Associate editor: M. Madhani
Cyclic GMP signaling in cardiovascular pathophysiology and therapeutics

https://doi.org/10.1016/j.pharmthera.2009.02.009Get rights and content

Abstract

Cyclic guanosine 3',5'-monophosphate (cGMP) mediates a wide spectrum of physiologic processes in multiple cell types within the cardiovascular system. Dysfunctional signaling at any step of the cascade — cGMP synthesis, effector activation, or catabolism — have been implicated in numerous cardiovascular diseases, ranging from hypertension to atherosclerosis to cardiac hypertrophy and heart failure. In this review, we outline each step of the cGMP signaling cascade and discuss its regulation and physiologic effects within the cardiovascular system. In addition, we illustrate how cGMP signaling becomes dysregulated in specific cardiovascular disease states. The ubiquitous role cGMP plays in cardiac physiology and pathophysiology presents great opportunities for pharmacologic modulation of the cGMP signal in the treatment of cardiovascular diseases. We detail the various therapeutic interventional strategies that have been developed or are in development, summarizing relevant preclinical and clinical studies.

Introduction

Cyclic guanosine 3',5'-monophosphate (cGMP) is a ubiquitous intracellular second-messenger that mediates a vast array of physiologic processes, from ion channel conductance to cell growth and apoptosis to cellular mobility and contractility. In the cardiovascular system, cGMP signaling is vital to endothelial, vascular smooth muscle, and cardiac myocyte function. Generated by guanylyl cyclase isoforms in response to natriuretic peptides (NPs) and nitric oxide (NO), cGMP exerts its actions through cGMP-gated cation channels, cGMP-dependent protein kinases (PKGs), and cGMP-regulated phosphodiesterases (PDEs) that in turn hydrolyze cyclic nucleotides. Since the discovery of cGMP in rat urine nearly 50 years ago (Ashman et. al, 1963), the field of cGMP signaling research has grown exponentially. Abnormalities at each step of the cGMP signaling cascade, from cGMP synthesis to its degradation, have been implicated in cardiovascular disease and thus represent potential targets for pharmacologic therapies.

cGMP has two distinct pathways that regulate its synthesis, one coupled to natriuretic peptide hormone, and the other a simple gas (nitric oxide) (Fig. 1). No other second messenger, not even cyclic adenosine monophosphate (cAMP), is activated by a gas. The significance of NO-cGMP signaling was recognized by the 1998 Nobel Prize in Physiology and Medicine that was awarded for the major discoveries surrounding nitric oxide (Arnold et al., 1977, Katsuki et al., 1977, Schultz et al., 1977, Ignarro et al., 1987a, Ignarro et al., 1987b). Natriuretic peptide-mediated cGMP signaling was discovered in the early 1980s, when a polypeptide hormone was isolated from heart atrial muscle tissue and found to have potent diuretic (natriuretic) and hypotensive properties (de Bold, 1982, Ackermann et al., 1984, Atarashi et al., 1984, Atlas et al., 1984, Bloch et al., 1985, de Bold, 1985). The discovery of atrial natriuretic peptide was momentous in its implication of the heart as more than a circulatory pump or electrically conductive tissue but also an endocrine organ; a finding which ultimately helped shift the conceptual paradigm of heart failure to the current neurohormonal model.

