2007). It acts as a selective pulmonary vasodilator at rest and during exercise in patients with systolic HF and secondary pulmonary hypertension (Lewis et al., 2007). In line with these observations, a meta‐analysis of randomized clinical trials, comparing sildenafil with a placebo, indicates that sildenafil improves the haemodynamic parameters in HF patients with reduced ejection fraction (Zhuang et al., 2014). Additionally, Bocchi et al. (2002) have shown that a single oral administration of sildenafil to patients with HF was associated with an improvement in exercise capacity. Because sildenafil decreases the left ventricular afterload, it acutely improves cardiac performance in patients with chronic HF (Hirata et al., 2005). Also Guazzi et al. (2007) showed that in chronic HF, sildenafil improves ventilation during exercise and improves aerobic efficiency. This effect of sildenafil was found to be significantly associated with an endothelium‐mediated attenuation of muscle over‐signalling during exercise (Guazzi et al., 2007). Additionally, intermediate‐term administration (4 weeks) of sildenafil to outpatients with stable but chronic HF improves exercise performance and ventilatory efficiency, oxygen uptake kinetics and pulmonary hypertension (Behling et al., 2008). However, these beneficial effects were not confirmed by the RELAX trial (Redfield et al., 2013). In this trial, chronic treatment with sildenafil for 24 weeks did not improve exercise capacity, quality of life, clinical status or left ventricular remodelling in 216 elderly HF with preserved ejection fraction patients when compared with a placebo. The lack of beneficial effects of sildenafil in this RELAX trial is not fully understood but may be attributable to the fact that the patients did not have pulmonary hypertension or to the fact that an impaired cGMP ‘production’, rather than an increased ‘degradation’, may be the predominant pathophysiological mechanism in HF with preserved ejection fraction (Redfield et al., 2013). According to novel experimental data, long‐term PDE5 inhibition might serve as a useful tool to prevent the development of HF with preserved ejection fraction in high risk patients (such as diabetics), rather than to treat/reverse the symptoms of this disease (Matyas et al., 2017).

Potential mechanisms

The potential mechanisms by which PDE5 inhibition improves myocardial function are complex and are summarized in Figure 2. The prominent expression of PDE in bronchial smooth muscle and in blood vessels suggests that PDE5 inhibitors exert their beneficial cardiac effects by affecting systemic and regional haemodynamics. However, experimental studies suggest sildenafil has a direct protective effect on adult cardiomyocytes against necrosis and apoptosis following ischaemia/reoxygenation injury. The underlying mechanism involves the NO signalling pathway (Das et al., 2005). In addition, Guazzi (2008) has shown that the increase in cGMP activity evoked by inhibition of PDE5 has a direct effect on the myocardium, independent of the vascular action that may block adrenergic, hypertrophic and pro‐apoptotic signalling and contribute to this cardioprotective effect.

Figure 2.

Figure 2

Proposed mechanisms for the cardioprotective actions of PDE5 inhibitors. PDE5 inhibitors activate PKG by preventing the breakdown of cGMP and increasing the generation of NO‐driven cGMP. The NO–cGMP–PKG signalling pathway has a direct cardioprotective effect on the myocardium by opening mitochondrial ATP‐dependent potassium channels (mitoKATP) and stimulating calcium activated potassium channels (KCa). In addition, PDE5 inhibition promotes anti‐apoptotic and anti‐inflammatory effects, decreases hypertrophy, ventricular arrhythmias and myocardial fibrosis and also reduces oxidative stress by activating SIRT1. The NO–cGMP–PKG pathway also leads to beneficial cardiac effects by inducing direct vasodilator responses. Hence, basic science data and clinical studies suggest PDE5 inhibitors have potential as a treatment for myocardial IR injury and chronic HF.

Apart from inhibiting PDE5, PDE5 inhibitors also possess antioxidant activity. It has been shown that oxidative stress increases the expression of PDE5 in the cardiomyocytes of failing hearts (Lu et al., 2010) and sildenafil attenuated oxidative stress by inhibiting free radical formation and by facilitating antioxidant redox systems (Bivalacqua et al., 2009; Perk et al., 2008). One of the mechanisms of action of PDE inhibitors might be to reduce oxidative stress levels, as PDE levels are up‐regulated in conditions such as pulmonary hypertension, chronic HF and right ventricular hypertrophy (Corbin et al., 2005; Forfia et al., 2007; Nagendran et al., 2007) and oxidative stress mediates the dysfunctional NO–cGMP–PKG signalling in cardiovascular disease. Therefore, PDE5 inhibitors may have therapeutic benefits in a variety of pathologies (Das et al., 2015).

