Open access peer-reviewed chapter

G Protein-Coupled Receptor Regulation in Cardiovascular Disease: Role of G Protein-Coupled Receptor Kinases

Written By

Asma S. Alonazi, Anfal F. Bin Dayel, Tahani K. Alshammari and Nouf M. Alrasheed

Submitted: 24 April 2022 Reviewed: 13 May 2022 Published: 18 June 2022

DOI: 10.5772/intechopen.105403

From the Edited Volume

Novel Pathogenesis and Treatments for Cardiovascular Disease

Edited by David C. Gaze

Chapter metrics overview

164 Chapter Downloads

View Full Metrics

Abstract

G protein-coupled receptor kinases (GRKs), the negative regulators of G protein-coupled receptors (GPCRs), have a key role in cardiovascular disease pathophysiology. Alteration in GRKs’ expressions and/or kinase activity has been reported in preclinical animal models as well as in patients with cardiovascular diseases. This alteration might be a contributing factor to disease progression by a variety of mechanisms such as non-canonical transduction pathways. The current chapter is aimed to expand our knowledge and understanding of the function of GRKs in cardiovascular diseases, highlight their involvement, and illustrate the possible mechanistic role of GRKs in hypertensive vascular diseases and cardiac myopathy. The current chapter also is endeavoured to identify the potential molecular mechanisms by which GRKs participate in cardiovascular disease progression. Building the basics knowledge about GRKs in cardiovascular diseases will help to assess the potential utilization of GRKs as therapeutic targets and to examine the possible approaches to modulate their protein expression or to inhibit their kinase activity to prevent or attenuate cardiovascular disease progression.

Keywords

  • GRK2
  • GRK5
  • GPCR regulation
  • cardiovascular diseases
  • heart failure
  • hypertension
  • myocardial infarction

1. Introduction

Cardiovascular diseases consider as one of the major causes of death, contributing to approximately 30% of all deaths globally. In Kingdom of Saudi Arabia, approximately 37% deaths are caused by cardiovascular diseases [1]. Elevated cardiovascular disease-related morbidity and mortality result from a complex pathophysiological process including activation of many signaling transduction pathways, resulting in modification in cardiac/vascular structure, remodeling, and ultimately alteration in the functionality, which contributes to disease progression. Of importance, G protein-coupled receptors (GPCR) play a key fundamental role in various transductions signaling that participate in cardiovascular diseases pathophysiological progression. GPCRs are a superfamily of heptahelical integral membrane proteins, which respond to various stimuli. They are responsible for transduction of a plethora of signaling networks that involve in physiological and pathological actions in cardiovascular system [2, 3]. The activation of α-adrenergic, β-adrenergic, muscarinic, angiotensin II type 1 receptor (AT1) and endothelin (ETA) receptors are involved in cardiac contractility, vascular resistance, vascular and cardiac remodeling. In addition, the effect of neurohumoral systems on cardiac contractility and blood vessel tone mainly transmit their signals via corresponding GPCR. These types of receptors and their downstream transduction systems are targets of various drugs used in the treatment of cardiovascular diseases [4].

In case of hypertension, GPCRs play fundamental function in blood vessel diameter, which is mainly controlled by either contraction or relaxation of vascular smooth muscle cells. During contraction, GPCR mediated phosphorylation of contractile proteins [5]. Vasoactive peptides such as noradrenaline, angiotensin II, endothelin 1, and vasopressin activated their corresponding Gαq coupled GPCR, results in stimulation of phospholipase C-β, resulting in the formation of inositol-1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 binds to their corresponding receptors which is inositol trisphosphate receptors (IP3Rs) in the sarcoplasmic reticulum; the intracellular Ca2+ store. Activation of IP3Rs resulting in efflux of Ca2+ into the cytoplasm. On other hand, DAG activates protein kinase C (PKC), promoting Ca2+ influx via enhancing the vascular channels activity such as voltage-dependent L-type Ca2+ channels. Elevated intracellular Ca2+ concentration [Ca2+]i binds to calmodulin, creating Ca2+-calmodulin complex which activate MLC kinase (MLCK) followed by phosphorylation of contractile proteins that promote myosin-actin filament interactions and consequently smooth muscle contraction [6, 7, 8, 9, 10, 11]. Contraction of vascular smooth muscle cells could persist via regulation of MLC phosphatase (MLCP). Vasoactive peptides also regulate the dephosphorylation of MLC phosphatase via different mechanisms. They utilize the PLC-DAG-PKC pathway, which in turn inhibits phosphatase activity and thus stimulates persistent contraction [12]. In addition, they activate RhoA-Rho kinase pathway, which phosphorylates MLCP and inhibits its activity [13, 14]. On the other hand, a low [Ca2+]i concentration and increased activity of MLC phosphatase promoting vascular smooth muscle cell relaxation [15]. Gαs-coupled GPCR mediated blood vessel relaxation. Adrenaline, as vasodilator, acts on corresponding receptors, recruiting Gαs to stimulate adenylyl cyclase (AC), leading to the formation of cAMP and then activation of protein kinase A (PKA). PKA plays important role in decreasing [Ca2+]i concentrations via phosphorylation of MLCK. This results in activation of calcium pumps in the plasma membrane and sarcoplasmic reticulum. Furthermore, promotes cell hyperpolarization by opening K+ channels promoting relaxation of vascular smooth muscle cells [8, 16, 17].

