HHS Public Access Author manuscript Author Manuscript

Maturitas. Author manuscript; available in PMC 2017 November 01. Published in final edited form as: Maturitas. 2016 November ; 93: 65–72. doi:10.1016/j.maturitas.2016.03.022.

Adrenergic signaling in heart failure and cardiovascular aging Gaetano Santulli1 and Guido Iaccarino2 1College

of Physicians & Surgeons, Columbia University Medical Center, New York, NY, USA

2Division

of Internal Medicine, Department of Medicine and Surgery, University of Salerno, Italy

Abstract Author Manuscript

Both cardiovascular disease and aging are associated with changes in the sympathetic nervous system. Indeed, mounting evidence indicates that adrenergic receptors are functionally involved in numerous processes underlying both aging and cardiovascular disorders, in particular heart failure. This article will review the pathophysiological role of the sympathetic nervous system in heart failure and cardiovascular aging.

Keywords Sympathetic nervous system; beta adrenergic receptors; aging; multimorbidity; cardiovascular system; G protein-coupled receptor kinases; heart failure

Author Manuscript

INTRODUCTION Aging is associated with a progressive decline in physiological processes. Cardiovascular disease, and heart failure in particular, remains the leading cause of death among the elderly [1]. Therefore, it is indispensable to explore the molecular mechanisms underlying agerelated cardiovascular disorders. Many of the physiological paradigms developed during the last decades of research in the biological and medical fields may reveal inadequate to understand the aging pathophysiology, in which adaptive and maladaptive mechanisms combine in a new and delicate equilibrium, a thin red line that divides the healthy aging from the fragile individual. The sympathetic nervous system is one of such mechanisms that with aging undergo a series of rearrangements, from baroreflex sensitivity to adrenergic signaling. Over the course of the

Author Manuscript

Corresponding author: Gaetano Santulli: [email protected] or [email protected]. Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. Contributors Both authors contributed significantly to the work, read the manuscript, and agreed to its submission. Conflict of interest None declared. Provenance and peer review This article has undergone peer review.

Santulli and Iaccarino

Page 2

Author Manuscript

years the sympathetic nervous system changes its ability to regulate many physiological functions including, not exclusively, the cardiovascular system. Cardiac aging is characterized by several complex modifications including diastolic dysfunction, left ventricular hypertrophy, increased risk of atrial fibrillation, valvular degeneration, leading to a decreased maximal exercise capacity. Intriguingly, the sympathetic nervous system plays a key role in both aging and cardiovascular disease. Indeed, adrenergic receptors (ARs) are involved in a variety of pathophysiological processes and mounting evidence indicates their participation in aging and cardiovascular physiopathology. The Sympathetic nervous system

Author Manuscript

The aging process is accompanied by a series of changes in the autonomic control of the cardiovascular system, favoring heightened cardiac sympathetic tone with parasympathetic withdrawal and blunted cardiovagal baroreflex sensitivity. In a study of older males, the peripheral plasma norepinephrine concentration was approximately 66% higher than that observed in younger men. This finding is explained by the reduction in the rate of clearance of norepinephrine from plasma and 29% higher whole-body norepinephrine spillover to plasma The cardiac spillover of catecholamines is higher in older than in younger men [2, 3]. Different factors can play a role in determining the rate of spillover of norepinephrine from sympathetic nerve terminals to plasma including nerve traffic, pre-junctional modulation, and the rate of catecholamine reuptake, principally via neuronal uptake. Adrenergic receptors and G protein-coupled receptor kinases

Author Manuscript

Adrenergic receptors (ARs) belong to the guanine nucleotide-binding G protein-coupled receptor (GPCR) superfamily, and are membrane receptors that activate heterotrimeric G proteins. GPCRs consist of one extracellular N-terminal domain, seven membrane-spanning domains, three intra- and three extracellular loops, and one intracellular C-terminal tail. G proteins typically stimulate (Gs protein) or inhibit (Gi protein) the enzyme adenylyl-cyclase, or activate (Gq protein) phospholipase C (PLC). GPCR signaling is stopped by the family of G protein-coupled receptor kinases (GRKs). GRK-mediated phosphorylation leads to an increased affinity of GPCRs for the arrestin class of proteins, which then uncouples the phosphorylated receptor from G protein and targets the receptor for internalization [4].

Author Manuscript

Two classes of ARs have been identified: α and β. Phenylephrine is a selective agonist of αAR while isoproterenol is a non-selective agonist for βAR [5]. The subfamily of α1AR (Gq coupled receptors) consists of three highly homologous subtypes, including α1A-, α1B- and α1D-AR [6]. The α2AR subfamily (coupled to Gi) comprises three subtypes: α2A-, α2Band α2C-AR [7]. Some species express a fourth α2D-AR [8]. In the βAR family there are three receptor subtypes: β1AR is highly expressed in the heart [9], β2AR is widely distributed throughout the body [10], and β3AR is found, although not exclusively, in the white and brown adipose tissue [11]. All βARs couple primarily to Gαs and subsequent cAMP-related pathways, however under certain conditions they can couple to Gαi [12].

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 3

Author Manuscript

Notably, β2AR and β3AR signaling can also occur via mechanisms independent from G protein [13]. Additionally, the response to GPCR stimulus can be modified by various parameters, including chronic stimulation, cell hypoxia, acidosis, and aging [14-16].

Author Manuscript

GRKs have a significant role in the regulation of adrenergic responses. Generally, GRK2 is considered the most important isoform of the 7 mammalian GRKs. Indeed mice with homozygous deletion of GRK2 exhibit embryonic lethality whereas gene ablation for the other GRKs resulted in relatively mild phenotypes [17-19]. GRK-mediated desensitization does not rely only on its catalytic activity but also on protein-protein interactions occurring in different cellular compartments [20]. Both up-regulated and reduced levels of GRK2 may affect cellular functioning. Alterations in GRK2 activity and expression have been described in numerous diseases, including myocardial infarction [21], Alzheimer’s disease [22], rheumatoid arthritis [23], multiple sclerosis [24], inflammatory pain [25, 26], opioid addiction [26], thyroid gland disorders [27], pituitary adenomas [28], ovarian cancer [29], and cystic fibrosis [30].