Conclusions

While evidence from experimental data and to a lesser extent clinical studies suggest that PDE5 inhibitors are cardioprotective, carefully conducted clinical trials are required before extrapolating the encouraging results of these experimental studies to clinical indications. The re‐purposing of these drugs represents an attractive option for the treatment of patients with advanced HF and diabetic patients with HF, and also may be suitable for a wide range of other clinical areas, such as heart transplants, coronary bypass surgery, acute myocardial infarction and pathological cardiac hypertrophy.

Conflict of interest

The authors declare no conflicts of interest.

Acknowledgements

The authors thank Dr Mihály Ruppert for his constructive comments and Paige Brlecic for English corrections. This work was supported by the Land Baden‐Württemberg, Germany, by the Medical Faculty of the University of Heidelberg, Germany (to Dr S. Korkmaz‐Icöz), by the János Bolyai Research Scholarship of the Hungarian Academy of Sciences (to Dr T. Radovits) and by the National Research, Development and Innovation Office of Hungary (NKFIA; NVKP‐16‐1‐2016‐0017).

Korkmaz‐Icöz, S. , Radovits, T. , and Szabó, G. (2018) Targeting phosphodiesterase 5 as a therapeutic option against myocardial ischaemia/reperfusion injury and for treating heart failure. British Journal of Pharmacology, 175: 223–231. doi: 10.1111/bph.13749.