In case of heart failure, GPCR such as β-adrenergic receptors plays an essential role in cardiac function and in cardiac myocytes contractility. β1-adrenergic receptors couple to Gαs that activates adenylate cyclase (AC) and enhances cAMP mediate protein kinase A (PKA) activation which regulates different intracellular, sarcolemma, and myofibrillar substrates, mediating positive inotropic and chronotropic effects [18]. Gβγ subunits also activate downstream effectors that participate in cardiac transduction pathways. Moreover, it has been reported that overexpression of β1-adrenergic receptors triggers early myocytes hypertrophy and interstitial fibrosis followed by marked cardiac dysfunction in mice [18]. In addition, β1-adrenergic receptors activate various downstream signaling participating in cardiac pathophysiological processes such as cardiac hypertrophy, which might progress to heart failure development [19, 20, 21]. β2-adrenergic receptors can couple to a dual Gαs/Gαi subunits, it has been implicated in differential β2-adrenergic receptors mediated signaling such as in myocyte apoptosis [22]. Therefore, β-adrenoceptor blockers are one of the standard pharmacotherapeutics agents used in the treatment of heart failure patients [23]. β-blockers also have been shown to reduce disease progression, mortality, and morbidity in patients with heart failure with reduced ejection fraction (HFrEF) [24]. This effect appears primarily related to the ability of β-adrenoceptor blockers to protect the heart from the harmful effects of receptor over-stimulation [25].

As the effect of neurohumoral systems on cardiovascular system mainly transmits their signals via GPCRs, understanding of the GPCR regulation and their G-protein dependent/independent signaling reveals a novel therapeutic approach that could attenuate cardiovascular-related complications. In current chapter, we provide insight into the potential effect of GPCR negative regulators, focusing particularly on G protein-coupled receptor kinases (GRKs) and their possible effect on cardiovascular diseases.

Advertisement

2. GPCRs regulations

Repeated or prolonged/continues agonist stimulation of GPCRs resulted in loss of receptor response characterized as receptor desensitization. It can be described as physical uncoupling of G proteins from their associate receptors subsequent in diminish their ability to initiate intracellular signaling cascades [26]. As shown in Figure 1, receptor desensitization process is initiated by receptor phosphorylation. It can be mediated by G protein-coupled receptor kinases (GRKs), which phosphorylate agonist-bound active receptor inducing homologous type of receptor desensitization [27]. Receptor phosphorylation can be also mediated by second messenger kinases such as PKA and PKC, which can phosphorylate receptors regardless of whether the GPCR is occupied by agonist or not, thus producing heterologous type of receptor desensitization [27, 28, 29]. Receptor phosphorylation subsequently increases the affinity of the receptors for β-arrestins proteins binding, consequently prevents further receptors-G protein interactions and therefore termination of G protein-related signaling [30]. Accordingly, the phosphorylated GPCR/β-arrestin complex is subjected for clathrin-mediated endocytosis, followed by either recycling, or degradation [31, 32, 33]. Importantly, β-arrestins function as ligand-regulated adaptor scaffolds that enable the transduction of signaling pathways in non-canonical manner [34].

Figure 1.

GPCR desensitization and related signaling transductions pathways. GPCR desensitization by GRKs and β-arrestins process initiated with ligand binding, receptor activation and dissociation of G protein. Consequently, GRKs phosphorylate agonist-occupied GPCRs (activated receptors) at third intracellular loop or C-terminal. Receptor phosphorylation resulting in enhances the affinity for β-arrestin recruitment then binding to the receptors causing termination of G-protein dependent transduction signaling and receptor desensitization. After that, the receptors undergo internalization and initiation of G protein-independent transduction signaling.

Advertisement

3. G protein-coupled receptors kinases (GRKs)

G Protein-Coupled Receptors Kinases (GRKs) are family of seven members of serine/threonine kinases [35]. They are allocated into three subcategories: the first category is visual GRKs including GRK1 and GRK7; the second and third categories are non-visual GRKs including β-adrenergic receptor kinase subcategory, containing GRK2 (β-ARK1) and GRK3 (β-ARK2) and; the GRK4 subcategory, containing GRK4, GRK5, and GRK6 [27]. Regarding GRKs tissue distribution, GRK1 and GRK7 are primarily expressed in the retina mediating photoreceptor regulation. GRK2, GRK3, GRK5, and GRK6 are ubiquitously expressed in various tissues; however, GRK4 is limitedly distributed to testes, kidneys, and some areas of the brain. Hence, GRK2, GRK3, GRK5, or GRK6 is the potential regulator for the majority of GPCRs [36, 37, 38].

In cardiovascular system, the distribution pattern and expression levels of GRK are crucial factors contributing to their functionality in various cell types. Previous reports show that GRK2, GRK3, and GRK5 are highly expressed in the human heart [39]. GRK isoforms distribution is different among various heart cells. GRK2 and GRK5 are expressed in almost all cardiac cells, while GRK3 distribution is limited to cardiac myocytes [4, 40]. GRK2 is expressed in the vascular endothelium, arterial smooth muscle, and in the myocardium. GRK2 is also expressed in the kidney, especially in the renal proximal tubule [41].

The structure of GRKs comprises of three domains: N-terminal; an amino terminal domain, central serine/threonine protein kinase/catalytic domain, and C-terminal; a carboxyl terminal domain. The N-terminal domain is implicated in receptor recognition. It includes a region of a regulator of G protein signaling (RGS) homology domain (RH) [31, 35]. In GRK2, Gβγ binding site has been mapped in the N-terminal region causing binding of GRK2 to cell membrane [42]. The central domain is a serine/threonine protein that exerts the kinase catalytic function in all GRKs. The C-terminal domain structure is different among GRKs subfamilies. It is implicated in GRK membrane localization. For instant, GRK5 is located at cell membrane level as the C-terminus of GRK5 contains lipid-binding sites that interact with the phospholipid in the cell membrane. On other hand, GRK2 and GRK3 are cytoplasmic proteins that are recruited to the plasma membrane upon agonist binding and receptor activation. Their C-terminal domains contain pleckstrin homology (PH) domain, which comprises binding sites for the cell membrane phospholipid (PIP2) and Gβγ subunits [26, 35, 43]. As a multi-domain protein, GRK acts as a negative GPCR signaling regulator, terminating G protein-dependent signaling and initiating other G protein-independent signaling pathways [30, 44]. For instance, reported evidence reported that GRK functions are expanded more than receptors phosphorylation. GRKs are able to interact with various cellular proteins mediating non-canonical GPCR signaling [31, 45]. Of importance, GRK expression and activity are changed in many cardiovascular diseases such as in case of hypertension or heart failure. Thus, better understanding of the diseases associated with alteration in GRKs’ expression as well as their functional roles in cardiovascular system is fundamental to develop a new therapeutic target.

