Skip to main content
Iranian Journal of Public Health logoLink to Iranian Journal of Public Health
. 2023 May;52(5):870–879. doi: 10.18502/ijph.v52i5.12704

Phosphodiesterase-5 Inhibitors as Therapeutics for Cardiovascular Diseases: A Brief Review

Milena Corbic 1,*, Jasmina Sretenovic 2, Vladimir Zivkovic 2,3, Vladimir Jakovljevic 2,4, Tamara Nikolic Turnic 5,6
PMCID: PMC10362213  PMID: 37484720

Abstract

Background:

Three selective and most used inhibitors of PDE-5-sildenafil, vardenafil and tadalafil- have been successfully used for the treatment of erectile dysfunction. Erectile dysfunction and cardiovascular diseases might be considered as two dissimilar clinical signs of the identical systemic disease. PDE-5 inhibitors can through different models and mechanisms induce vasodilation, decrease apoptosis and cell proliferation, and they are widely present in various tissues that make them promising targets in a range of cardiovascular diseases.

Methods:

PubMed was explored to identify papers published from 1990–2019, presenting data for the most used PDE-5 inhibitors (sildenafil, tadalafil or vardenafil) in treatment of cardiovascular diseases.

Results:

This article analyses the therapeutic potentials of PDE-5 inhibitors in cardiovascular diseases and discusses mechanisms, possible risks and limitations. Comparable to earlier studies, newer studies suggest cardioprotective effects of PDE-5 inhibitors, which include different models and mechanisms and do not indicate an increased rate of significant cardiovascular adverse reactions. Dissimilarity in the pharmacokinetics and pharmacodynamics of PDE-5 inhibitors are significant to their risk- benefit profile and clinical use. Some of the studies suggesting infarct size reduction after PDE-5 inhibition described the especially close dose-effect relation, other studies dosage adaptation in drug- drug interactions.

Conclusion:

PDE-5 inhibitors indicate the encouraging useful effects by ischemia/reperfusion injury, myocar-dial infarction, cardiac hypertrophy, cardiomyopathy and systolic and diastolic congestive heart failure. Therefore, this and similar reviews can help for additional clinical targeting in the therapy of cardiovascular diseases.

Keywords: Cardiovascular disease, Cardioprotective effects, Phosphodiesterase-5 inhibitors

Introduction

Phosphodiesterases (PDEs) are metallohydrolases that play the essential role in terminating cyclic nucleotides signaling by causing the hydrolysis of 3′, 5′ cyclic adenosine monophosphate (cAMP) and/or 3′, 5′ cyclic guanosine monophosphate (cGMP), thus modulating the duration and intensity of their biological actions and controlling the intracellular concentrations (1,2). They are classified into 11 families (PDE1–PDE11) according to their sequence of homology, biochemical and pharmacological effects, and the distribution differs among dissimilar tissues (2,3). PDE families vary with respect to their primary structures, tissue distribution, and sensitivity to specific inhibitors, subcellular localization and mechanisms of regulation. There are some PDEs specified only for the hydrolysis of cAMP or cGMP, and others have combined specificity. They are building complexes by connecting to a big number of proteins that connect to each other by protein-protein interactions (4).

The most common selective inhibitors of PDE-5-sildenafil, vardenafil and tadalafil have been used for the treatment of erectile dysfunction (ED), and for the treatment of pulmonary hypertension (PH) (sildenafil and tadalafil). Numerous clinical studies have suggested potential use of PDE-5 inhibitors (PDE-5i), including ischemia/reperfusion injury (IRI), cardiomyopathy, myocardial infarction (MI), cardiac hypertrophy, heart failure (HF), stroke, neurodegenerative diseases and other circulatory disorders (5). Because of the similar risk factors, pathophysiologic pathways and potential mechanisms, patients with cardiovascular diseases (CVD) have frequently also ED (6). Therapy of ED after a first MI was related with a lower mortality and the risk was lower among men who had been treated with PDE-5i, which seemed to be dose-dependent (7). This article reviews the therapeutic potentials of PDE5 inhibitors in different cardiovascular diseases, and discusses mechanisms, possible risks and limitations.

Methods

PubMed was explored to find papers published from 1990–2019 showing relations between 3 most frequent used PDE5i and the CVD. Here are used the following search terms: (phosphodiesterase type 5 inhibitors AND cardiovascular diseases). The search was focused to English language, humans and animals, revealing possible cardioprotective profile of PDE5 inhibitors, considering mechanisms, perspectives, and experimental restrictions of these effects. This article will focus on the 3 most frequent used PDE5i: sildenafil, tadalafil and vardenafil.

Results

Ischemia/Reperfusion Injury and Myocardial Infarction

The myocardial protection from the loss of contractile function among the patients having an acute MI presents a great health challenge. The prognosis depends on it how successful is the reduction of the infarct size (5). IRI is a multi-factorial process what leads to severe tissue destruction, represents a paradoxical phenomenon that results in additional destruction, resulting in dysfunction of the organ, and occurs in previously ischemic tissues after the recovery of blood flow (8). The first that the preconditioning-like effect of sildenafil were demonstrated against myocardial IRI in an in vivo rabbit model by opening of mitochondrial K (ATP) channels (9).