Author Manuscript

GRK2 levels in peripheral blood lymphocytes have been reported to mirror changes in kinase expression in other organs under several pathophysiological settings [21]. In particular, GRK2 levels and activity are increased in lymphocytes from hypertensive patients. Impairment of β-adrenergic mediated vasodilation has been reported in both human hypertensive subjects and animal models of hypertension, and such an alteration has been related to the increased GRK2 abundance and activity [31]. Decreased β-adrenergic signaling due to increased GRK2 activity would reduce the vasodilatative response, leading to high blood pressure. This view is supported by the inverse correlation of GRK2 expression with blood pressure. Additionally, data from spontaneously hypertensive rats and Dahl salt-sensitive rats confirm increased levels of GRK2 in vascular smooth muscle cells, consistent with the observations in peripheral lymphocytes [32]. As discussed below in the section dedicated to adrenergic signaling and metabolism, elevated GRK2 levels could imply metabolic alterations and lead to insulin-resistance [13, 33, 34]. To summarize, control of endothelium homeostasis relies on a complex interaction between adrenergic system and nitroxidative stress, where specific molecules as GRKs may interplay with and modulate their crosstalk. Adrenergic receptors and cardiovascular aging

Author Manuscript

The age-associated decrease in catecholamine-responsiveness in the elderly is well established. In particular, an age-related reduction in βAR sensitivity and density has been shown in the myocardium and has been attributed to down-regulation and impaired coupling of βARs to adenylyl-cyclase {Xiao, 1998 #1544}. The age-linked decline in cardiac βAR response appears to be due to a down-regulation of β1ARs, as reported in experiments in aged explanted human hearts [35]. Additionally, a decreased sensitivity of βARs, measured by isoproterenol-induced changes in the catecholamine-stimulated adenylyl-cyclase activity has been shown in the cardiovascular system [10]. βARs compartmentalization may also participate in the age-associated decreased βAR responsiveness [36]. Indeed, whereas

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 4

Author Manuscript

β1ARs are widely distributed on the plasma membrane, β2ARs are usually located in the transverse tubules, containing specialized proteins that couple membrane depolarization (excitation) to calcium-mediated myofilament shortening (contraction) [10, 37-39]. Henceforward, the localization of β2AR in cardiac cells leads to the generation of spatially restricted cAMP production, affecting calcium-dependent proteins that control the contraction of myofilaments [40, 41]. Several conditions presenting a decreased myocardial function can elicit activity from the sympathetic nervous system (SNS) that eventually diverts blood flow to critical organs and increases cardiac output. The main players involved in such system are catecholamines, whose release is strictly orchestrated by the GPCR system, relating the adrenal gland and the heart [42].

Author Manuscript

Most of the modifications in the sympathetic nervous system occurring with aging, including decreased βAR responsiveness, increased circulating catecholamines, and overall hyposensitivity to adrenergic stress, are also observed in patients with failing hearts. Moreover, young people are more reactive to isoproterenol-induced increase in blood flow than elderly subjects [31].

Author Manuscript

Vascular tone is finely regulated by the intimal (endothelial cells, EC) and the medial (vascular smooth muscle cells, VSMC) layers [43-45] and both EC and VSMC express β2AR [46-48]. The age-related decline in β2AR function and successive cAMP generation is observed in diverse cardiovascular disorders, including atherosclerosis, hypertension, vascular insufficiency, and orthostatic hypotension, all conditions with significant mortality and morbidity [49-53]. The increased incidence of restenosis and atherosclerosis in aged people may also rely on the age-associated deterioration in βAR-mediated cAMP production, since cAMP may inhibit VSMC proliferation [45, 54]. A reduced responsiveness with age has been also reported for αAR in healthy subjects [55], with potential implications for reduced muscle blood flow and augmented blood pressure during exercise [56-58].

Author Manuscript

During the late 90’s the idea that the EC possessed functional βARs became more and more evident, and soon this system was seen as a therapeutic target [59, 60]. Nevertheless, the proangiogenic effects of the β2AR was firstly identified by our group in 2005 [47]. The picture was completed, when we finally demonstrated that the endothelium is the source of catecholamines that stimulate in a paracrine manner endothelial βARs [46]. These findings were further corroborated by the observation that also endothelial progenitor cells express β2AR and that the ability of this receptor to signaling to eNOS promotes angiogenesis through multiple mechanisms [61]. This ability is lost during aging. Strategies that aim to restore β2AR signaling in models where it is lost are effective to restore also impaired angiogenesis [47, 62]. For instance, in aging the enhancement of βAR signaling leads to the correction of impaired angiogenesis [10, 63, 64]. The overall incidence of cardiac disorders including heart failure, left ventricular hypertrophy, and arrhythmias, increases considerably with age [1, 65]. Elderly people appear to be particularly predisposed to the development of heart failure: such a diagnosis is the leading cause of hospitalizations in people >65 years of age [1, 15]. Additionally, alterations in ventricular relaxation and filling have been described with aging [64, 66].

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 5

Author Manuscript

The prevalence of atrial fibrillation reaches values of 17.8% in people >85 years [1, 67, 68]. The development of a non-regular pattern of electrical activity might have detrimental consequences in hearts that are relatively stiff and relax slowly [1, 69, 70]. Likewise, the prevalence of left ventricular hypertrophy increases with rising blood pressure and body mass index [71, 72] and studies in normotensive people indicate that myocardial wall thickness increases progressively with age [73]. In the vasculature an age-linked increase in intimal thickening has been reported, accompanied by luminal dilatation and reduction in distensibility, resulting in increased vessel stiffness. Indeed, pulse wave velocity, a noninvasive index of vascular stiffening, increases with age and has been associated with structural alterations in the media [13, 44, 48, 49, 64, 74-76].