3.1 GRKs in hypertension

In hypertension, continuous activation of Gαq coupled receptors such as ETA and AT1 mediate vascular smooth muscle contraction and enhance peripheral vascular resistance [46]. Current evidence shows that GRK2 plays an important function in regulation of prolonged Gαq-related signaling in vascular smooth muscle cells and consider as the main negative regulator of vasoactive peptide corresponding GPCRs. Moreover, previously published studies reported that GRK2 negatively regulate ETA and P2Y2 receptors in aortic smooth muscle cells [47, 48]. Reported evidence shows that inhibition of GRK2 kinase activity diminishes the desensitization process of AngII/AT1 and UTP/P2Y2 induced arterial smooth muscle contractions [49]. Indeed, published studies show that GRK2 expression is augmented in hypertension, in both hypertensive animal models and hypertensive patients [50, 51, 52, 53, 54, 55]. Therefore, enhanced GRK2 expression may possibly participate in the pathophysiology of hypertension development. For instant, GRK2 has been reported to attenuate endothelial NO production [56]. Furthermore, GRK2 is reported to mediate the desensitization of β-adrenoceptors, which mediates vasodilation. Thus, enhanced GRK2 expressions may impair vasodilation in hypertension, which possibly contributes to enhancing vascular tone and elevation of blood pressure [53]. Additionally, it has been reported that GRK2 overexpression in vascular smooth muscle cells resulted in a 30% increase in vascular wall thickness [52], suggesting a possible link between GRK2 over-expression in hypertension and hypertension-induced vascular remodeling [57]. Recently published paper show that elevated GRK2 expression hypertension has a potential to promote vascular smooth muscle growth and proliferation possibly via PI3K-Akt signaling, followed by release the GSK3-mediated inhibition of cell cycling progression, therefore aggravate hypertensive induced pathophysiological vascular remodeling [58].

Still, it is not clear if the changes in GRK2 expression are a contributing factor for hypertension development or a consequence of hypertension, which needs further investigation. Moreover, further investigations are required to understand the molecular mechanisms underlying these changes and how the alterations in GRK expression implicated in triggering or progression of hypertension might contribute to the development of novel diagnostic and/or therapeutic strategies to control hypertension or prevent its complications.

3.2 GRKs in heart failure

Myocardial GRK2 and GRK5 have been shown to be involved in the pathophysiology of heart failure [40]. Indeed, several evidences highlight GRK2 as well as GRK5 as the key regulators of β-adrenoceptor [59, 60]. Of importance, recently published paper describes that GRK2 and GRK5 are new therapeutic targets for pathological cardiac hypertrophy and may attenuate morbidity and mortality rates [61]. Dysregulation of β-adrenoceptor is a pathological characteristic of heart failure; in particular, the receptors are considerably downregulated and desensitized as a result of the upregulated levels of GRK2 and GRK5 [16]. Enhanced expression and activity of GRK2 are associated with the loss of β-adrenoceptor functions that induces deleterious effects in the heart functionality contribute to progression of heart failure [62]. Overstimulation of β-adrenoceptor as a subsequent of continuous sympathetic activation, resulting in GRK and β-arrestins induced desensitization and downregulation of β-adrenoceptor [63]. Initially, this process is adaptive response to overcome receptor overactivation. However, prolonged excessive stimulation mediated receptor downregulation has been reported inducing harmful effect to the heart and consequently heart failure development [63, 64]. Notably, alterations in GRKs have been observed in heart failure [39, 65, 66]. Indeed, several evidences highlight GRK2 as well as GRK5 as the key regulators of β-adrenoceptor [59, 60]. Several reported evidence have shown that GRK2 expression and activity are significantly increased in the failing heart [39, 67, 68]. Enhanced GRK2 expression and altered functionality have been found in heart failure status [39, 65, 66, 69]. Moreover, up-regulation of GRK2 level was detected in end-stage dilated heart failure patient [65]. Even though the mechanism of β-adrenoceptor overstimulation mediated GRK2 upregulation is not clearly understood, published reports show that GRK2 dysfunction plays an essential function in the pathophysiology of heart failure [70] suggesting that alteration in GRK2 function participates in heart failure pathology. Altered GRK2 expression or activity appears to contribute to disease progression through various molecular mechanisms. Therefore, targeting GRK2 expression or inhibition of its activity has been suggested as a therapeutics strategy for treatment of heart failure patients [7172].

Reported evidence shows that overexpression of a peptide inhibitor of GRK2; carboxy terminal domain (βARKct) which lacks to membrane translocation function inhibits GRK2 activity and prevents desensitization of the receptors resulting in restoring of β-adrenoceptor function and enhanced cardiac contractility in experimental animals of heart failure [73, 74, 75]. Moreover, Gβγ-GRK2 inhibition reduces pathological effect of myofibroblast activation. Thus, Gβγ-GRK2 inhibition might be a potential therapeutic strategy to attenuate pathological myofibroblast activation, interstitial fibrosis, cardiac remodeling, and progression of heart failure [76].

It has been reported that cardiac dysfunction could be attenuated by inhibition of GRK2 activity [62]. Interestingly, it has been reported that paroxetine, selective serotonin re-uptake inhibitor (SSRI) approved by FDA for treatment of depression, significantly inhibited GRK2 kinase activity [49, 77, 78]. Published studies showed capability paroxetine as GRK2 inhibitor in reversing cardiac remodeling in experimental models of acute myocardial infarction [79, 80]. Therefore, paroxetine may perhaps be used as a therapeutic approach for targeting GRK2 catalytic activity and potentially provide a protective role against cardiac hypertrophy development via its function as GRK2 inhibitor.