Studies in animal MI demonstrated antiarrhythmic effects of PDE-5i (10,11) as well as recovery of postischemic ventricular contractile function (10,12). In IR of isolated rat hearts, sildenafil pre-treatment at 0.05 mg/kg resulted in improved ventricular recovery, a reduced incidence of ventricular fibrillation (VF) and decreased MI, through increasing cGMP content in the heart. At higher doses, it caused frequently VF and at very low without influence on cardiac function (10).

Sildenafil und vardenafil just before reperfusion have the infarct-limiting effect. They protect the ischemic myocardium against IRI through a mechanism dependent on mitochondrial K (ATP) channel opening (13,14). cGMP-dependent protein kinase (PKG) signaling plays a major role in cardioprotection, and its activation with tadalafil when given 30 min to 120 min before coronary occlusion limits MI and maintains left ventricular function through H (2)S signaling (15). Ischemic preconditioning is first described in experimental preparations in which short episodes of IR applied prior to a longer coronary artery occlusion reduce myocardial infarct size, and it have been presented in many experimental studies (16).

Sildenafil-induced cardioprotection is partly mediated by increased cGMP synthesis and PKG activation, which opens myocardial mitochondrial large-conductance Ca2+-sensitive potassium (mBKCa) channels responsible for myocardial ischemic preconditioning (17).

“Postconditioning” is the adjusted ischemia-reperfusion and represents the rescue of heart tissue applied after a myocardial infarction, through protective interventions with pharmacological stimulus (pharmacological postconditioning) and with repeated short intervals of ischemia (ischemic postconditioning) (18). One further study investigated whether atorvastatin and sildenafil have synergistic effects on myocardial infarct size reduction and increase nitric oxide synthase (NOS) expression. Low-dose atorvastatin in the combination with sildenafil increased the expression of P-eNOS and iNOS and limit MI size, and had a synergistic effect on P-eNOS and iNOS expression and on myocardial protection without leading to serious hypotension or tachycardia (19). Hydrogen sulfide (H2S) is a gaseous molecule that exerts many physiological actions in the cardiovascular system. Similar to PKG, H2S seems to protect the heart via opening of mitoKATP channel (20). PKG activation with tadalafil restricts MI when given 30 min to 120 min before coronary occlusion and preserves left ventricular function through H2S signaling (15). Sildenafil-induced cardioprotection depends on activating of protein kinase C (PKC) and selective translocation of three PKC isoforms (PKC-α, - θ and −δ) (21).

The pathogenesis of IRI includes activation and infiltration of neutrophils and platelets, excessive production of reactive oxygen species and release of proinflammatory and diverse proapoptotic cytokines (2225). All these pathways are damaging mitochondrial- and intracellular calcium homeostasis, causing cardiac myocyte necrosis with additionally damage of contractile function (24) and they happen very early after restoration of flow and they are compatible with the fact that most changes related with cell death, occur during the very first minutes of reflow (26). PDE-5i have been shown to protect the heart against IRI (5,13,27).

In recent years, several studies identified the expression and the induction of nitric oxide synthases (28,29 accumulation of cGMP, activation of kinases with promotion of hydrogen sulfide production (15) and opening of mitochondrial KATP channels as part of the signaling cascade (9). Many reports document the results of animal studies that showed the ability of sildenafil to induce angiogenesis in MI model (9,10,28,30).

Sildenafil being given after early reperfusion has increased the levels of vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang-1) mRNA, which leads to neovascularization. Sildenafil at a specific dose can reduce endothelial cell injury after ischemia and reperfusion, stimulates myocardial angiogenesis and angiogenic gene/protein expression and improve left ventricular contractile functional reserve (31). Another study has suggested the importance of the combined therapy with nitric oxide (NO) inhalation and selective PDE5 inhibition using tadalafil by persistent increase in cGMP. This treatment suggests how important that is for useful long-term structural and functional remodeling (32). Guanylate cyclase can be activated through NO and enlarge the formation of cGMP (10,12) cGMP could activate PKG, and possibly open sarcolemmal (12) and mitochondrial ATP-sensitive K+ (KATP) channels (9,10,12). cGMP and opening of mitochondrial K(ATP) channel play a key role in preconditioning of the heart following ischemia/reperfusion (I/R) injury. Sildenafil and vardenafil protect the ischemic myocardium against reperfusion injury through a pathway that depends on mitochondrial K (ATP) channel opening (14).