Author Manuscript

Adrenergic signaling in heart failure The SNS has prominent effects on cardiac physiology, including increases in atrioventricular conduction (positive dromotropy), heart rate (positive chronotropy), cardiac contractility (positive inotropy), and cardiac relaxation (positive lusitropy). Crucially, the SNS plays a crucial role in the regulation of vascular tone by controlling both peripheral resistance and cardiac output [77, 78].

Author Manuscript Author Manuscript

Heart failure is a chronic syndrome in which the heart is unable of pumping a adequate supply of blood to meet the metabolic requirements of the body or generating the required elevated ventricular filling pressures to maintain output [34]. Despite considerable advances in the treatment of heart failure, such a disease still represents a severe social and clinical burden [42, 79, 80]. The cardiovascular system is best viewed as a complex dynamic system, continually adapting to optimize organ perfusion. During heart failure, diverse neurohormonal mechanisms are triggered to maintain cardiac output [4]. Heart failure is considered a progressive disease that begins long before signs or symptoms become clinically evident: initially there is a complex adaptive neurohormonal activation - required to compensate for cardiac dysfunction - which includes nervous system, renin-angiotensinaldosterone system, endothelin, natriuretic peptides, and vasopressin. The process progressively becomes maladaptive, leading to increased mechanical stress on the failing heart and causing harmful electrical and structural events [81-83]. Thus, β-blockers, Angiotensin II AT1 receptor blockers, angiotensin-converting enzyme inhibitors, and mineralocorticoid receptor antagonists represent cornerstones for the treatment of patients with heart failure. Given the decreased myocardial contractility observed in heart failure and the positive inotropic response obtained with βAR agonists, it was initially attractive to attempt to overcome the βAR desensitization in order to ameliorate cardiac function via the stimulation of the adrenergic system [84]. Several authors also provided experimental evidence that β2AR overexpression might have been a new treatment for heart failure [85, 86]. Other investigators demonstrated that the overexpression of β1AR [87] or β2AR [88] leads to cardiomyopathy and heart failure, as reflected by decreased cardiac function, increased fibrosis, cardiomyocyte apoptosis, and overall mortality.

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 6

Author Manuscript

The central part of the adrenal gland, called adrenal medulla, is the main source of catecholamines and is comprised of groups of adrenergic and noradrenergic chromaffin cells and, to a lesser extent, ganglionic neurons [42, 89]. The adrenal gland can be seen as a specialized sympathetic ganglion, receiving inputs from the sympathetic nervous system via pre-ganglionic fibers. Yet, the adrenal gland directly secretes neurohormones into the bloodstream [89]. The sympathetic overdrive observed in heart failure correlates with a higher risk of arrhythmias and left ventricular dysfunction [90]. Moreover, augmented levels of catecholamines can cause myocardial damage via enhanced cardiac oxygen demand and by increasing peroxidative metabolism [91] and ultimately leading to structural alterations, including focal necrosis and inflammation, increased collagen deposition, and interstitial fibrosis [92-94].

Author Manuscript

Systemically circulating or locally released catecholamines trigger two main classes of ARs: α1AR and β2AR, causing vasoconstriction and vasodilatation, respectively [46, 47, 95]. With aging, such a fine equilibrium is progressively shifted toward increased vasoconstriction, most likely due to a defective vasodilatation in response to βAR stimulation. Increased basal levels of circulating catecholamines have been observed both in heart failure and with advancing age, mirrored by a decrease in the number of high-affinity βARs. These findings suggest that these alterations might be attributable to βAR desensitization rather than an actual reduction in βAR density [10, 21, 96].

Author Manuscript

Both expression and activity of GRK2 increase in vascular tissue with aging [97]. Equally important, an impairment in βAR-mediated vasorelaxation has been observed in hypertensive patients [31] and in animal models of hypertension [75, 97]: such an alteration has been related to increased GRK2 abundance and activity. Transgenic overexpression of GRK2 in the vasculature impairs βAR signaling and the vasodilatative response, eliciting a hypertensive phenotype in rodents. This aspect has been confirmed in humans: GRK2 expression correlates with blood pressure and impaired βAR-mediated adenylyl-cyclase activity [97]. Additionally, variants of the gene encoding for β2AR have been associated with longevity [10].

Author Manuscript

The decreased βAR-mediated response has been attributed to different mechanisms, including an attenuation of PKA activation, an impaired generation of cyclic AMP, a reduced receptor density, and a less efficient coupling to adenylyl-cyclase [10]. However, currently there is no single molecular or cellular factor that can fully explain the decline in βAR function. Nonetheless, the etiology seems to be most likely associated with alterations in the ability of βAR to respond to agonists at the cellular level. Adrenergic signaling and metabolism The adrenergic system is involved in regulating several metabolic pathways. Increased circulating catecholamines and activation of the different ARs present in the various organs produce important metabolic responses, including increased gluconeogenesis by the liver to

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 7

Author Manuscript

provide substrate for the brain, increased lipolysis and elevated levels of fatty acids in plasma, and modulation of insulin secretion by pancreatic islets of Langerhans. Such responses are detrimental to the functioning of different organs (e.g. the heart). Metabolic modifications including insulin resistance, altered glucose and lipid metabolism, and mitochondrial dysfunction represent common features of many conditions involving adrenergic overdrive. Notably, these alterations are seen in a number of different pathological conditions and are generally highly correlated with the level of activation of the sympathetic system.

Author Manuscript

Chronic βAR stimulation induces insulin resistance and in this context the β2AR has a key role in overall glucose homeostasis by modulating pancreatic islet hormone secretion as well as liver and muscle glucose homeostasis. Short- and long-term stimulation of the β2AR has been associated with the modulation of fatty acid and glucose metabolism [98]. Indeed, acute treatment with β2AR agonists of myocytes or skeletal muscle increases glucose uptake to levels comparable to those seen after insulin stimulation [99].