GRK5 another GRK member that mediates phosphorylation and desensitization of β-adrenoceptor is well known to regulate heart functions [72]. Several studies suggest that GRK5 plays a crucial role in various cardiovascular diseases. For instance, previous studies show that GRK5 overexpressing mice developed cardiac hypertrophy, which rapidly progressed to heart failure [81]. Moreover, GRK5 knockout mice showed attenuated hypertrophic responses [82]. Furthermore, GRK5 overexpressing mice showed an alteration in myocardial performance including attenuation of contractility, cardiac output, stroke work, and stroke volume [83]. Of note, GRK5 levels were shown to be markedly elevated in heart failure patients and patients with left ventricular volume overload disorders and dilated cardiomyopathic hearts [84, 85, 86].

GRK5 overexpressed transgenic mice exhibited enhanced susceptibility to pressure overload-induced cardiac hypertrophy and cardiac dysfunction [87]. Furthermore, cardiac-specific GRK5 transgenic mice demonstrated reduced cardiac function and increased adverse cardiac remodeling in a myocardial infarction-induced heart failure mice model [64]. On other hand, heart hypertrophic responses were attenuated in GRK5 knockout mice [88], these studies demonstrated the possible functions of GRK5 in pathological cardiac remodeling development. Interestingly, several lines of evidence show that GRK5 can translocate to the nucleus, exerting its non-canonical functions. For instance, it was shown that GRK5 in the cardiomyocyte nuclei acts as a class II histone deacetylase (HDAC) kinase, phosphorylating HDAC5 and leading to de-repression of myocyte enhancer factor 2 (MEF2)-mediated hypertrophic gene transcription [81, 89]. In addition, it was demonstrated that GRK5 interacts with hypertrophic transcription factors like nuclear factor of activated T cell (NFAT) and nuclear factor κ-B (NF-κB) [81, 87, 90, 91]. These studies indicate that GRK5 has a major role in the pathogenesis of the cardiovascular disorders and GRK5 might be a therapeutic target for heart failure. Recently, it has been demonstrated that KR-39038, a novel small molecule inhibitor of GRK5, significantly inhibited cellular hypertrophy and HDAC5 phosphorylation in neonatal rat ventricular myocytes. This inhibitor was able to minimize the left ventricular weight, improve cardiac function and ameliorate myocardial remodeling in animal model of heart failure [92]. Another important agent proposed as a GRK5 inhibitor is an anti-inflammatory and anti-allergic immunomodulator, named amlexanox [93]. This agent was able to inhibit GRK5 induced MEF2 activation in neonatal rat ventricular myocytes and inhibit GRK5 mediated HDAC5 phosphorylation in cellular model of cardiac hypertrophy [93, 94].

3.3 GRKs in myocardial infarction

It has been reported that GRK2 expressions upregulated in peripheral blood lymphocytes in patients with acute ST-segment elevation myocardial infarction. Enhanced lymphocyte GRK2 expressions are associated with worse cardiac functionality. These studies indicate that GRK2 could be predictive of myocardial remodeling after myocardial infarction [95, 96]. Enhanced GRK2 levels and activity are deleterious to post-ischemic myocardium in acute ischemia/reperfusion (I/R) injury animal model [97]. It has been reported that GRK2 peptide inhibitor; βARKct provides cardioprotective effect, which modulate GRK2-mediated PI3K-Akt-NOS signaling pathway in the ischemic heart which validates GRK2-related effect on survival and apoptotic signaling in the ischemic heart [97]. βARKct expression mediated GRK2 inhibition modulate Akt downstream pro-survival signaling such as reduced Caspase-3 activity, increased eNOS activation and NO production and then reduced apoptosis and cell death [97]. Furthermore, decreasing GRK2 expression in cardiomyocytes attenuate myocyte apoptosis possibly via Akt/Bcl-2 mediated mitochondrial protection and limits I/R- provoked injury and improves post-ischemia recovery in the heart [98]. Additionally, it has been reported that fibroblast specific GRK2 knockout has a protective effect after myocardial I/R injury in mice. GRK2 fibroblast knockout mice decreased the infarct size, increased ejection fraction, preserved cardiac function, and also reduced tumor necrosis factor-α expression, fibrotic gene expression, and fibrosis development [99].

Advertisement

4. Conclusions

Cardiovascular diseases are a leading cause of death worldwide. The pathophysiological mechanisms are regulated by a GPCR mediated complex network of transduction pathways. The functions of GRKs, negative regulators of GPCR, are not limited to receptors desensitization. It is expanded further to activations of many transductions in non-classical manner. As the expression and kinase activity of GRK2 and GRK5 are altered in cardiovascular diseases, Therefore, better knowledge of the transduction events which mediated by up-regulated GRK2 and/or GRK5 in terms of the expression, activity, and localization would help to develop a novel strategy for targeting their expressions or inhibiting activity. This will participate in building a knowledge-based platform identifying a new therapeutic target to prevent the progression of cardiovascular diseases. Many different approaches could be applied, including small molecule inhibitors, gene therapy, and the use of advanced drug delivery systems to potentially prevent the progression of cardiovascular disease. Overall, GRKs play an important role in cardiovascular diseases progression. Pharmacological intervention of GRK5 as well as GRK2 would provide a novel possible future target for cardiovascular disease progression prevention.