Cardiomyopathy

By doxorubicin-induced cardiotoxicity, numerous studies with sildenafil have demonstrated protective effects in vivo treatment of mice before the therapy with doxorubicin, in which sildenafil attenuated myocyte apoptosis and reduced left ventricular dysfunction and frequency of a prolonged ST segment. The sildenafil protective effects were abolished by either L-NAME (an inhibitor of NO synthase) or 5-HD (a blocker of mitochondrial K (ATP)) (33).

cGMP binds and activates cGMP-dependent protein kinase, also known as protein kinase G (PKG). One of the targets of PKG Iα is RhoA (34), and this formation activated by doxorubicin limits its interaction, and suppresses classical cGMP-PKG Iα pro-survival signaling, resulting in intensified apoptosis. PDE5 inhibition can protect from doxorubicin-induced injury by elevating cGMP, and as a result, cGMP connecting to PKG Iα reduces oxidant-induced disulfide formation (35).

Coadministration tadalafil with doxorubicin reduced oxidative stress, upgraded antioxidant capacity by upregulation of mitochondrial superoxide dismutase and prevented the depletion of prosurvival proteins including Bcl-2 and GATA-4 (36,37).

Sildenafil seems to be involved in controlling diabetic cardiomyopathy progression through inter-leukin-8, which is a proinflammatory C-X-C chemokine that plays significant role in inflammation. Sildenafil in diabetic cardiomyopathy seriously reduced Th1 type chemokine CXCL10 level, in blood and in human cardiomyocytes that participates in heart damage initiation and progression (38).

There are evidence that suggest the participation of long non-coding RNAs in cardiovascular illness (39) and the part of long non-coding RNAs in diabetic cardiomyopathy (40).

A bigger expression of the long non-coding RNAs metastasis associated lung adenocarcinoma transcript 1 (MALAT1) has been detected in diabetic cardiomyopathy (41) and Sildenafil can suppress the increase of those levels (42). Recovering the intracellular impact of NO signaling can be possibly relevant to controlling transcription of long non-coding RNAs involved in cardiac injury and in the development of cardiomyopathy related to dysmetabolic conditions including diabetes (42).

Sildenafil being chronic administrated right after permanent occlusion of the left anterior descending coronary artery in mice decreased ischemic cardiomyopathy (43). These cardioprotective effects are mediated by activation of PKG, increased Bcl-2-to-Bax ratio and expression of endothelial and inducible nitric oxide (NO) synthase (eNOS and iNOS, respectively) (27,28,29).

Therapy during three months with 100 mg/day sildenafil in mice with diabetic dilative cardiomyopathy, caused a reduction in circulating monocyte chemoattractant protein (MCP)-1 and tumor growth factor (TGF)-β, without the influence of any other vasodilatory or endothelial effects and as a result improved cardiac geometry and kinetics (44).

Vascular Endothelial Dysfunction

Endothelial dysfunction represents reduction of the bioavailability of vasodilators, particularly NO, and/or an increase in endothelium-derived contracting factors. This lack of balance leads to destruction of endothelium-dependent vasodilation, which is related with many of the ordinary risk factors for CVD and plays a key role in the progress of atherosclerosis and acute coronary syndromes (45, 46).

The endothelial cell dysfunction and cardiac events are well reviewed (47,48), and also the effects of PDE-5i endothelial function have been largely investigated in recent years. The presence of the PDE-5 in the systemic artery endothelium and smooth muscle cells opened a new field of investigations of conditions that affects systemic arteries. Cumulative data from numerous studies with sildenafil have showed partial recovery of endothelial function in experimental models of hypertension and hypertensive subjects. Sildenafil beneficial effects on endothelial and kidney dysfunctions are power to reduce the levels of angiotensin II and increase angiotensin 1–7 and to improve NO bioavailability in angiotensin-dependent hypertension (49).

In a meta-analysis, endothelial dysfunction was the important independent risk factor for MI, stroke, cardiac death, and the need for coronary revascularization (50).

Acute administration of sildenafil 25 and 50 mg increased endothelium-dependent flow-mediated vasodilation in the brachial artery among patients with chronic heart failure (51). Sildenafil was dilated the epicardial coronary arteries, reduced exercise-induced ischemia, improved endothelial dysfunction in the brachial artery, and suppress platelet activation in patients with coronary artery disease (52). In human sildenafil administrated oral cause’s powerful protection against IR-induced endothelial dysfunction through opening of K (ATP) channels (53).

In one study, the effects of sildenafil on vascular disturbances were examined in a mouse model of Angiotensin II-induced hypertension, and sildenafil decreased the damaging effects of Angiotensin II on resistance vessels probably through restoring the balance of ROS/NO/eicosanoids (54).

Cardiac Hypertrophy and Heart Failure

Continual cardiac pressure overload induces pathological ventricular remodeling and hypertrophy that can common lead to HF. Blocking the intrinsic catabolism of cGMP with sildenafil reduces chamber and myocyte hypertrophy, and improves in vivo heart function in mice with chronic exposure to the pressure overload induced by transverse aortic constriction. Takimoto et al have showed how multiple signaling pathways activated by the pressure load are being deactivated by sildenafil, such as calcineurin/NFAT, PI3K/Akt and ERK1/2 (55).