Each of these pathways couples to a distinct guanylyl cyclase isoform — soluble (sGC) and particulate (pGC) guanylyl cyclase respectively. These cyclases differ in their intracellular distribution, with sGC historically described as a cytosolic protein and pGC being a membrane bound protein. However, their intracellular localization is more nuanced, and recent studies support distinct pools of cGMP generation with different downstream effects (Castro et al., 2006, Takimoto et al., 2007, Nausch et al., 2008). Cyclic GMP activates three types of effector molecules, with cGMP-dependent protein kinases (PKGs) and phosphodiesterases (PDEs) predominating in the cardiovascular system. A third type of effector molecule, cGMP-gated cation channels, exists in retinal and olfactory neuroepithelium and nephrons, but neither protein expression nor physiological function of these channels have been established in the cardiovascular system. Cyclic GMP-PKG signaling within the vascular endothelium stimulates cell proliferation and increases permeability (Draijer et al., 1995a, Draijer et al., 1995b, Vaandrager and de Jonge, 1996, Holschermann et al., 1997, Hood and Granger, 1998, Smolenski et al., 2000, Kook et al., 2003); it inhibits cell proliferation and mediates vasorelaxation in vascular smooth muscle (Archer et al., 1994, Bolotina et al., 1994, Cornwell et al., 1994, Murad et al., 1985); while in cardiac myocardium, it inhibits hypertrophy and modulates contractility (Lohmann et al., 1991, Shah et al., 1994, Takimoto et al., 2005b, Kinugawa et al., 1997, Vila-Petroff et al., 1999, Tatsumi et al., 2000). In all three tissues, cGMP-PKG signaling also mediates cellular apoptosis (Fukuo et al., 1996, Wu et al., 1997, DeMeester et al., 1998, Arstall et al., 1999, Suenobu et al., 1999, Taimor et al., 2000).

Lastly, cGMP catabolism is regulated by a subgroup of the 11 member phospho-diesterase superfamily. PDEs play a role in not only spatiotemporal regulation of cGMP signal but also cross-regulation of the cAMP signal. The strategy of inhibiting PDEs to enhance cGMP and related signaling has already been harnessed with the PDE5A inhibitor sildenafil, a common treatment for erectile dysfunction (Boolell et al., 1996). PDE inhibition has been further examined for the treatment of a variety of cardiovascular diseases, including pulmonary hypertension and now chronic heart failure, and this continues to be a highly active and promising field of research (Bethke et al., 1992a, Bethke et al., 1992b, Eddahibi et al., 1998, Gillies et al., 2002, Reffelmann and Kloner, 2003, Guazzi and Samaja, 2007, Lewis et al., 2007a, Attina et al., 2008, Baliga et al., 2008, Park, 2008).

This review summarizes our current understanding of cGMP signaling within the cardiovascular system, specifically in vascular endothelial and smooth muscle cells and cardiac myocytes. Several others have reviewed specific aspects of nitric oxide and natriuretic peptide, PKG, and PDE5 signaling (Kass et al., 2007a, Hofmann et al., 2009, Gillies et al., 2002, Saraiva and Hare, 2006, Rastaldo et al., 2007, Schulz et al., 2008, Woodard and Rosado, 2008). After discussing the major elements of the cGMP signaling pathway, we focus on the role of dysfunctional cGMP signaling in cardiovascular disease and the potential targets that this poses for the pharmacological treatment of cardiovascular diseases.

Section snippets

Generation by guanylyl cyclases

The biosynthesis of cyclic GMP from guanosine triphosphate (GTP) is catalyzed by two different isoforms of guanylyl cyclase, one which functions as the biosensor for nitric oxide and the other, as the plasma membrane receptor for natriuretic peptides.

Vascular smooth muscle cells

cGMP signaling directs vascular tone and smooth muscle cell (SMC) proliferation and differentiation (Fig. 3) (Murad et al., 1985, Munzel et al., 2003). Vascular tone is regulated by changes in intracellular free calcium concentrations within SMCs. In general, SMC contraction is triggered by the receptor-mediated generation of the second-messenger inositol 1,4,5-trisphosphate (IP3). IP3 induces release of free calcium from intracellular stores, further provoking influx of extracellular calcium

Pathophysiological role of cGMP signaling in cardiovascular disease

With cGMP signaling being as vital as it is to the physiologic functions of the heart and vasculature, it comes as no surprise that dysfunction at any level of the cGMP signaling pathway is a factor in many cardiovascular diseases. Endothelial cell dysfunction contributes to hypertensive disease, both systemic and pulmonary, and atherosclerosis; vascular smooth muscle dysfunction, systemic and pulmonary hypertensive and ischemic heart disease; and cardiac myocyte dysfunction, hypertrophic and

Pharmacologic modulation of cGMP signaling in cardiovascular disease

Given that cGMP signaling is dysregulated in a wide spectrum of cardiovascular diseases and can be altered at multiple levels, from upstream triggering events to downstream effectors and regulatory molecules, each step of the cascade becomes a promising target for pharmacologic therapy (Table 1).