Advertisement

Acknowledgments

The authors extend their appreciation to the Deputyship for Research & Innovation, Ministry of Education in Saudi Arabia for funding this research work through project no. (IFKSUDR_H150).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Organization WH. Noncommunicable Diseases Country Profiles 2018. 2018
  2. 2. Fredriksson R, Lagerström MC, Lundin L-G, Schiöth HB. The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Molecular Pharmacology. 2003;63(6):1256-1272
  3. 3. Gether U. Uncovering molecular mechanisms involved in activation of G protein-coupled receptors. Endocrine Reviews. 2000;21(1):90-113
  4. 4. Penela P, Murga C, Ribas C, Tutor AS, Peregrín S, Mayor F Jr. Mechanisms of regulation of G protein-coupled receptor kinases (GRKs) and cardiovascular disease. Cardiovascular Research. 2006;69(1):46-56
  5. 5. Touyz RM, Alves-Lopes R, Rios FJ, Camargo LL, Anagnostopoulou A, Arner A, et al. Vascular smooth muscle contraction in hypertension. Cardiovascular Research. 2018;114(4):529-539
  6. 6. Lymperopoulos A, Bathgate A. Arrestins in the cardiovascular system. Progress in Molecular Biology and Translational Science. 2013;118:297-334
  7. 7. Hill-Eubanks DC, Werner ME, Heppner TJ, Nelson MT. Calcium signaling in smooth muscle. Cold Spring Harbor Perspectives in Biology. 2011;3(9):a004549
  8. 8. Harris DM, Cohn HI, Pesant S, Eckhart AD. GPCR signalling in hypertension: Role of GRKs. Clinical Science (Lond). 2008;115(3):79-89
  9. 9. Lozinskaya I, Matsuda K, Cox RH. Augmented contributions of voltage-Gated Ca2 channels to contractile responses in spontaneously hypertensive rat mesenteric arteries*. American Journal of Hypertension. 1997;10(11):1231-1239
  10. 10. Allen BG, Walsh MP. The biochemical basis of the regulation of smooth-muscle contraction. Trends in Biochemical Sciences. 1994;19(9):362-368
  11. 11. Ghosh D, Syed AU, Prada MP, Nystoriak MA, Santana LF, Nieves-Cintron M, et al. Calcium channels in vascular smooth muscle. Advances in Pharmacology. 2017;78:49-87
  12. 12. Ringvold HC, Khalil RA. Protein kinase C as regulator of vascular smooth muscle function and potential target in vascular disorders. Advances in Pharmacology. 2017;78:203-301
  13. 13. Loirand G, Pacaud P. The role of Rho protein signaling in hypertension. Nature Reviews Cardiology. 2010;7(11):637-647
  14. 14. Puetz S, Lubomirov LT, Pfitzer G. Regulation of smooth muscle contraction by small GTPases. Physiology (Bethesda). 2009;24:342-356
  15. 15. Webb RC. Smooth muscle contraction and relaxation. Advances in Physiology Education. 2003;27(1-4):201-206
  16. 16. Fukunaga K, Kume H, Oguma T, Shigemori W, Tohda Y, Ogawa E, et al. Involvement of Ca(2+) signaling in the synergistic effects between muscarinic receptor antagonists and β2-adrenoceptor agonists in airway smooth muscle. International Journal of Molecular Sciences. 2016;17(9):1590
  17. 17. Consigny PM. Vascular smooth muscle contraction and relaxation: Pathways and chemical modulation. Journal of Vascular and Interventional Radiology. 1991;2(3):309-317
  18. 18. Engelhardt S, Hein L, Wiesmann F, Lohse MJ. Progressive hypertrophy and heart failure in beta1-adrenergic receptor transgenic mice. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(12):7059-7064
  19. 19. Xiang Y, Kobilka BK. Myocyte adrenoceptor signaling pathways. Science. 2003;300(5625):1530-1532
  20. 20. Bernstein D, Fajardo G, Zhao M, Urashima T, Powers J, Berry G, et al. Differential cardioprotective/cardiotoxic effects mediated by β-adrenergic receptor subtypes. American Journal of Physiology-Heart and Circulatory Physiology. 2005;289(6):H2441-H24H9
  21. 21. Khamssi M, Brodde O-E. The role of cardiac beta1-and beta2-adrenoceptor stimulation in heart failure. Journal of Cardiovascular Pharmacology. 1990;16:S133-S137
  22. 22. Communal C, Singh K, Sawyer DB, Colucci WS. Opposing effects of beta(1)- and beta(2)-adrenergic receptors on cardiac myocyte apoptosis: Role of a pertussis toxin-sensitive G protein. Circulation. 1999;100(22):2210-2
  23. 23. Chavey WE. The importance of beta blockers in the treatment of heart failure. American Family Physician. 2000;62(11):2453-2462
  24. 24. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JG, Coats AJ, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. European Journal of Heart Failure. 2016;18(8):891-975
  25. 25. Bristow MR. beta-adrenergic receptor blockade in chronic heart failure. Circulation. 2000;101(5):558-569
  26. 26. Penela P, Ribas C, Mayor F. Mechanisms of regulation of the expression and function of G protein-coupled receptor kinases. Cellular Signalling. 2003;15(11):973-981
  27. 27. Willets JM, Challiss RA, Nahorski SR. Non-visual GRKs: Are we seeing the whole picture? Trends in Pharmacological Sciences. 2003;24(12):626-633
  28. 28. Hausdorff WP, Caron MG, Lefkowitz RJ. Turning off the signal: Desensitization of beta-adrenergic receptor function. FASEB Journal. 1990;4(11):2881-2889
  29. 29. Stadel JM, Nambi P, Shorr RG, Sawyer DF, Caron MG, Lefkowitz RJ. Catecholamine-induced desensitization of turkey erythrocyte adenylate cyclase is associated with phosphorylation of the beta-adrenergic receptor. Proceedings of the National Academy of Sciences. 1983;80(11):3173-3177