In experimental models, cGK1 activity is being stimulated by the PDE5A inhibition to suppress numerous cardiac signaling pathways involved in HF and pathological hypertrophy (56). That refers to blockade of calcineurin/NFAT signaling (55), its activation of regulators of G-protein signaling (RGS2/4) to block Gq-activated cascades (e.g. from angiotensin or endothelin-1) (57), enhanced mitochondrial and consequent cytoprotection against ischemic injury connected to glycogen synthesis kinase 3-β and mitogen activated kinase ERK1/2 (27), improvement of the cGK1 diastolic function by phosphorylating titin to increase distensibility (58), improvement of proteasome degradation of misfolded proteins (59), inhibition of transient receptor potential canonical ion channel - type 6 (Trpc6) (60) and other mechanisms.

The cardioprotection of sildenafil through NO signaling pathway can be used for slowing the ventricular hypertrophy and remodeling as the indicators of HF (29).

Chronic treatment with sildenafil directly following MI attenuated also ischemic cardiomyopathy and reduced apoptosis in the border zone of the infarcted myocardium (61).

Activation of the sympathetic nervous system in patients with HF, particularly the cardiac sympathetic nerves, has been suggested as an isolated predictor of mortality in HF patients (62). In one study among patients with chronic HF, the acute administration of sildenafil as a bolus dose was connected with a moderate reduction in systemic arterial blood pressure and a more valuable reduction in pulmonary arterial pressure, which resulted in a 20% reduction in cardiac noradrena-line spillover (63).

HF with preserved ejection fraction represents a clinical syndrome of HF with LVEF >50% and frequently has multiple comorbidities, including hypertension, coronary artery disease, atrial fibrillation and diabetes mellitus inducing a systemic proinflammatory state which further induces coronary microvascular endothelial inflammation. It reduces NO bioavailability and PKG activity that leads to hypertrophy development. Stiffness of myocyte and extracellular matrix components of ventricular muscle contribute to high diastolic left ventricular stiffness and HF development (64).

The prognosis of HF with preserved ejection fraction is getting worse with pulmonary hypertension (PH) and right ventricular failure development. Patients had pulmonary artery pressure (PAP) >40 mm Hg (right heart catheterisation and right ventricular systolic dysfunction, and PDE5i largely improved PAP and right ventricular function (65).

Studies with animal models support a cardioprotective effects of PDE5is, as well as improving contractile function in HF with reduced ejection fraction (HFrEF) (29,66) and regressing left ventricular hypertrophy (55,61).

The effects of PDE5i against inflammation und apoptosis have been demonstrated among the studies in animal models of left ventricular dysfunction (67). Sildenafil seriously decreases left ventricular remodeling and has the prophylactic effect by exercise intolerance in rats of chronic mitral regurgitation by reducing perivascular fibrosis, apoptosis and hypertrophy (68).

PDE-5i improve diastolic function in animal models of systolic HF and clinical studies of HFrEF by decreasing hypertrophy, fibrosis and improving chamber compliance. In the hypertensive heart after chronic renin-angiotensin aldosterone system stimulation and angiotension II-induced HF, sildenafil increased diastolic and systolic performance, and reduced left ventricular hypertrophy (67).

PDE5i suppress beta-adrenergic receptor (β-AR) stimulation in left ventricular myocytes via reduced myofilament Ca sensitivity through PKG-phosphorylation of cardiac Troponin I (69).

Sildenafil improves the ability to exercise, in patients with systolic HF with secondary PH, decreases pulmonary arterial pressure contributing to decreased pulmonary vascular resistance (70).

Possible Risks and Caution by Using Pde5

The patients receiving nitrate therapy need to be considered with attention and separately, because PDE-5i increased the hypotensive effects of acute and chronic nitrates (71). PDE-5i and glyceryl nitrate, isosorbide salts, sodium nitroprus-side, amyl nitrite, nicorandil, and organic nitrates should not be used simultaneously (72).

The therapy of ED should be applied only to patients with low-risk of cardiac events, because sexual activity and treatment of ED may cause cardiac events in patients with preexisting CVD. On the other hand, men with high risk of CVD should receive cardiovascular risk assessment before attempting sexual activity and receiving ED treatment (73).

The early predominating safety concern refers to potential risk of arrhythmia, especially while many cases of unpredicted death have been described with sildenafil in patients predisposed to ischemic cardiac events. The patients that have limited cardiac repolarization reserve can be prone to arrhythmia if treated with medications that prolong ventricular repolarization. Sildenafil at therapeutic concentrations does not prolong cardiac repolarization (74) and do not modify action potential duration or QTc (75).

Even though many studies have not shown a significant distinction in the risk of severe cardiovascular events in patients treated with PDE5 inhibitors and placebo, the risks and benefits need to be valued on an individual basis.

Discussion

The reviewed studies, mostly among studies with animals, suggest cardioprotective effects of PDE-5i with various models and mechanisms. Variations in the pharmacokinetics and pharmacodynamics of PDE-5i are significant to their risk-benefit profile and medical use. Some of the studies examining the reduction of the infarct size after PDE-5 inhibition described a particularly close dose-effect relation, other studies dosage adaptations in drug- drug interactions and special populations based on the specific demands of the patients. The progress in developing of new PDE-5I with improved selectivity, faster onset of action, increased potency, less side effects and improved tolerability should be the aim. Further researches should probably compare PDE-5i together to determine the patient groups in which the drug would be most effective.