Author Manuscript

A potential mechanism for β2AR function in insulin resistance involves the activation of PI3K and its downstream signal pathway and in particular the phosphorylation and inactivation of TBC1D4 by AKT [100]. TBC1D4 inhibits the translocation to the plasma membrane of the glucose transporter type 4 (GLUT4) [100]. Moreover, TBC1D4 is also targeted by AMPK, which represents a pivotal mechanism in the regulation of insulinindependent glucose uptake [100, 101]. Strikingly, higher levels of AMPK phosphorylation and activity are seen in response to βAR stimulation [102, 103] as a result of changes in the AMP/ATP ratio or activation of upstream AMPK kinases [104]. Besides, in vivo studies show a greater efficiency of carvedilol, a non-selective βAR antagonist, in ameliorating myocardial insulin sensitivity and glucose extraction in an animal model of heart failure, compared to the selective β1AR antagonist metoprolol [105]. Chronic adrenergic stimulation, as seen during heart failure, would be detrimental by mechanisms involving mechanisms such as JNK, β-arrestins and GRKs [4, 106]. Insulin resistance highly correlates with adrenergic function [75, 107-111]. In both type 2 diabetes mellitus (T2DM) and heart failure circulating insulin levels are elevated, causing a persistent stimulation of insulin receptors [52, 112-117]. Hyperactive insulin signaling can accelerate adverse left ventricular remodeling [72, 118]. Recently, insulin has been demonstrated to directly impair adrenergic pathways for contractile function via an insulin receptor/β2AR signaling complex [114].

Author Manuscript

In numerous conditions associated with insulin resistance, such as hypertension and T2DM, there are elevated GRK2 levels [97, 119]. In vitro experiments demonstrated that insulin increases GRK2 levels, causing GRK2–IRS1 association [97, 120]. On these bases, GRK2 inhibition has been proposed to be beneficial. Indeed, chronic treatment of spontaneously hypertensive rats with an inhibitor of GRK2 kinase activity ameliorates glucose homeostasis and decreases blood pressure [120]. Growing evidence indicates that GRKs can exert different effects within the cell depending on the stimulus, cell type, and localization [97, 121]. In this sense, we were the first to

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 8

Author Manuscript

demonstrate a mitochondrial localization for GRK2 [122], later confirmed by other investigators [123], establishing a functional role for GRK2 in organelle biogenesis and ATP production [122, 124]. Sympathetic system and remodeling Histopathologic evaluation in murine hearts reveals subendocardial and interstitial fibrosis, vacuolization of cytoplasm and mineralization [16, 125-127]. Furthermore, morphometric analysis of cardiomyocytes demonstrates a progressive hypertrophy, increased apoptosis alongside with fibrosis and amyloid deposition [128].

Author Manuscript

Several organs undergo fibrotic remodeling as a function of age resulting in overall decreased functionality. The precise mechanisms leading to the age-dependent accumulation of collagen have yet to be fully identified. Mounting evidence reveals the emerging role of collagen cross-linkers and matrix metalloproteinases (MMP) in the turnover of collagens, finely regulated by proteolytic MMP activity and their endogenous tissue inhibitors (TIMPs) [129, 130]. All of these mechanisms are modulated by neuroendocrine activation and βAR signal transduction modifications. Indeed, chronic sympathetic activation significantly contributes to progression from compensated left ventricular hypertrophy to myocardial dysfunction through detrimental cardiac matrix remodeling. Besides, sympathetic overactivity achieved via β1AR overexpression (β1TG mouse) [87, 131] is accompanied by interstitial matrix remodeling and turnover by inducing MMP/TIMPs. In β1TG pro-collagen type-I and type-III mRNA and interstitial collagen protein expression is progressively increased from four-fold at 5 months to 17-fold at 12 months compared to 3 months of age. In β1TG mice at 5 months of age with compensated cardiac hypertrophy, an increased deposition of type-I and -III collagen fibers surrounding the cardiomyocytes is observed [131]. Contrariwise, the β1TG group at 12 months of age exhibits an asymmetrical distribution of the collagen fibers with disruption of the collagen network structure [132, 133].

Author Manuscript

A positive correlation between MMP-2 and noradrenaline in patients with heart failure has been demonstrated, also corroborated by in vitro experiments in human cardiac fibroblasts [134], supporting thereby the link between sympathetic stimulation and collagen turnover. Conclusion

Author Manuscript

With aging the sympathetic nervous system undergoes a series of rearrangements, from baroreflex sensitivity to adrenergic signaling, and over the course of the years, it changes its ability to regulate countless physiological functions. The exaggerated sympathetic activity observed in aging, discussed in this report, has to be put into the perspective of a whole scenario, in which the effectors of catecholamines are also undergoing a plasticity with the ultimate intent of allowing an adequate signaling.

Acknowledgments Funding Dr. Gaetano Santulli, MD, PhD is supported by the National Institutes of Health (K99DK107895).