  30. 30. Brinks HL, Eckhart AD. Regulation of GPCR signaling in hypertension. Biochimica et Biophysica Acta. 2010;1802(12):1268-1275
  31. 31. Penela P, Murga C, Ribas C, Lafarga V, Mayor F Jr. The complex G protein-coupled receptor kinase 2 (GRK2) interactome unveils new physiopathological targets. British Journal of Pharmacology. 2010;160(4):821-832
  32. 32. Kelly E, Bailey CP, Henderson G. Agonist-selective mechanisms of GPCR desensitization. British Journal of Pharmacology. 2008;153(Suppl. 1):S379-SS88
  33. 33. Oakley RH, Laporte SA, Holt JA, Caron MG, Barak LS. Differential affinities of visual arrestin, βArrestin1, and βArrestin2 for G protein-coupled receptors delineate two major classes of receptors. Journal of Biological Chemistry. 2000;275(22):17201-17210
  34. 34. Luttrell LM, Miller WE. Arrestins as regulators of kinases and phosphatases. Progress in Molecular Biology and Translational Science. 2013;118:115-147
  35. 35. Homan KT, Tesmer JJG. Structural insights into G protein-coupled receptor kinase function. Current Opinion in Cell Biology. 2014;27:25-31
  36. 36. Premont RT, Gainetdinov RR. Physiological roles of G protein-coupled receptor kinases and arrestins. Annual Review of Physiology. 2007;69:511-534
  37. 37. Pugh EN, Lamb TD. Chapter 5 Phototransduction in vertebrate rods and cones: Molecular mechanisms of amplification, recovery and light adaptation. In: Stavenga DG, DeGrip WJ, Pugh EN, editors. Handbook of Biological Physics. vol. 3. North-Holland; 2000. pp. 183-255. DOI: 10.1016/S1383-8121(00)80008-1
  38. 38. Premont RT, Macrae AD, Stoffel RH, Chung N, Pitcher JA, Ambrose C, et al. Characterization of the G protein-coupled receptor kinase GRK4. Identification of four splice variants. The Journal of Biological Chemistry. 1996;271(11):6403-6410
  39. 39. Ungerer M, Böhm M, Elce J, Erdmann E, Lohse M. Altered expression of beta-adrenergic receptor kinase and beta 1-adrenergic receptors in the failing human heart. Circulation. 1993;87(2):454-463
  40. 40. Vinge LE, Øie E, Andersson Y, Grøgaard HK, Andersen G, Attramadal H. Myocardial distribution and regulation of GRK and beta-arrestin isoforms in congestive heart failure in rats. The American Journal of Physiology-Heart and Circulatory Physiology. 2001;281(6):H2490-H2499
  41. 41. Yang J, Villar VAM, Armando I, Jose PA, Zeng C. G protein-coupled receptor kinases: Crucial regulators of blood pressure. Journal of the American Heart Association. 2016;5(7):e003519
  42. 42. Eichmann T, Lorenz K, Hoffmann M, Brockmann J, Krasel C, Lohse MJ, et al. The amino-terminal domain of G-protein-coupled receptor kinase 2 is a regulatory Gbeta gamma binding site. The Journal of Biological Chemistry. 2003;278(10):8052-8057
  43. 43. Ribas C, Penela P, Murga C, Salcedo A, Garcia-Hoz C, Jurado-Pueyo M, et al. The G protein-coupled receptor kinase (GRK) interactome: Role of GRKs in GPCR regulation and signaling. Biochimica et Biophysica Acta. 2007;1768(4):913-922
  44. 44. Reiter E, Lefkowitz RJ. GRKs and β-arrestins: Roles in receptor silencing, trafficking and signaling. Trends in Endocrinology & Metabolism. 2006;17(4):159-165
  45. 45. Shenoy SK, Lefkowitz RJ. β-arrestin-mediated receptor trafficking and signal transduction. Trends in Pharmacological Sciences. 2011;32(9):521-533
  46. 46. Harris DM, Cohn HI, Pesant S, Zhou RH, Eckhart AD. Vascular smooth muscle G(q) signaling is involved in high blood pressure in both induced renal and genetic vascular smooth muscle-derived models of hypertension. The American Journal of Physiology-Heart and Circulatory Physiology. 2007;293(5):H3072-H3079
  47. 47. Morris GE, Nelson CP, Everitt D, Brighton PJ, Standen NB, Challiss RA, et al. G protein-coupled receptor kinase 2 and arrestin2 regulate arterial smooth muscle P2Y-purinoceptor signalling. Cardiovascular Research. 2011;89(1):193-203
  48. 48. Morris GE, Nelson CP, Standen NB, Challiss RA, Willets JM. Endothelin signalling in arterial smooth muscle is tightly regulated by G protein-coupled receptor kinase 2. Cardiovascular Research. 2010;85(3):424-433
  49. 49. Rainbow RD, Brennan S, Jackson R, Beech AJ, Bengreed A, Waldschmidt HV, et al. Small-molecule G Protein-coupled receptor kinase inhibitors attenuate G protein-coupled Receptor Kinase 2-mediated desensitization of vasoconstrictor-induced arterial contractions. Molecular Pharmacology. 2018;94(3):1079-1091
  50. 50. Willets JM, Nash CA, Rainbow RD, Nelson CP, Challiss RA. Defining the roles of arrestin2 and arrestin3 in vasoconstrictor receptor desensitization in hypertension. The American Journal of Physiology-Cell Physiology. 2015;309(3):C179-C189
  51. 51. Cohn HI, Harris DM, Pesant S, Pfeiffer M, Zhou RH, Koch WJ, et al. Inhibition of vascular smooth muscle G protein-coupled receptor kinase 2 enhances alpha1D-adrenergic receptor constriction. The American Journal of Physiology-Cell Physiology. 2008;295(4):H1695-H1704
  52. 52. Eckhart AD, Ozaki T, Tevaearai H, Rockman HA, Koch WJ. Vascular-targeted overexpression of G protein-coupled receptor kinase-2 in transgenic mice attenuates beta-adrenergic receptor signaling and increases resting blood pressure. Molecular Pharmacology. 2002;61(4):749-758