Conclusion

The future carefully designed clinical trials should consequently lead to new applications of PDE-5i and give encouraging results with the perspective of clinical therapeutic strategies to upgrade symptoms and prognosis in CVD.

Journalism Ethics considerations

Ethical issues (Including plagiarism, informed consent, misconduct, data fabrication and/or falsification, double publication and/or submission, redundancy, etc.) have been completely observed by the authors.

Acknowledgements

No funding.

Footnotes

Conflict of interest

The authors declare that there is no conflict of interest.

References

  • 1.Beavo JA. (1995). Cyclic nucleotide phosphodiesterases: functional implications of multiple isoforms. Physiol Rev, 75(4):725–48. [DOI] [PubMed] [Google Scholar]
  • 2.Bender AT, Beavo JA. (2006). Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev, 58:488–520. [DOI] [PubMed] [Google Scholar]
  • 3.Corbin JD, Francis SH. (2002). Pharmacology of phosphodiesterase-5 inhibitors. Int J Clin Pract, 56(6):453–9. [PubMed] [Google Scholar]
  • 4.Al-Nema MY, Gaurav A. (2019). Protein-Protein Interactions of Phosphodiesterases. Curr Top Med Chem, 19(7):555–564. [DOI] [PubMed] [Google Scholar]
  • 5.Kukreja RC, Salloum FN, et al. (2011). Emerging new uses of phosphodiesterase-5 inhibitors in cardiovascular diseases. Exp Clin Cardiol, 16(4): e30–5. [PMC free article] [PubMed] [Google Scholar]
  • 6.Cai Z, Zhang J, Li H. (2019). Two Birds with One Stone: Regular Use of PDE5 Inhibitors for Treating Male Patients with Erectile Dysfunction and Cardiovascular Diseases. Cardiovasc Drugs Ther, 33(1):119–128. [DOI] [PubMed] [Google Scholar]
  • 7.Andersson DP, Trolle Lagerros Y, Grotta A, et al. (2017). Association between treatment for erectile dysfunction and death or cardiovascularoutcomes after myocardial infarction. Heart, 103(16):1264–1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yellon DM, Hausenloy DJ. (2007). Myocardial reperfusion injury. N Engl J Med, 357:1121–1135. [DOI] [PubMed] [Google Scholar]
  • 9.Ockaili R, Salloum F, Hawkins J, et al. (2002). Sildenafil (Viagra) induces powerful cardioprotective effect via opening of mitochondrial K(ATP) channels in rabbits. Am J Physiol Heart Circ Physiol, 283(3):H1263–9. [DOI] [PubMed] [Google Scholar]
  • 10.Das S, Maulik N, Das DK, et al. (2002). Cardioprotection with sildenafil, a selective inhibitor of cyclic 3′,5′-monophosphate-specific phosphodiesterase 5. Drugs Exp Clin Res, 28(6):213–9. [PubMed] [Google Scholar]
  • 11.Nagy O, Hajnal A, Parratt JR, et al. (2004). Sildenafil (Viagra) reduces arrhythmia severity during ischaemia 24 h after oral administration in dogs. Br J Pharmacol, 141(4):549–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.du Toit EF, Rossouw E, Salie R, et al. (2005). Effect of sildenafil on reperfusion function, infarct size, and cyclic nucleotide levels in the isolated rat heart model. Cardiovasc Drugs Ther, 19(1):23–31. [DOI] [PubMed] [Google Scholar]
  • 13.Elrod JW, Greer JJ, Lefer DJ. (2007). Sildenafil-mediated acute cardioprotection is independent of the NO/cGMP pathway. Am J Physiol Heart Circ Physiol, 292(1):H342–7. [DOI] [PubMed] [Google Scholar]
  • 14.Salloum FN, Takenoshita Y, Ockaili RA, 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(2): 453–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Salloum FN, Chau VQ, Hoke NN, et al. (2009). Phosphodiesterase-5 inhibitor, tadalafil, protects against myocardial ischemia/reperfusion through protein-kinase G dependent generation of hydrogen sulfide. Circulation, 120(110): S31–S36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kloner RA, Rezkalla SH. (2006). Preconditioning, postconditioning and their application to clinical cardiology. Cardiovasc Res, 70(2):297–307. [DOI] [PubMed] [Google Scholar]
  • 17.Behmenburg F, Dorsch M, Huhn R, et al. (2015). Impact of Mitochondrial Ca2+-Sensitive Potassium (mBKCa) Channels in Sildenafil-Induced Cardioprotection in rats. PLoS One, 10(12): e0144737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Skyschally A, van CP, Iliodromitis EK, et al. (2009). Ischemic postconditioning: experimental models and protocol algorithms. Basic Res Cardiol, 104(5):469–83. [DOI] [PubMed] [Google Scholar]