References

  1. Alexander SPH, Kelly E, Marrion N, Peters JA, Benson HE, Faccenda E et al. (2015a). The Concise Guide to PHARMACOLOGY 2015/16: Overview. Br J Pharmacol 172: 5729–5143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Alexander SPH, Davenport AP, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015b). The Concise Guide to PHARMACOLOGY 2015/16: G protein‐coupled receptors. Br J Pharmacol 172: 5744–5869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alexander SPH, Cidlowski JA, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015c). The Concise Guide to PHARMACOLOGY 2015/16: Nuclear hormone receptors. Br J Pharmacol 172: 5956–5978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alexander SPH, Kelly E, Marrion N, Peters JA, Benson HE, Faccenda E et al. (2015d). The Concise Guide to PHARMACOLOGY 2015/16: Transporters. Br J Pharmacol 172: 6110–6202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Alexander SPH, Fabbro D, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015e). The concise guide to PHARMACOLOGY 2015/16: Enzymes. Br J Pharmacol 172: 6024–6109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Ambrosio G, Tritto I (2001). Clinical manifestations of myocardial stunning. Coron Artery Dis 12: 357–361. [DOI] [PubMed] [Google Scholar]
  7. Anderson SG, Hutchings DC, Woodward M, Rahimi K, Rutter MK, Kirby M et al. (2016). Phosphodiesterase type‐5 inhibitor use in type 2 diabetes is associated with a reduction in all‐cause mortality. Heart (British Cardiac Society) 102: 1750–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Behling A, Rohde LE, Colombo FC, Goldraich LA, Stein R, Clausell N (2008). Effects of 5′‐phosphodiesterase four‐week long inhibition with sildenafil in patients with chronic heart failure: a double‐blind, placebo‐controlled clinical trial. J Card Fail 14: 189–197. [DOI] [PubMed] [Google Scholar]
  9. Bender AT, Beavo JA (2006). Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev 58: 488–520. [DOI] [PubMed] [Google Scholar]
  10. Bivalacqua TJ, Sussan TE, Gebska MA, Strong TD, Berkowitz DE, Biswal S et al. (2009). Sildenafil inhibits superoxide formation and prevents endothelial dysfunction in a mouse model of secondhand smoke induced erectile dysfunction. J Urol 181: 899–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bocchi EA, Guimaraes G, Mocelin A, Bacal F, Bellotti G, Ramires JF (2002). Sildenafil effects on exercise, neurohormonal activation, and erectile dysfunction in congestive heart failure: a double‐blind, placebo‐controlled, randomized study followed by a prospective treatment for erectile dysfunction. Circulation 106: 1097–1103. [DOI] [PubMed] [Google Scholar]
  12. Boolell M, Allen MJ, Ballard SA, Gepi‐Attee S, Muirhead GJ, Naylor AM et al. (1996). Sildenafil: an orally active type 5 cyclic GMP‐specific phosphodiesterase inhibitor for the treatment of penile erectile dysfunction. Int J Impot Res 8: 47–52. [PubMed] [Google Scholar]
  13. Chrysant SG, Chrysant GS (2012). The pleiotropic effects of phosphodiesterase 5 inhibitors on function and safety in patients with cardiovascular disease and hypertension. J Clin Hypertens (Greenwich) 14: 644–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Coquil JF, Franks DJ, Wells JN, Dupuis M, Hamet P (1980). Characteristics of a new binding protein distinct from the kinase for guanosine 3′:5′‐monophosphate in rat platelets. Biochim Biophys Acta 631: 148–165. [DOI] [PubMed] [Google Scholar]
  15. Corbin JD, Beasley A, Blount MA, Francis SH (2005). High lung PDE5: a strong basis for treating pulmonary hypertension with PDE5 inhibitors. Biochem Biophys Res Commun 334: 930–938. [DOI] [PubMed] [Google Scholar]
  16. Croom KF, Curran MP (2008). Sildenafil: a review of its use in pulmonary arterial hypertension. Drugs 68: 383–397. [DOI] [PubMed] [Google Scholar]
  17. Das A, Durrant D, Salloum FN, Xi L, Kukreja RC (2015). PDE5 inhibitors as therapeutics for heart disease, diabetes and cancer. Pharmacol Ther 147: 12–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Das A, Xi L, Kukreja RC (2005). Phosphodiesterase‐5 inhibitor sildenafil preconditions adult cardiac myocytes against necrosis and apoptosis. Essential role of nitric oxide signaling. J Biol Chem 280: 12944–12955. [DOI] [PubMed] [Google Scholar]