Conclusions and perspectives

The central role of cGMP in mediating various physiological processes within the cardiovascular system presents this second messenger as a prime target for pharmacologic modulation. Many different approaches have been undertaken to enhance the cGMP signal. Supplementation with essential co-factors for eNOS can restore the bioavailability of NO in diseased states. NO-donor drugs and synthetic chimeric natriuretic peptides can trigger cGMP production through their respective guanylyl cyclases.

References (419)

  • ButtE. et al.

    Endothelial nitric-oxide synthase (type III) is activated and becomes calcium independent upon phosphorylation by cyclic nucleotide-dependent protein kinases

    J Biol Chem

    (2000)
  • ChenH. et al.

    Atrial natriuretic peptide-initiated cGMP pathways regulate vasodilator-stimulated phosphoprotein phosphorylation and angiogenesis in vascular endothelium

    J Biol Chem

    (2008)
  • ChicheJ.D. et al.

    Adenovirus-mediated gene transfer of cGMP-dependent protein kinase increases the sensitivity of cultured vascular smooth muscle cells to the antiproliferative and pro-apoptotic effects of nitric oxide/cGMP

    J Biol Chem

    (1998)
  • CrabosM. et al.

    Reduced basal NO-mediated dilation and decreased endothelial NO-synthase expression in coronary vessels of spontaneously hypertensive rats

    J Mol Cell Cardiol

    (1997)
  • D'SouzaS.P. et al.

    Autocrine and paracrine actions of natriuretic peptides in the heart

    Pharmacol Ther

    (2004)
  • DasA. et al.

    Cyclic GMP-dependent protein kinase Ialpha attenuates necrosis and apoptosis following ischemia/reoxygenation in adult cardiomyocyte

    J Biol Chem

    (2006)
  • DasA. et al.

    Protein kinase G dependent cardioprotective mechanism of phosphodiesterase-5 inhibition involves phosphorylation of ERK and GSK3beta

    J Biol Chem

    (2008)
  • DeverG.J. et al.

    Phospholipid chlorohydrin induces leukocyte adhesion to ApoE−/− mouse arteries via upregulation of P-selectin

    Free Radic Biol Med

    (2008)
  • DrexlerH. et al.

    Expression, activity and functional significance of inducible nitric oxide synthase in the failing human heart

    J Am Coll Cardiol

    (1998)
  • FischmeisterR. et al.

    Species- and tissue-dependent effects of NO and cyclic GMP on cardiac ion channels

    Comp Biochem Physiol A Mol Integr Physiol

    (2005)
  • AckermannU. et al.

    Cardiovascular effects of atrial extracts in anesthetized rats

    Can J Physiol Pharmacol

    (1984)
  • AicherA. et al.

    Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells

    Nat Med

    (2003)
  • AldertonW.K. et al.

    Nitric oxide synthases: structure, function and inhibition

    Biochem J

    (2001)
  • AndreassiM.G. et al.

    Up-regulation of ‘clearance’ receptors in patients with chronic heart failure: a possible explanation for the resistance to biological effects of cardiac natriuretic hormones

    Eur J Heart Fail

    (2001)
  • ArcherS.L. et al.

    Nitric oxide and cGMP cause vasorelaxation by activation of a charybdotoxin-sensitive K channel by cGMP-dependent protein kinase

    Proc Natl Acad Sci U S A

    (1994)
  • ArnoldW.P. et al.

    Nitric oxide activates guanylate cyclase and increases guanosine 3':5'-cyclic monophosphate levels in various tissue preparations

    Proc Natl Acad Sci U S A

    (1977)
  • ArstallM.A. et al.