  53. 53. Gros R, Chorazyczewski J, Meek MD, Benovic JL, Ferguson SS, Feldman RD. G-protein-coupled receptor kinase activity in hypertension: Increased vascular and lymphocyte G-protein receptor kinase-2 protein expression. Hypertension. 2000;35(1 Pt 1):38-42
  54. 54. Cohn HI, Xi Y, Pesant S, Harris DM, Hyslop T, Falkner B, et al. G protein-coupled receptor kinase 2 expression and activity are associated with blood pressure in black Americans. Hypertension. 2009;54(1):71-76
  55. 55. Gros Robert BJL, Tan Christopher M, Feldman Ross D. G-protein–coupled receptor kinase activity is increased in hypertension. Journal of Clinical Investigation. 1997;99:2087-2093
  56. 56. Feldman RD. Deactivation of vasodilator responses by GRK2 overexpression: A mechanism or the mechanism for hypertension? Molecular Pharmacology. 2002;61(4):707-709
  57. 57. Ramos-Ruiz R, Penela P, Penn RB, Mayor F Jr. Analysis of the human G protein-coupled receptor kinase 2 (GRK2) gene promoter: Regulation by signal transduction systems in aortic smooth muscle cells. Circulation. 2000;101(17):2083-2089
  58. 58. Alonazi ASA, Willets JM. G protein-coupled receptor kinase 2 is essential to enable vasoconstrictor-mediated arterial smooth muscle proliferation. Cell Signal. 2021;88:110152
  59. 59. Hu LA, Chen W, Premont RT, Cong M, Lefkowitz RJ. G protein-coupled receptor kinase 5 regulates β1-adrenergic receptor association with PSD-95. Journal of Biological Chemistry. 2002;277(2):1607-1613
  60. 60. Huang ZM, Gao E, Chuprun JK, Koch WJ. GRK2 in the heart: A GPCR kinase and beyond. Antioxidants & Redox Signaling. 2014;21(14):2032-2043
  61. 61. Lieu M, Koch WJ. GRK2 and GRK5 as therapeutic targets and their role in maladaptive and pathological cardiac hypertrophy. Expert Opinion on Therapeutic Targets. 2019;23(3):201-214
  62. 62. Petrofski JA, Koch WJ. The beta-adrenergic receptor kinase in heart failure. Journal of Molecular and Cellular Cardiology. 2003;35(10):1167-1174
  63. 63. Lymperopoulos A, Rengo G, Koch WJ. Adrenergic nervous system in heart failure: Pathophysiology and therapy. Circulation Research. 2013;113(6):739-753
  64. 64. de Lucia C, Eguchi A, Koch WJ. New insights in cardiac β-adrenergic signaling during heart failure and aging. Frontiers in Pharmacology. 2018;9:904
  65. 65. Ungerer M, Parruti G, Böhm M, Puzicha M, DeBlasi A, Erdmann E, et al. Expression of beta-arrestins and beta-adrenergic receptor kinases in the failing human heart. Circulation Research. 1994;74(2):206-213
  66. 66. Leineweber K, Rohe P, Beilfuß A, Wolf C, Sporkmann H, Bruck H, et al. G-protein-coupled receptor kinase activity in human heart failure: Effects of β-adrenoceptor blockade. Cardiovascular Research. 2005;66(3):512-519
  67. 67. Harris CA, Chuang TT, Scorer CA. Expression of GRK2 is increased in the left ventricles of cardiomyopathic hamsters. Basic Research in Cardiology. 2001;96(4):364-368
  68. 68. Sato PY, Chuprun JK, Schwartz M, Koch WJ. The evolving impact of G protein-coupled receptor kinases in cardiac health and disease. Physiological Reviews. 2015;95(2):377-404
  69. 69. Bohm M, Lohse MJ. Quantification of beta-adrenoceptors and beta-adrenoceptor kinase on protein and mRNA levels in heart failure. European Heart Journal. 1994;15(Suppl. D):30-34
  70. 70. Mangmool S, Parichatikanond W, Kurose H. Therapeutic targets for treatment of heart failure: Focus on GRKs and β-Arrestins affecting βAR signaling. Frontiers in Pharmacology. 2018;9:1336
  71. 71. Williams ML, Hata JA, Schroder J, Rampersaud E, Petrofski J, Jakoi A, et al. Targeted beta-adrenergic receptor kinase (betaARK1) inhibition by gene transfer in failing human hearts. Circulation. 2004;109(13):1590-1593
  72. 72. Cannavo A, Liccardo D, Koch WJ. Targeting cardiac β-adrenergic signaling via GRK2 inhibition for heart failure therapy. Frontiers in Physiology. 2013;4:264
  73. 73. Koch WJ, Rockman HA, Samama P, Hamilton RA, Bond RA, Milano CA, et al. Cardiac function in mice overexpressing the beta-adrenergic receptor kinase or a beta ARK inhibitor. Science. 1995;268(5215):1350-1353
  74. 74. Tachibana H, Naga Prasad SV, Lefkowitz RJ, Koch WJ, Rockman HA. Level of beta-adrenergic receptor kinase 1 inhibition determines degree of cardiac dysfunction after chronic pressure overload-induced heart failure. Circulation. 2005;111(5):591-597
  75. 75. Rengo G, Lymperopoulos A, Leosco D, Koch WJ. GRK2 as a novel gene therapy target in heart failure. Journal of Molecular and Cellular Cardiology. 2011;50(5):785-792
  76. 76. Travers JG, Kamal FA, Valiente-Alandi I, Nieman ML, Sargent MA, Lorenz JN, et al. Pharmacological and activated fibroblast targeting of Gβγ-GRK2 after myocardial ischemia attenuates heart failure progression. Journal of the American College of Cardiology. 2017;70(8):958-971
  77. 77. Sugarman MA, Loree AM, Baltes BB, Grekin ER, Kirsch I. The efficacy of paroxetine and placebo in treating anxiety and depression: A meta-analysis of change on the Hamilton Rating Scales. PLoS One. 2014;9(8):e106337-e
  78. 78. Thal DM, Yeow RY, Schoenau C, Huber J, Tesmer JJ. Molecular mechanism of selectivity among G protein-coupled receptor kinase 2 inhibitors. Molecular Pharmacology. 2011;80(2):294-303