  19. Das A, Xi L, Kukreja RC (2008). Protein kinase G‐dependent cardioprotective mechanism of phosphodiesterase‐5 inhibition involves phosphorylation of ERK and GSK3beta. J Biol Chem 283: 29572–29585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Das S, Maulik N, Das DK, Kadowitz PJ, Bivalacqua TJ (2002). Cardioprotection with sildenafil, a selective inhibitor of cyclic 3′,5′‐monophosphate‐specific phosphodiesterase 5. Drugs Exp Clin Res 28: 213–219. [PubMed] [Google Scholar]
  21. De Santo LS, Mastroianni C, Romano G, Amarelli C, Marra C, Maiello C et al. (2008). Role of sildenafil in acute posttransplant right ventricular dysfunction: successful experience in 13 consecutive patients. Transplant Proc 40: 2015–2018. [DOI] [PubMed] [Google Scholar]
  22. Fisher PW, Salloum F, Das A, Hyder H, Kukreja RC (2005). Phosphodiesterase‐5 inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular dysfunction in a chronic model of doxorubicin cardiotoxicity. Circulation 111: 1601–1610. [DOI] [PubMed] [Google Scholar]
  23. Forfia PR, Lee M, Tunin RS, Mahmud M, Champion HC, Kass DA (2007). Acute phosphodiesterase 5 inhibition mimics hemodynamic effects of B‐type natriuretic peptide and potentiates B‐type natriuretic peptide effects in failing but not normal canine heart. J Am Coll Cardiol 49: 1079–1088. [DOI] [PubMed] [Google Scholar]
  24. Francis SH, Lincoln TM, Corbin JD (1980). Characterization of a novel cGMP binding protein from rat lung. J Biol Chem 255: 620–626. [PubMed] [Google Scholar]
  25. Giannetta E, Isidori AM, Galea N, Carbone I, Mandosi E, Vizza CD et al. (2012). Chronic Inhibition of cGMP phosphodiesterase 5A improves diabetic cardiomyopathy: a randomized, controlled clinical trial using magnetic resonance imaging with myocardial tagging. Circulation 125: 2323–2333. [DOI] [PubMed] [Google Scholar]
  26. Goldberg S, Greenspon AJ, Urban PL, Muza B, Berger B, Walinsky P et al. (1983). Reperfusion arrhythmia: a marker of restoration of antegrade flow during intracoronary thrombolysis for acute myocardial infarction. Am Heart J 105: 26–32. [DOI] [PubMed] [Google Scholar]
  27. Guazzi M (2008). Sildenafil and phosphodiesterase‐5 inhibitors for heart failure. Curr Heart Fail Rep 5: 110–114. [DOI] [PubMed] [Google Scholar]
  28. Guazzi M, Samaja M, Arena R, Vicenzi M, Guazzi MD (2007). Long‐term use of sildenafil in the therapeutic management of heart failure. J Am Coll Cardiol 50: 2136–2144. [DOI] [PubMed] [Google Scholar]
  29. Hearse DJ, Bolli R (1992). Reperfusion induced injury: manifestations, mechanisms, and clinical relevance. Cardiovasc Res 26: 101–108. [DOI] [PubMed] [Google Scholar]
  30. Hegedus P, Li S, Korkmaz‐Icoz S, Radovits T, Mayer T, Al Said S et al. (2016). Dimethyloxalylglycine treatment of brain‐dead donor rats improves both donor and graft left ventricular function after heart transplantation. J Heart Lung Transplant 35: 99–107. [DOI] [PubMed] [Google Scholar]
  31. Henrie AM, Nawarskas JJ, Anderson JR (2015). Clinical utility of tadalafil in the treatment of pulmonary arterial hypertension: an evidence‐based review. Core Evid 10: 99–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Hirata K, Adji A, Vlachopoulos C, O'Rourke MF (2005). Effect of sildenafil on cardiac performance in patients with heart failure. Am J Cardiol 96: 1436–1440. [DOI] [PubMed] [Google Scholar]
  33. Kim KH, Kim YJ, Ohn JH, Yang J, Lee SE, Lee SW et al. (2012). Long‐term effects of sildenafil in a rat model of chronic mitral regurgitation: benefits of ventricular remodeling and exercise capacity. Circulation 125: 1390–1401. [DOI] [PubMed] [Google Scholar]
  34. Kloner RA (1993). Does reperfusion injury exist in humans? J Am Coll Cardiol 21: 537–545. [DOI] [PubMed] [Google Scholar]
  35. Kloner RA (2004). Cardiovascular effects of the 3 phosphodiesterase‐5 inhibitors approved for the treatment of erectile dysfunction. Circulation 110: 3149–3155. [DOI] [PubMed] [Google Scholar]
  36. Koka S, Aluri HS, Xi L, Lesnefsky EJ, Kukreja RC (2014). Chronic inhibition of phosphodiesterase 5 with tadalafil attenuates mitochondrial dysfunction in type 2 diabetic hearts: potential role of NO/SIRT1/PGC‐1alpha signaling. Am J Physiol 306: H1558–H1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Koka S, Das A, Salloum FN, Kukreja RC (2013). Phosphodiesterase‐5 inhibitor tadalafil attenuates oxidative stress and protects against myocardial ischemia/reperfusion injury in type 2 diabetic mice. Free Radic Biol Med 60: 80–88. [DOI] [PubMed] [Google Scholar]