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 9

Author Manuscript

References

Author Manuscript Author Manuscript Author Manuscript

[1]. Santulli G. Epidemiology of cardiovascular disease in the 21st century: updated numbers and updated facts. Journal of Cardiovascular Disease (JCvD). 2013; 1(1):1–2. [2]. Esler M, Lambert G, Kaye D, Rumantir M, Hastings J, Seals DR. Influence of ageing on the sympathetic nervous system and adrenal medulla at rest and during stress. Biogerontology. 2002; 3(1-2):45–9. [PubMed: 12014841] [3]. Kaye D, Esler M. Sympathetic neuronal regulation of the heart in aging and heart failure. Cardiovascular research. 2005; 66(2):256–64. [PubMed: 15820194] [4]. Hatton R, Cvjeticanin A, Lymperopoulos A. The adrenergic system of the adrenal glands as a remote control of cardiac function. JCvD. 2015; 5(3):394–397. [5]. Mei Y, Yin N, Jin X, He J, Yin Z. The regulatory role of the adrenergic agonists phenylephrine and isoproterenol on fetal hemoglobin expression and erythroid differentiation. Endocrinology. 2013; 154(12):4640–9. [PubMed: 24080366] [6]. Lampri, E.; Ioachim, E. Angiogenesis: Something Old, Something New. In: Santulli, Gaetano, editor. Angiogenesis: Insight from a systematic overview. Nova Science Publishers; 2013. p. 1-30. [7]. O’Connell TD, Jensen BC, Baker AJ, Simpson PC. Cardiac alpha1-adrenergic receptors: novel aspects of expression, signaling mechanisms, physiologic function, and clinical importance. Pharmacological reviews. 2014; 66(1):308–33. [PubMed: 24368739] [8]. Ruuskanen JO, Laurila J, Xhaard H, Rantanen VV, Vuoriluoto K, Wurster S, Marjamaki A, Vainio M, Johnson MS, Scheinin M. Conserved structural, pharmacological and functional properties among the three human and five zebrafish alpha 2-adrenoceptors. Br J Pharmacol. 2005; 144(2): 165–77. [PubMed: 15655522] [9]. Vicco MH, Pujato N, Bontempi I, Rodeles L, Marcipar I, Bottasso OA. beta1-Selective Adrenoceptor Antagonists Increase Plasma Levels of Anti-p2beta Antibodies and Decrease Cardiac Involvement in Chronic Progressive Chagas Heart Disease. Can J Cardiol. 2014; 30(3): 332–7. [PubMed: 24370375] [10]. Santulli G, Iaccarino G. Pinpointing beta adrenergic receptor in ageing pathophysiology: victim or executioner? Evidence from crime scenes. Immunity & ageing: I & A. 2013; 10(1):10. [PubMed: 23497413] [11]. Belge C, Hammond J, Dubois-Deruy E, Manoury B, Hamelet J, Beauloye C, Markl A, Pouleur AC, Bertrand L, Esfahani H, Jnaoui K, Gotz KR, Nikolaev VO, Vanderper A, Herijgers P, Lobysheva I, Iaccarino G, Hilfiker-Kleiner D, Tavernier G, Langin D, Dessy C, Balligand JL. Enhanced expression of beta3-adrenoceptors in cardiac myocytes attenuates neurohormoneinduced hypertrophic remodeling through nitric oxide synthase. Circulation. 2014; 129(4):451– 62. [PubMed: 24190960] [12]. Xiao RP. Beta-adrenergic signaling in the heart: dual coupling of the beta2-adrenergic receptor to G(s) and G(i) proteins. Science’s STKE: signal transduction knowledge environment. 2001; 2001(104):re15. [PubMed: 11604549] [13]. Santulli G, Lombardi A, Sorriento D, Anastasio A, Del Giudice C, Formisano P, Beguinot F, Trimarco B, Miele C, Iaccarino G. Age-related impairment in insulin release: the essential role of beta(2)-adrenergic receptor. Diabetes. 2012; 61(3):692–701. [PubMed: 22315324] [14]. Kaya AI, Onaran HO, Ozcan G, Ambrosio C, Costa T, Balli S, Ugur O. Cell contact-dependent functional selectivity of beta2-adrenergic receptor ligands in stimulating cAMP accumulation and extracellular signal-regulated kinase phosphorylation. The Journal of biological chemistry. 2012; 287(9):6362–74. [PubMed: 22241475] [15]. Santulli G, Marks AR. Essential Roles of Intracellular Calcium Release Channels in Muscle, Brain, Metabolism, and Aging. Curr Mol Pharmacol. 2015; 8(2):206–22. [PubMed: 25966694] [16]. Yuan Q, Chen Z, Santulli G, Gu L, Yang ZG, Yuan ZQ, Zhao YT, Xin HB, Deng KY, Wang SQ, Ji G. Functional Role of Calstabin2 in Age-related Cardiac Alterations. Scientific reports. 2014; 4:7425. [PubMed: 25502776]