    Cytokine-mediated apoptosis in cardiac myocytes: the role of inducible nitric oxide synthase induction and peroxynitrite generation

    Circ Res

    (1999)
  • Ashman, D.F., Lipton, R., Melicow, M.M., Price,T.D. (1963). Isolation of adenosine 3', 5'-monophosphate and guanosine...
  • AtarashiK. et al.

    Inhibition of aldosterone production by an atrial extract

    Science

    (1984)
  • AtlasS.A. et al.

    Purification, sequencing and synthesis of natriuretic and vasoactive rat atrial peptide

    Nature

    (1984)
  • AttinaT.M. et al.

    Phosphodiesterase type 5 inhibition reverses impaired forearm exercise-induced vasodilatation in hypertensive patients

    J Hypertens

    (2008)
  • BaligaR.S. et al.

    Synergy between natriuretic peptides and phosphodiesterase 5 inhibitors ameliorates pulmonary arterial hypertension

    Am J Respir Crit Care Med

    (2008)
  • BalligandJ.L. et al.

    Nitric oxide synthases and cardiac muscle. Autocrine and paracrine influences

    Arterioscler Thromb Vasc Biol

    (1997)
  • BalligandJ.L. et al.

    Induction of NO synthase in rat cardiac microvascular endothelial cells by IL-1 beta and IFN-gamma

    Am J Physiol

    (1995)
  • BauersachsJ. et al.

    Vasodilator dysfunction in aged spontaneously hypertensive rats: changes in NO synthase III and soluble guanylyl cyclase expression, and in superoxide anion production

    Cardiovasc Res

    (1998)
  • BendayanD. et al.

    Combination therapy with prostacyclin and tadalafil for severe pulmonary arterial hypertension: a pilot study

    Respirology

    (2008)
  • BenderA.T. et al.

    Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use

    Pharmacol Rev

    (2006)
  • BenderA.T. et al.

    Selective up-regulation of PDE1B2 upon monocyte-to-macrophage differentiation

    Proc Natl Acad Sci U S A

    (2005)
  • BethkeT. et al.

    Phosphodiesterase inhibition by enoximone in preparations from nonfailing and failing human hearts

    Arzneimittelforschung

    (1992)
  • BethkeT. et al.

    Phosphodiesterase inhibition in ventricular cardiomyocytes from guinea-pig hearts

    Br J Pharmacol

    (1992)
  • BirukovaA.A. et al.

    Epac/Rap and PKA are novel mechanisms of ANP-induced Rac-mediated pulmonary endothelial barrier protection

    J Cell Physiol

    (2008)
  • BlochK.D. et al.

    Biosynthesis and secretion of proatrial natriuretic factor by cultured rat cardiocytes

    Science

    (1985)
  • BlumenthalD.K. et al.

    Phosphorylation of cardiac troponin by guanosine 3':5'-monophosphate-dependent protein kinase

    J Biol Chem

    (1978)
  • BoerrigterG. et al.

    Targeting heme-oxidized soluble guanylate cyclase in experimental heart failure

    Hypertension

    (2007)
  • BoerrigterG. et al.

    Cardiorenal and humoral properties of a novel direct soluble guanylate cyclase stimulator BAY 41-2272 in experimental congestive heart failure

    Circulation

    (2003)
  • BolotinaV.M. et al.

    Nitric oxide directly activates calcium-dependent potassium channels in vascular smooth muscle

    Nature

    (1994)
  • BoolellM. et al.

    Sildenafil, a novel effective oral therapy for male erectile dysfunction

    Br J Urol

    (1996)
  • BorlaugB.A. et al.

    Sildenafil inhibits beta-adrenergic-stimulated cardiac contractility in humans

    Circulation

    (2005)
  • BrovkovychV. et al.

    Augmented inducible nitric oxide synthase expression and increased NO production reduce sepsis-induced lung injury and mortality in myeloperoxidase-null mice

    Am J Physiol Lung Cell Mol Physiol

    (2008)
  • BruneB. et al.

    Activation of soluble guanylate cyclase by carbon monoxide and inhibition by superoxide anion

    Eur J Biochem

    (1990)
  • Cited by (336)

    View all citing articles on Scopus
    View full text