  • 19.Rosanio S, Ye Y, Atar S, et al. (2006). Enhanced cardioprotection against ischemia-reperfusion injury with combining sildenafil with low-dose atorvastatin. Cardiovasc Drugs Ther, 20(1):27–36. [DOI] [PubMed] [Google Scholar]
  • 20.Elrod JW, Calvert JW, Morrison J, et al. (2007). Hydrogen sulfide attenuates myocardial ischemia-reperfusion injury by preservation of mitochondrial function. Proc Natl Acad Sci USA, 104(39):15560–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Das A, Ockaili R, Salloum F, et al. (2004). Protein kinase C plays an essential role in sildenafil-induced cardioprotection in rabbits. Am J Physiol Heart Circ Physiol, 286(4):H1455–60. [DOI] [PubMed] [Google Scholar]
  • 22.Carden DL, Granger DN. (2000). Pathophysiology of ischaemia-reperfusion injury. J Pathol, 190(3):255–266. [DOI] [PubMed] [Google Scholar]
  • 23.Vinten-Johansen J. (2004). Involvement of neutrophils in the pathogenesis of lethal myocardial reperfusion injury. Cardiovasc Res, 61(3):481–97. [DOI] [PubMed] [Google Scholar]
  • 24.Heusch G, Boengler K, Schulz R. (2008). Cardioprotection: nitric oxide, protein kinases, and mitochondria. Circulation, 118(19):1915–9. [DOI] [PubMed] [Google Scholar]
  • 25.McAlindon E, Bucciarelli-Ducci C, Suleiman MS, et al. (2015). Infarct size reduction in acute myocardial infarction. Heart, 101:155–160. [DOI] [PubMed] [Google Scholar]
  • 26.Halestrap AP, Clarke SJ, Javadov SA. (2004). Mitochondrial permeability transition pore opening during myocardial reperfusion—a target for cardioprotection. Cardiovasc Res, 61(3):372–85. [DOI] [PubMed] [Google Scholar]
  • 27.Das A, Xi L, Kukreja RC. (2008). Protein kinase G-dependent cardioprotective mechanism of phosphodiesterase-5 inhibition involves phosphorylation of ERK and GSK3b. J Biol Chem, 283(43):29572–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Salloum F, Yin C, Xi L, et al. (2003). Sildenafil induces delayed preconditioning through inducible nitric oxide synthase-dependent pathway in mouse heart. Circ Res, 92(6):595–7. [DOI] [PubMed] [Google Scholar]
  • 29.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(13):12944–55. [DOI] [PubMed] [Google Scholar]
  • 30.Kukreja RC, Salloum F, Das A, et al. (2005). Pharmacological preconditioning with sildenafil: basic mechanisms and clinical implications. Vascul Pharmacol, 42(5–6):219–32. [DOI] [PubMed] [Google Scholar]
  • 31.Koneru S, Varma Penumathsa S, Thirunavukkarasu M, et al. (2008). Sildenafil-mediated neovascularization and protection against myocardial ischaemiareperfusion injury in rats: role of VEGF/angiopoietin-1. J Cell Mol Med, 12(6b): 2651–2664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lux A, Pokreisz P, Swinnen M, et al. (2016). Concomitant Phosphodiesterase 5 Inhibition Enhances Myocardial Protection by Inhaled Nitric Oxide in Ischemia-Reperfusion Injury. J Pharmacol Exp Ther, 356(2):284–92. [DOI] [PubMed] [Google Scholar]
  • 33.Fisher PW, Salloum F, Das A, et al. (2005). Phosphodiesterase-5 inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular dysfunction in a chronic model of doxorubicin cardiotoxicity. Circulation, 111(13):1601–10. [DOI] [PubMed] [Google Scholar]
  • 34.Li F, Jiang Q, Shi KJ, et al. (2013). RhoA modulates functional and physical interaction between ROCK1 and Erk1/2 in selenite-induced apoptosis of leukaemia cells. Cell Death Dis, 4(7):e708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Prysyazhna O, Burgoyne JR, Scotcher J, et al. (2016). Phosphodiesterase 5 Inhibition Limits Doxorubicin-induced Heart Failure by Attenuating Protein Kinase G Iα Oxidation. J Biol Chem, 291(33):17427–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Koka S, Das A, Zhu SG, et al. (2010). Long-acting phosphodiesterase-5 inhibitor tadalafil attenuates doxorubicin-induced cardiomyopathy without interfering with chemotherapeutic effect. J Pharmacol Exp Ther, 334(3):1023–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Koka S, Kukreja RC. (2010). Attenuation of doxorubicin-induced cardiotoxicity by tadalafil: A long acting phosphodiesterase-5 inhibitor. Mol Cell Pharmacol, 2(5):173–178. [PMC free article] [PubMed] [Google Scholar]