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 10

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

[17]. Gainetdinov RR, Bohn LM, Sotnikova TD, Cyr M, Laakso A, Macrae AD, Torres GE, Kim KM, Lefkowitz RJ, Caron MG, Premont RT. Dopaminergic supersensitivity in G protein-coupled receptor kinase 6-deficient mice. Neuron. 2003; 38(2):291–303. [PubMed: 12718862] [18]. Lyubarsky AL, Chen C, Simon MI, Pugh EN Jr. Mice lacking G-protein receptor kinase 1 have profoundly slowed recovery of cone-driven retinal responses. J Neurosci. 2000; 20(6):2209–17. [PubMed: 10704496] [19]. Walker JK, Gainetdinov RR, Feldman DS, McFawn PK, Caron MG, Lefkowitz RJ, Premont RT, Fisher JT. G protein-coupled receptor kinase 5 regulates airway responses induced by muscarinic receptor activation. Am J Physiol Lung Cell Mol Physiol. 2004; 286(2):L312–9. [PubMed: 14565944] [20]. Fusco A, Santulli G, Sorriento D, Cipolletta E, Garbi C, Dorn GW 2nd, Trimarco B, Feliciello A, Iaccarino G. Mitochondrial localization unveils a novel role for GRK2 in organelle biogenesis. Cell Signal. 2011; 24(2):468–75. [PubMed: 21983013] [21]. Santulli G, Campanile A, Spinelli L, Assante di Panzillo E, Ciccarelli M, Trimarco B, Iaccarino G. G protein-coupled receptor kinase 2 in patients with acute myocardial infarction. Am J Cardiol. 2011; 107(8):1125–30. [PubMed: 21296320] [22]. Suo Z, Wu M, Citron BA, Wong GT, Festoff BW. Abnormality of G-protein-coupled receptor kinases at prodromal and early stages of Alzheimer’s disease: an association with early betaamyloid accumulation. J Neurosci. 2004; 24(13):3444–52. [PubMed: 15056724] [23]. Lombardi MS, Kavelaars A, Schedlowski M, Bijlsma JW, Okihara KL, Van de Pol M, Ochsmann S, Pawlak C, Schmidt RE, Heijnen CJ. Decreased expression and activity of G-protein-coupled receptor kinases in peripheral blood mononuclear cells of patients with rheumatoid arthritis. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 1999; 13(6):715–25. [PubMed: 10094932] [24]. Vroon A, Kavelaars A, Limmroth V, Lombardi MS, Goebel MU, Van Dam AM, Caron MG, Schedlowski M, Heijnen CJ. G protein-coupled receptor kinase 2 in multiple sclerosis and experimental autoimmune encephalomyelitis. Journal of immunology. 2005; 174(7):4400–6. [25]. Singhmar P, Huo X, Eijkelkamp N, Berciano SR, Baameur F, Mei FC, Zhu Y, Cheng X, Hawke D, Mayor F Jr. Murga C, Heijnen CJ, Kavelaars A. Critical role for Epac1 in inflammatory pain controlled by GRK2-mediated phosphorylation of Epac1. Proceedings of the National Academy of Sciences of the United States of America. 2016 [26]. Zhang J, Ferguson SS, Law PY, Barak LS, Caron MG. Agonist-specific regulation of delta-opioid receptor trafficking by G protein-coupled receptor kinase and beta-arrestin. J Recept Signal Transduct Res. 1999; 19(1-4):301–13. [PubMed: 10071766] [27]. Metaye T, Menet E, Guilhot J, Kraimps JL. Expression and activity of g protein-coupled receptor kinases in differentiated thyroid carcinoma. The Journal of clinical endocrinology and metabolism. 2002; 87(7):3279–86. [PubMed: 12107237] [28]. Gatto F, Feelders R, van der Pas R, Kros JM, Dogan F, van Koetsveld PM, van der Lelij AJ, Neggers SJ, Minuto F, de Herder W, Lamberts SW, Ferone D, Hofland LJ. beta-Arrestin 1 and 2 and G protein-coupled receptor kinase 2 expression in pituitary adenomas: role in the regulation of response to somatostatin analogue treatment in patients with acromegaly. Endocrinology. 2013; 154(12):4715–25. [PubMed: 24169548] [29]. King DW, Steinmetz R, Wagoner HA, Hannon TS, Chen LY, Eugster EA, Pescovitz OH. Differential expression of GRK isoforms in nonmalignant and malignant human granulosa cells. Endocrine. 2003; 22(2):135–42. [PubMed: 14665717] [30]. Roux J, McNicholas CM, Carles M, Goolaerts A, Houseman BT, Dickinson DA, Iles KE, Ware LB, Matthay MA, Pittet JF. IL-8 inhibits cAMP-stimulated alveolar epithelial fluid transport via a GRK2/PI3K-dependent mechanism. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2013; 27(3):1095–106. [PubMed: 23221335] [31]. Izzo R, Cipolletta E, Ciccarelli M, Campanile A, Santulli G, Palumbo G, Vasta A, Formisano S, Trimarco B, Iaccarino G. Enhanced GRK2 expression and desensitization of betaAR vasodilatation in hypertensive patients. Clin Transl Sci. 2008; 1(3):215–20. [PubMed: 20443852] [32]. Gros R, Chorazyczewski J, Meek MD, Benovic JL, Ferguson SS, Feldman RD. G-Proteincoupled receptor kinase activity in hypertension: increased vascular and lymphocyte G-protein

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 11

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

receptor kinase-2 protein expression. Hypertension. 2000; 35(1 Pt 1):38–42. [PubMed: 10642272] [33]. De Boer MP, Meijer RI, Wijnstok NJ, Jonk AM, Houben AJ, Stehouwer CD, Smulders YM, Eringa EC, Serne EH. Microvascular dysfunction: a potential mechanism in the pathogenesis of obesity-associated insulin resistance and hypertension. Microcirculation. 2012; 19(1):5–18. [PubMed: 21883642] [34]. Santulli, G. The Adrenergic System in Cardiovascular Metabolism and Aging. In: Lymperopoulos, A., editor. The Cardiovascular Adrenergic System. Springer; 2015. p. 97-116. [35]. White M, Roden R, Minobe W, Khan MF, Larrabee P, Wollmering M, Port JD, Anderson F, Campbell D, Feldman AM, et al. Age-related changes in beta-adrenergic neuroeffector systems in the human heart. Circulation. 1994; 90(3):1225–38. [PubMed: 8087932] [36]. Nakou ES, Parthenakis FI, Kallergis EM, Marketou ME, Nakos KS, Vardas PE. Healthy aging and myocardium: A complicated process with various effects in cardiac structure and physiology. International journal of cardiology. 2016; 209:167–175. [PubMed: 26896615] [37]. Xie W, Santulli G, Guo X, Gao M, Chen BX, Marks AR. Imaging atrial arrhythmic intracellular calcium in intact heart. Journal of molecular and cellular cardiology. 2013; 64:120–3. [PubMed: 24041536] [38]. Umanskaya A, Santulli G, Xie W, Andersson DC, Reiken SR, Marks AR. Genetically enhancing mitochondrial antioxidant activity improves muscle function in aging. Proceedings of the National Academy of Sciences of the United States of America. 2014; 111(42):15250–5. [PubMed: 25288763] [39]. Santulli G, Xie W, Reiken SR, Marks AR. Mitochondrial calcium overload is a key determinant in heart failure. Proceedings of the National Academy of Sciences of the United States of America. 2015; 112(36):11389. [PubMed: 26217001] [40]. Kirk JA, Holewinski RJ, Kooij V, Agnetti G, Tunin RS, Witayavanitkul N, de Tombe PP, Gao WD, Van Eyk J, Kass DA. Cardiac resynchronization sensitizes the sarcomere to calcium by reactivating GSK-3beta. The Journal of clinical investigation. 2014; 124(1):129–38. [PubMed: 24292707] [41]. Wright PT, Nikolaev VO, O’Hara T, Diakonov I, Bhargava A, Tokar S, Schobesberger S, Shevchuk AI, Sikkel MB, Wilkinson R, Trayanova NA, Lyon AR, Harding SE, Gorelik J. Caveolin-3 regulates compartmentation of cardiomyocyte beta2-adrenergic receptor-mediated cAMP signaling. Journal of molecular and cellular cardiology. 2014; 67:38–48. [PubMed: 24345421] [42]. Santulli G. Adrenal signaling in heart failure: something more than a distant ship’s smoke on the horizon. Hypertension. 2014; 63(2):215–6. [PubMed: 24218430] [43]. Santulli G, Basilicata MF, De Simone M, Del Giudice C, Anastasio A, Sorriento D, Saviano M, Del Gatto A, Trimarco B, Pedone C, Zaccaro L, Iaccarino G. Evaluation of the anti-angiogenic properties of the new selective alphaVbeta3 integrin antagonist RGDechiHCit. J Transl Med. 2011; 9:7. [PubMed: 21232121] [44]. Santulli G, Wronska A, Kunihiro U, Diacovo T, Gao M, Marx S, Kitajewski J, Chilton J, Kemal M, Tuschl T, Marks AR, Totary Jain H. A selective microRNA-based strategy inhibits restenosis while preserving endothelial function. The Journal of clinical investigation. 2014; 124:4102– 4114. [PubMed: 25133430] [45]. Santulli G. microRNAs Distinctively Regulate Vascular Smooth Muscle and Endothelial Cells: Functional Implications in Angiogenesis, Atherosclerosis, and In-Stent Restenosis. Adv Exp Med Biol. 2015; 887:53–77. [PubMed: 26662986] [46]. Sorriento D, Santulli G, Del Giudice C, Anastasio A, Trimarco B, Iaccarino G. Endothelial cells are able to synthesize and release catecholamines both in vitro and in vivo. Hypertension. 2012; 60(1):129–36. [PubMed: 22665130] [47]. Iaccarino G, Ciccarelli M, Sorriento D, Galasso G, Campanile A, Santulli G, Cipolletta E, Cerullo V, Cimini V, Altobelli GG, Piscione F, Priante O, Pastore L, Chiariello M, Salvatore F, Koch WJ, Trimarco B. Ischemic neoangiogenesis enhanced by beta2-adrenergic receptor overexpression: a novel role for the endothelial adrenergic system. Circ Res. 2005; 97(11):1182– 9. [PubMed: 16239589]