  79. 79. Tian X, Wang Q , Guo R, Xu L, Chen QM, Hou Y. Effects of paroxetine-mediated inhibition of GRK2 expression on depression and cardiovascular function in patients with myocardial infarction. Neuropsychiatric Disease and Treatment. 2016;12:2333-2341
  80. 80. Schumacher SM, Gao E, Zhu W, Chen X, Chuprun JK, Feldman AM, et al. Paroxetine-mediated GRK2 inhibition reverses cardiac dysfunction and remodeling after myocardial infarction. Science Translational Medicine. 2015;7(277):277ra31
  81. 81. Martini JS, Raake P, Vinge LE, DeGeorge BR, Chuprun JK, Harris DM, et al. Uncovering G protein-coupled receptor kinase-5 as a histone deacetylase kinase in the nucleus of cardiomyocytes. Proceedings of the National Academy of Sciences. 2008;105(34):12457-12462
  82. 82. Gold JI, Gao E, Shang X, Premont RT, Koch WJ. Determining the absolute requirement of g protein–coupled receptor kinase 5 for pathological cardiac hypertrophy. Circulation Research. 2012;111(8):1048-1053
  83. 83. Chen EP, Bittner HB, Akhter SA, Koch WJ, Davis RD. Myocardial function in hearts with transgenic overexpression of the G protein-coupled receptor kinase 5. The Annals of Thoracic Surgery. 2001;71(4):1320-1324
  84. 84. Dzimiri N, Muiya P, Andres E, Al-Halees Z. Differential functional expression of human myocardial G protein receptor kinases in left ventricular cardiac diseases. European Journal of Pharmacology. 2004;489(3):167-177
  85. 85. Yi XP, Zhou J, Baker J, Wang X, Gerdes AM, Li F. Myocardial expression and redistribution of GRKs in hypertensive hypertrophy and failure. The Anatomical Record Part A: Discoveries in Molecular, Cellular, and Evolutionary Biology: An Official Publication of the American Association of Anatomists. 2005;282(1):13-23
  86. 86. Agüero J, Almenar L, D'Ocon P, Oliver E, Montó F, Moro J, et al. Correlation between beta-adrenoceptors and G-protein-coupled receptor kinases in pretransplantation heart failure. In: Transplantation Proceedings. Elsevier; 2008. The Journal of Heart and Lung Transplantation. DOI: 10.1016/j.healun.2009.06.003. Epub 2009 Sep 26.
  87. 87. Hullmann JE, Grisanti LA, Makarewich CA, Gao E, Gold JI, Chuprun JK, et al. GRK5-mediated exacerbation of pathological cardiac hypertrophy involves facilitation of nuclear NFAT activity. Circulation Research. 2014;115(12):976-985
  88. 88. Gold JI, Gao E, Shang X, Premont RT, Koch WJ. Determining the absolute requirement of G protein-coupled receptor kinase 5 for pathological cardiac hypertrophy: Short communication. Circulation Research. 2012;111(8):1048-1053
  89. 89. Traynham CJ, Cannavo A, Zhou Y, Vouga AG, Woodall BP, Hullmann J, et al. Differential role of G protein-coupled receptor kinase 5 in physiological versus pathological cardiac hypertrophy. Circulation Research. 2015;117(12):1001-1012
  90. 90. Islam KN, Bae J-W, Gao E, Koch WJ. Regulation of nuclear factor κB (NF-κB) in the nucleus of cardiomyocytes by G protein-coupled receptor kinase 5 (GRK5). Journal of Biological Chemistry. 2013;288(50):35683-35689
  91. 91. Gold JI, Martini JS, Hullmann J, Gao E, Chuprun JK, Lee L, et al. Nuclear translocation of cardiac G protein-coupled receptor kinase 5 downstream of select Gq-activating hypertrophic ligands is a calmodulin-dependent process. PloS One. 2013;8(3):e57324
  92. 92. Lee JH, Seo HW, Ryu JY, Lim CJ, Yi KY, Oh K-S, et al. KR-39038, a novel GRK5 inhibitor, attenuates cardiac hypertrophy and improves cardiac function in heart failure. Biomolecules & Therapeutics. 2020;28(5):482
  93. 93. Homan KT, Wu E, Cannavo A, Koch WJ, Tesmer JJ. Identification and characterization of amlexanox as a G protein-coupled receptor kinase 5 inhibitor. Molecules. 2014;19(10):16937-16949
  94. 94. Park CH, Lee JH, Lee MY, Lee JH, Lee BH, Oh K-S. A novel role of G protein-coupled receptor kinase 5 in urotensin II-stimulated cellular hypertrophy in H9c2 UT cells. Molecular and Cellular Biochemistry. 2016;422(1):151-160
  95. 95. Gao WQ , Han CG, Lu XC, Liu YX, Hui HP, Wang H. GRK 2 level in peripheral blood lymphocytes of elderly patients with acute myocardial infarction. Journal of Geriatric Cardiology. 2013;10(3):281-285
  96. 96. Santulli G, Campanile A, Spinelli L, Assante di Panzillo E, Ciccarelli M, Trimarco B, et al. G protein-coupled receptor kinase 2 in patients with acute myocardial infarction. The American Journal of Cardiology. 2011;107(8):1125-1130
  97. 97. Brinks H, Das A, Koch WJ. A role for GRK2 in myocardial ischemic injury: Indicators of a potential future therapy and diagnostic. Future Cardiology. 2011;7(4):547-556
  98. 98. Fan Q , Chen M, Zuo L, Shang X, Huang MZ, Ciccarelli M, et al. Myocardial ablation of G protein-coupled receptor kinase 2 (GRK2) decreases ischemia/reperfusion injury through an anti-intrinsic apoptotic pathway. PLoS One. 2013;8(6):e66234
  99. 99. Woodall MC, Woodall BP, Gao E, Yuan A, Koch WJ. Cardiac fibroblast GRK2 deletion enhances contractility and remodeling following ischemia/reperfusion injury. Circulation Research. 2016;119(10):1116-1127

Written By

Asma S. Alonazi, Anfal F. Bin Dayel, Tahani K. Alshammari and Nouf M. Alrasheed

Submitted: 24 April 2022 Reviewed: 13 May 2022 Published: 18 June 2022