  • 38.Di Luigi L, Corinaldesi C, Colletti M, et al. (2016). Phosphodiesterase Type 5 inhibitor sildenafil decreases the proinflammatory chemokine CXCL10 in human cardiomyocytes and in subjects with diabetic cardiomyopathy. Inflammation, 39(3):1238–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Greco S, Salgado Somoza A, Devaux Y, et al. (2017). Long noncoding RNAs and cardiac disease. Antioxid Redox Signal, 29(9):880–901. [DOI] [PubMed] [Google Scholar]
  • 40.Zhou Xiang, Zhang Wei, Jin Mengchao, et al. (2017). lncRNA MIAT functions as a competing endogenous RNA to upregulate DAPK2 by sponging miR-22-3p in diabetic cardiomyopathy. Cell Death & Disease, 8(7): e2929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Zhang Mengyao, Gu Huimin, Xu Weiting, et al. (2016). Down-regulation of lncRNA MALAT1 reduces cardiomyocyte apoptosis and improves left ventricular function in diabetic rats. Int J Cardiol, 203:214–6. [DOI] [PubMed] [Google Scholar]
  • 42.Bacci L, Barbati SA, Colussi C, et al. (2018). Sildenafil normalizes MALAT1 level in diabetic cardiomyopathy. Endocrine, 62(1):259–262. [DOI] [PubMed] [Google Scholar]
  • 43.Wang X, Fisher P, Xi L, et al. (2008). Activation of mitochondrial calcium-activated and ATP-sensitive potassium channels is essential for sildenafil-induced cardioprotection. J Mol Cell Cardiol, 44:105–13. [DOI] [PubMed] [Google Scholar]
  • 44.Giannetta E, Isidori AM, Galea N, 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(19):2323–33. [DOI] [PubMed] [Google Scholar]
  • 45.Hadi HA, Carr CS, Al Suwaidi J. (2005). Endothelial dysfunction: cardiovascular risk factors, therapy, and outcome. Vasc Health Risk Manag, 1(3):183–98. [PMC free article] [PubMed] [Google Scholar]
  • 46.Cayatte AJ, Palacino JJ, Horten K, et al. (1994). Chronic inhibition of nitric oxide production accelerates neointima formation and impairs endothelial function in hypercholesterolemic rabbits. Arterioscler Thromb, 14(5):753–9. [DOI] [PubMed] [Google Scholar]
  • 47.Widlansky ME, Gokce N, Keaney JF, Jr, et al. (2003). The clinical implications of endothelial dysfunction. J Am Coll Cardiol, 42(7):1149–60. [DOI] [PubMed] [Google Scholar]
  • 48.Ganz P, Vita JA. (2003). Testing endothelial vasomotor function: nitric oxide, a multipotent molecule. Circulation, 108(17):2049–53. [DOI] [PubMed] [Google Scholar]
  • 49.Vasquez EC, Gava AL, Graceli JB, et al. (2016). Novel Therapeutic Targets for Phosphodiesterase 5 Inhibitors: current state-of-theart on systemic arterial hypertension and atherosclerosis. Curr Pharm Biotechnol, 17(4):347–64. [DOI] [PubMed] [Google Scholar]
  • 50.Lerman A, Zeiher AM. (2005). Endothelial function: cardiac events. Circulation, 111(3):363–8. [DOI] [PubMed] [Google Scholar]
  • 51.Katz SD, Balidemaj K, Homma S, et al. (2000). Acute type 5 phosphodiesterase inhibition with sildenafil enhances flow-mediated vasodilation in patients with chronic heart failure. J Am Coll Cardiol, 36(3):845–51. [DOI] [PubMed] [Google Scholar]
  • 52.Halcox JP, Nour KR, Zalos G, et al. (2002). The effect of sildenafil on human vascular function, platelet activation, and myocardial ischemia. J Am Coll Cardiol, 40(7):1232–40. [DOI] [PubMed] [Google Scholar]
  • 53.Gori T, Sicuro S, Dragoni S, et al. (2005). Sildenafil prevents endothelial dysfunction induced by ischemia and reperfusion via opening of adenosine triphosphate-sensitive potassium channels: A human in vivo study. Circulation, 111(6):742–6. [DOI] [PubMed] [Google Scholar]
  • 54.Dias AT, Leal MAS, Zanardo TC, et al. (2018). Beneficial Morphofunctional Changes Promoted by Sildenafil in Resistance Vessels in the Angiotensin II-Induced Hypertension Model. Curr Pharm Biotechnol, 19(6):483–494. [DOI] [PubMed] [Google Scholar]
  • 55.Takimoto E, Champion HC, Li M, et al. (2005). Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat Med, 11(2):214–22. [DOI] [PubMed] [Google Scholar]