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 12

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

[48]. Santulli G. microRNAs and Endothelial (Dys) Function. J Cell Physiol. 2015 [49]. Santulli G, Cipolletta E, Sorriento D, Del Giudice C, Anastasio A, Monaco S, Maione AS, Condorelli G, Puca A, Trimarco B, Illario M, Iaccarino G. CaMK4 Gene Deletion Induces Hypertension. J Am Heart Assoc. 2012; 1(4):e001081. [PubMed: 23130158] [50]. Santulli G. Coronary heart disease risk factors and mortality. JAMA: the journal of the American Medical Association. 2012; 307(11):1137. [PubMed: 22436947] [51]. Vu TH, Stamler J, Liu K, McDermott MM, Lloyd-Jones DM, Pirzada A, Garside DB, Daviglus ML. Prospective relationship of low cardiovascular risk factor profile at younger ages to anklebrachial index: 39-year follow-up-the chicago healthy aging study. J Am Heart Assoc. 2012; 1(6):e001545. [PubMed: 23316312] [52]. Sardu C, Marfella R, Santulli G. Impact of diabetes mellitus on the clinical response to cardiac resynchronization therapy in elderly people. Journal of cardiovascular translational research. 2014; 7(3):362–8. [PubMed: 24500410] [53]. Ciccarelli M, Finelli R, Rivera N, Santulli G, Izzo R, De Luca N, Rozza F, Ceccarelli M, Pagnotta S, Uliano F, Tremigliozzi R, Condorelli G, Trimarco V, Iaccarino G. The possible role of chromosome X variability in hypertensive familiarity. J Hum Hypertens. 2016 [54]. Begum N, Shen W, Manganiello V. Role of PDE3A in regulation of cell cycle progression in mouse vascular smooth muscle cells and oocytes: implications in cardiovascular diseases and infertility. Current opinion in pharmacology. 2011; 11(6):725–9. [PubMed: 22051884] [55]. Lavi S, Nevo O, Thaler I, Rosenfeld R, Dayan L, Hirshoren N, Gepstein L, Jacob G. Effect of aging on the cardiovascular regulatory systems in healthy women. American journal of physiology. Regulatory, integrative and comparative physiology. 2007; 292(2):R788–93. [56]. Menotti A, Lanti M, Mancini M, Zanchetti A, Laurenzi M, Terradura Vagnarelli O, Puddu P. Moderate Leisure Time Physical Activity and Occurrence of Cardiovascular Events: A 10-year Follow-up in the Gubbio Population Study. JCvD. 2014; 2(5):218–224. [57]. Ross AJ, Gao Z, Pollock JP, Leuenberger UA, Sinoway LI, Muller MD. beta-Adrenergic receptor blockade impairs coronary exercise hyperemia in young men but not older men. American journal of physiology. Heart and circulatory physiology. 2014; 307(10):H1497–503. [PubMed: 25239806] [58]. Huang CJ, Slusher AL, Whitehurst M, Wells M, Mock JT, Maharaj A, Shibata Y. Acute aerobic exercise mediates G protein-coupled receptor kinase 2 expression in human PBMCs. Life sciences. 2015; 135:87–91. [PubMed: 26092485] [59]. Maffei A, Vecchione C, Aretini A, Poulet R, Bettarini U, Gentile MT, Cifelli G, Lembo G. Characterization of nitric oxide release by nebivolol and its metabolites. Am J Hypertens. 2006; 19(6):579–86. [PubMed: 16733229] [60]. Maffei A, Lembo G. Nitric oxide mechanisms of nebivolol. Ther Adv Cardiovasc Dis. 2009; 3(4):317–27. [PubMed: 19443516] [61]. De Rosa R, Galasso G, Piscione F, Santulli G, Pierri A, Sorriento D, Iaccarino G, Trimarco B, Chiariello M. Beta2-adrenergic receptor improves the endothelial progenitor cells angiogenic function. Eur Heart J. 2013; 30(S1):353. [62]. Ciccarelli M, Sorriento D, Cipolletta E, Santulli G, Fusco A, Zhou RH, Eckhart AD, Peppel K, Koch WJ, Trimarco B, Iaccarino G. Impaired neoangiogenesis in beta(2)-adrenoceptor genedeficient mice: restoration by intravascular human beta(2)-adrenoceptor gene transfer and role of NFkappaB and CREB transcription factors. British journal of pharmacology. 2011; 162(3):712– 21. [PubMed: 20958287] [63]. Ciccarelli M, Santulli G, Pascale V, Trimarco B, Iaccarino G. Adrenergic receptors and metabolism: role in development of cardiovascular disease. Frontiers in physiology. 2013; 4:265. [PubMed: 24106479] [64]. Santulli G, Ciccarelli M, Trimarco B, Iaccarino G. Physical activity ameliorates cardiovascular health in elderly subjects: the functional role of the beta adrenergic system. Frontiers in physiology. 2013; 4:209. [PubMed: 23964243] [65]. Sorriento D, Santulli G, Franco A, Cipolletta E, Napolitano L, Gambardella J, Gomez-Monterrey I, Campiglia P, Trimarco B, Iaccarino G, Ciccarelli M. Integrating GRK2 and NFkappaB in the