  38. Koka S, Das A, Zhu SG, Durrant D, Xi L, Kukreja RC (2010). Long‐acting phosphodiesterase‐5 inhibitor tadalafil attenuates doxorubicin‐induced cardiomyopathy without interfering with chemotherapeutic effect. J Pharmacol Exp Ther 334: 1023–1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Kukreja RC, Salloum F, Das A, Ockaili R, Yin C, Bremer YA et al. (2005). Pharmacological preconditioning with sildenafil: basic mechanisms and clinical implications. Vascul Pharmacol 42: 219–232. [DOI] [PubMed] [Google Scholar]
  40. Kukreja RC, Salloum FN, Das A (2012). Cyclic guanosine monophosphate signaling and phosphodiesterase‐5 inhibitors in cardioprotection. J Am Coll Cardiol 59: 1921–1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Lewis GD, Shah R, Shahzad K, Camuso JM, Pappagianopoulos PP, Hung J et al. (2007). Sildenafil improves exercise capacity and quality of life in patients with systolic heart failure and secondary pulmonary hypertension. Circulation 116: 1555–1562. [DOI] [PubMed] [Google Scholar]
  42. Lin CS (2004). Tissue expression, distribution, and regulation of PDE5. Int J Impot Res 16 (Suppl 1): S8–S10. [DOI] [PubMed] [Google Scholar]
  43. Lin CS, Lau A, Tu R, Lue TF (2000a). Expression of three isoforms of cGMP‐binding cGMP‐specific phosphodiesterase (PDE5) in human penile cavernosum. Biochem Biophys Res Commun 268: 628–635. [DOI] [PubMed] [Google Scholar]
  44. Lin CS, Lau A, Tu R, Lue TF (2000b). Identification of three alternative first exons and an intronic promoter of human PDE5A gene. Biochem Biophys Res Commun 268: 596–602. [DOI] [PubMed] [Google Scholar]
  45. Loganathan S, Korkmaz‐Icoz S, Radovits T, Li S, Mikles B, Barnucz E et al. (2015). Effects of soluble guanylate cyclase activation on heart transplantation in a rat model. J Heart Lung Transplant 34: 1346–1353. [DOI] [PubMed] [Google Scholar]
  46. Loganathan S, Radovits T, Hirschberg K, Korkmaz S, Barnucz E, Karck M et al. (2008). Effects of selective phosphodiesterase‐5‐inhibition on myocardial contractility and reperfusion injury after heart transplantation. Transplantation 86: 1414–1418. [DOI] [PubMed] [Google Scholar]
  47. Loughney K, Hill TR, Florio VA, Uher L, Rosman GJ, Wolda SL et al. (1998). Isolation and characterization of cDNAs encoding PDE5A, a human cGMP‐binding, cGMP‐specific 3′,5′‐cyclic nucleotide phosphodiesterase. Gene 216: 139–147. [DOI] [PubMed] [Google Scholar]
  48. Lu Z, Xu X, Hu X, Lee S, Traverse JH, Zhu G et al. (2010). Oxidative stress regulates left ventricular PDE5 expression in the failing heart. Circulation 121: 1474–1483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Matyas C, Nemeth BT, Olah A, Torok M, Ruppert M, Kellermayer D et al. (2017). Prevention of the development of heart failure with preserved ejection fraction by the phosphodiesterase‐5A inhibitor vardenafil in rats with type 2 diabetes. Eur J Heart Fail 19: 326–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Nagendran J, Archer SL, Soliman D, Gurtu V, Moudgil R, Haromy A et al. (2007). Phosphodiesterase type 5 is highly expressed in the hypertrophied human right ventricle, and acute inhibition of phosphodiesterase type 5 improves contractility. Circulation 116: 238–248. [DOI] [PubMed] [Google Scholar]
  51. Nagy O, Hajnal A, Parratt JR, Vegh A (2004). Sildenafil (Viagra) reduces arrhythmia severity during ischaemia 24 h after oral administration in dogs. Br J Pharmacol 141: 549–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Ockaili R, Salloum F, Hawkins J, Kukreja RC (2002). Sildenafil (Viagra) induces powerful cardioprotective effect via opening of mitochondrial K(ATP) channels in rabbits. Am J Physiol 283: H1263–H1269. [DOI] [PubMed] [Google Scholar]
  53. Omori K, Kotera J (2007). Overview of PDEs and their regulation. Circ Res 100: 309–327. [DOI] [PubMed] [Google Scholar]
  54. Perk H, Armagan A, Naziroglu M, Soyupek S, Hoscan MB, Sutcu R et al. (2008). Sildenafil citrate as a phosphodiesterase inhibitor has an antioxidant effect in the blood of men. J Clin Pharm Ther 33: 635–640. [DOI] [PubMed] [Google Scholar]
  55. Piper HM, Meuter K, Schafer C (2003). Cellular mechanisms of ischemia‐reperfusion injury. Ann Thorac Surg 75: S644–S648. [DOI] [PubMed] [Google Scholar]