  • 56.Kim GE, Kass DA. (2017). Cardiac Phosphodiesterases and Their Modulation for Treating Heart Disease. Handb Exp Pharmacol, 243:249–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Takimoto E, Koitabashi N, Hsu S, et al. (2009). Regulator of G protein signaling 2 mediates cardiac compensation to pressure overload and antihypertrophic effects of PDE5 inhibition in mice. J Clin Invest, 119(2):408–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bishu K, Hamdani N, Mohammed SF, et al. (2011). Sildenafil and B-type natriuretic peptide acutely phosphorylate titin and improve diastolic distensibility in vivo. Circulation, 124(25):2882–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Seo K, Rainer PP, Lee DI, et al. (2014). Hyperactive Adverse Mechanical Stress Responses in Dystrophic Heart Are Coupled to Transient Receptor Potential Canonical 6 and blocked by cGMP-Protein Kinase G Modulation. Circ Res, 114:823–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ranek MJ, Terpstra EJ, Li J, et al. (2013). Protein kinase g positively regulates proteasome-mediated degradation of misfolded proteins. Circulation, 128(4): 365–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Salloum FN, Abbate A, Das A, et al. (2008). Sildenafil (Viagra) attenuates ischemic cardiomyopathy and improves left ventricular function in mice. Am J Physiol Heart Circ Physiol, 294(3):H1398–406. [DOI] [PubMed] [Google Scholar]
  • 62.Kaye DM, Lefkovits J, Jennings GL, et al. (1995). Adverse consequences of high sympathetic nervous activity in the failing human heart. J Am Coll Cardiol, 26(5):1257–63. [DOI] [PubMed] [Google Scholar]
  • 63.Al Hesayen A, Floras JS, Parker JD. (2006). The effects of intravenous sildenafil on hemodynamics and cardiac sympathetic activity in chronic human heart failure. Eur J Heart Fail, 8(8):864–8. [DOI] [PubMed] [Google Scholar]
  • 64.Paulus WJ, Tschöpe C. (2013). A novel paradigm for heart failure with preserved ejection fraction: comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J Am Coll Cardiol, 62(4):263–71. [DOI] [PubMed] [Google Scholar]
  • 65.Guazzi M, Vicenzi M, Arena R, et al. (2011). Pulmonary hypertension in heart failure with preserved ejection fraction: a target of phosphodiesterase-5 inhibition in a 1-year study. Circulation, 124:164–74. [DOI] [PubMed] [Google Scholar]
  • 66.Senzaki H, Smith CJ, Juang GJ, et al. (2001). Cardiac phosphodiesterase 5 (cGMP-specific) modulates beta-adrenergic signaling in vivo and is down-regulated in heart failure. FASEB J, 15:1718–26. [DOI] [PubMed] [Google Scholar]
  • 67.Westermann D, Becher PM, Lindner D, et al. (2012). Selective PDE5A inhibition with sildenafil rescues left ventricular dysfunction, inflammatory immune response and cardiac remodeling in angiotensin II-induced heart failure in vivo. Basic Res Cardiol, 107(6):308. [DOI] [PubMed] [Google Scholar]
  • 68.Kim KH, Kim YJ, Ohn JH, 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(11):1390–401. [DOI] [PubMed] [Google Scholar]
  • 69.Lee DI, Vahebi S, Tocchetti CG, et al. (2010). PDE5A suppression of acute beta-adrenergic activation requires modulation of myocyte beta-3 signaling coupled to PKG-mediated troponin I phosphorylation. Basic Res Cardiol, 105(3):337–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lewis GD, Lachmann J, Camuso J, et al. (2007). Sildenafil improves exercise hemodynamics and oxygen uptake in patients with systolic heart failure. Circulation, 115:59–66. [DOI] [PubMed] [Google Scholar]
  • 71.Gandaglia G, Briganti A, Montorsi P, et al. (2016). Diagnostic and Therapeutic Implications of Erectile Dysfunction in Patients with Cardiovascular Disease. Eur Urol, 70(2):219–22. [DOI] [PubMed] [Google Scholar]
  • 72.Rezvanfar MA, Rahimi HR, Abdollahi M. (2012). ADMET considerations for phosphodiesterase-5 inhibitors. Expert Opin Drug Metab Toxicol, 8(10):1231–45. [DOI] [PubMed] [Google Scholar]
  • 73.D'Andrea S, Barbonetti A, Martorella A, et al. (2019). Effect of prolonged treatment with phosphodiesterase-5-inhibitors on endothelial dysfunction in vascular diseases and vascular risk conditions: A systematic review analysis and meta-analysis of randomized double-blind placebo-controlled trials. Int J Clin Pract, 73(2): e13296. [DOI] [PubMed] [Google Scholar]
  • 74.Chiang CE, Luk HN, Wang TM, et al. (2002). Effects of sildenafil on cardiac repolarization. Cardiovasc Res, 55(2):290–9. [DOI] [PubMed] [Google Scholar]
  • 75.Dustan Sarazan R, Crumb WJ, Beasley CM, et al. (2004). Absence of clinically important HERG channel blockade by three compounds that inhibit phosphodiesterase 5--sildenafil, tadalafil, and vardenafil. Eur J Pharmacol, 502(3):163–7. [DOI] [PubMed] [Google Scholar]

Articles from Iranian Journal of Public Health are provided here courtesy of Tehran University of Medical Sciences

RESOURCES