  56. Pokreisz P, Vandenwijngaert S, Bito V, Van den Bergh A, Lenaerts I, Busch C et al. (2009). Ventricular phosphodiesterase‐5 expression is increased in patients with advanced heart failure and contributes to adverse ventricular remodeling after myocardial infarction in mice. Circulation 119: 408–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Radovits T, Bomicke T, Kokeny G, Arif R, Loganathan S, Kecsan K et al. (2009). The phosphodiesterase‐5 inhibitor vardenafil improves cardiovascular dysfunction in experimental diabetes mellitus. Br J Pharmacol 156: 909–919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Redfield MM, Chen HH, Borlaug BA, Semigran MJ, Lee KL, Lewis G et al. (2013). Effect of phosphodiesterase‐5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. JAMA 309: 1268–1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Reed JC (1994). Bcl‐2 and the regulation of programmed cell death. J Cell Biol 124: 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Reffelmann T, Kloner RA (2003). Effects of sildenafil on myocardial infarct size, microvascular function, and acute ischemic left ventricular dilation. Cardiovasc Res 59: 441–449. [DOI] [PubMed] [Google Scholar]
  61. Salloum F, Yin C, Xi L, Kukreja RC (2003). Sildenafil induces delayed preconditioning through inducible nitric oxide synthase‐dependent pathway in mouse heart. Circ Res 92: 595–597. [DOI] [PubMed] [Google Scholar]
  62. Salloum FN, Abbate A, Das A, Houser JE, Mudrick CA, Qureshi IZ et al. (2008). Sildenafil (Viagra) attenuates ischemic cardiomyopathy and improves left ventricular function in mice. Am J Physiol 294: H1398–H1406. [DOI] [PubMed] [Google Scholar]
  63. Salloum FN, Chau VQ, Hoke NN, Kukreja RC (2014). Tadalafil prevents acute heart failure with reduced ejection fraction in mice. Cardiovasc Drugs Ther 28: 493–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Salloum FN, Takenoshita Y, Ockaili RA, Daoud VP, Chou E, Yoshida K et al. (2007). Sildenafil and vardenafil but not nitroglycerin limit myocardial infarction through opening of mitochondrial K(ATP) channels when administered at reperfusion following ischemia in rabbits. J Mol Cell Cardiol 42: 453–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Schofield RS, Edwards DG, Schuler BT, Estrada J, Aranda JM, Pauly DF et al. (2003). Vascular effects of sildenafil in hypertensive cardiac transplant recipients. Am J Hypertens 16: 874–877. [DOI] [PubMed] [Google Scholar]
  66. Senzaki H, Smith CJ, Juang GJ, Isoda T, Mayer SP, Ohler A et al. (2001). Cardiac phosphodiesterase 5 (cGMP‐specific) modulates beta‐adrenergic signaling in vivo and is down‐regulated in heart failure. FASEB J 15: 1718–1726. [DOI] [PubMed] [Google Scholar]
  67. Southan C, Sharman JL, Benson HE, Faccenda E, Pawson AJ, Alexander SP et al. (2016). The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. Nucl Acids Res 44: D1054–D1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Szabo G, Bahrle S, Stumpf N, Sonnenberg K, Szabo EE, Pacher P et al. (2002). Poly(ADP‐ribose) polymerase inhibition reduces reperfusion injury after heart transplantation. Circ Res 90: 100–106. [DOI] [PubMed] [Google Scholar]
  69. Szabo G, Radovits T, Veres G, Krieger N, Loganathan S, Sandner P et al. (2009). Vardenafil protects against myocardial and endothelial injuries after cardiopulmonary bypass. Eur J Cardiothorac Surg 36: 657–664. [DOI] [PubMed] [Google Scholar]
  70. Takimoto E, Champion HC, Li M, Belardi D, Ren S, Rodriguez ER et al. (2005). Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat Med 11: 214–222. [DOI] [PubMed] [Google Scholar]
  71. Vandeput F, Krall J, Ockaili R, Salloum FN, Florio V, Corbin JD et al. (2009). cGMP‐hydrolytic activity and its inhibition by sildenafil in normal and failing human and mouse myocardium. J Pharmacol Exp Ther 330: 884–891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Varma A, Das A, Hoke NN, Durrant DE, Salloum FN, Kukreja RC (2012). Anti‐inflammatory and cardioprotective effects of tadalafil in diabetic mice. PLoS One 7: e45243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Wallis RM, Corbin JD, Francis SH, Ellis P (1999). Tissue distribution of phosphodiesterase families and the effects of sildenafil on tissue cyclic nucleotides, platelet function, and the contractile responses of trabeculae carneae and aortic rings in vitro. Am J Cardiol 83: 3C–12C. [DOI] [PubMed] [Google Scholar]
  74. Wang X, Fisher PW, Xi L, Kukreja RC (2008). Essential role of mitochondrial Ca2+−activated and ATP‐sensitive K+ channels in sildenafil‐induced late cardioprotection. J Mol Cell Cardiol 44: 105–113. [DOI] [PubMed] [Google Scholar]
  75. Zhuang XD, Long M, Li F, Hu X, Liao XX, Du ZM (2014). PDE5 inhibitor sildenafil in the treatment of heart failure: a meta‐analysis of randomized controlled trials. Int J Cardiol 172: 581–587. [DOI] [PubMed] [Google Scholar]

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

RESOURCES