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 13

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Pathophysiology of Cardiac Hypertrophy. Journal of cardiovascular translational research. 2015; 8(8):493–502. [PubMed: 26224342] [66]. Czuriga D, Paulus WJ, Czuriga I, Edes I, Papp Z, Borbely A. Cellular mechanisms for diastolic dysfunction in the human heart. Current pharmaceutical biotechnology. 2012; 13(13):2532–8. [PubMed: 22280428] [67]. Xie W, Santulli G, Reiken SR, Yuan Q, Osborne BW, Chen BX, Marks AR. Mitochondrial oxidative stress promotes atrial fibrillation. Scientific reports. 2015; 5:11427. [PubMed: 26169582] [68]. Santulli G, Iaccarino G, De Luca N, Trimarco B, Condorelli G. Atrial fibrillation and microRNAs. Frontiers in physiology. 2014; 5:15. [PubMed: 24478726] [69]. D’Ascia SL, D’Ascia C, Marino V, Lombardi A, Santulli R, Chiariello M, Santulli G. Cardiac resynchronisation therapy response predicts occurrence of atrial fibrillation in non-ischaemic dilated cardiomyopathy. International journal of clinical practice. 2011; 65(11):1149–55. [PubMed: 21995693] [70]. Santulli G, D’Ascia S L, D’Ascia C. Development of atrial fibrillation in recipients of cardiac resynchronization therapy: the role of atrial reverse remodelling. The Canadian journal of cardiology. 2012; 28(2):245 e17. [PubMed: 22244772] [71]. Lanni F, Santulli G, Izzo R, Rubattu S, Zanda B, Volpe M, Iaccarino G, Trimarco B. The Pl(A1/A2) polymorphism of glycoprotein IIIa and cerebrovascular events in hypertension: increased risk of ischemic stroke in high-risk patients. J Hypertens. 2007; 25(3):551–6. [PubMed: 17278970] [72]. Sorriento D, Santulli G, Fusco A, Anastasio A, Trimarco B, Iaccarino G. Intracardiac injection of AdGRK5-NT reduces left ventricular hypertrophy by inhibiting NF-kappaB-dependent hypertrophic gene expression. Hypertension. 2010; 56(4):696–704. [PubMed: 20660817] [73]. Hees PS, Fleg JL, Lakatta EG, Shapiro EP. Left ventricular remodeling with age in normal men versus women: novel insights using three-dimensional magnetic resonance imaging. The American journal of cardiology. 2002; 90(11):1231–6. [PubMed: 12450604] [74]. Cantini C, Kieffer P, Corman B, Liminana P, Atkinson J, Lartaud-Idjouadiene I. Aminoguanidine and aortic wall mechanics, structure, and composition in aged rats. Hypertension. 2001; 38(4): 943–8. [PubMed: 11641314] [75]. Iaccarino G, Ciccarelli M, Sorriento D, Cipolletta E, Cerullo V, Iovino GL, Paudice A, Elia A, Santulli G, Campanile A, Arcucci O, Pastore L, Salvatore F, Condorelli G, Trimarco B. AKT participates in endothelial dysfunction in hypertension. Circulation. 2004; 109(21):2587–93. [PubMed: 15136501] [76]. Santulli G, Pagano G, Sardu C, Xie W, Reiken S, D’Ascia SL, Cannone M, Marziliano N, Trimarco B, Guise TA, Lacampagne A, Marks AR. Calcium release channel RyR2 regulates insulin release and glucose homeostasis. The Journal of clinical investigation. 2015; 125(5): 1968–78. [PubMed: 25844899] [77]. Heinonen I, Wendelin-Saarenhovi M, Kaskinoro K, Knuuti J, Scheinin M, Kalliokoski KK. Inhibition of alpha-adrenergic tone disturbs the distribution of blood flow in the exercising human limb. Am J Physiol Heart Circ Physiol. 2013; 305(2):H163–72. [PubMed: 23666670] [78]. Perino A, Ghigo A, Ferrero E, Morello F, Santulli G, Baillie GS, Damilano F, Dunlop AJ, Pawson C, Walser R, Levi R, Altruda F, Silengo L, Langeberg LK, Neubauer G, Heymans S, Lembo G, Wymann MP, Wetzker R, Houslay MD, Iaccarino G, Scott JD, Hirsch E. Integrating cardiac PIP3 and cAMP signaling through a PKA anchoring function of p110gamma. Mol Cell. 2011; 42(1):84–95. [PubMed: 21474070] [79]. Wu L, Liu L. Systematic Review and Meta-analysis Evaluating the Impact of Vitamin D on the Risk of Heart Failure: New Evidence from Population-based Studies. JCvD. 2014; 2(3):159–173. [80]. Al Maluli H, DeStephan C. Hemodynamic Monitoring in the Intensive Care Unit. JCvD. 2014; 2(2):101–115. [81]. Sardu C, Marfella R, Santulli G, Paolisso G. Functional role of miRNA in cardiac resynchronization therapy. Pharmacogenomics. 2014; 15(8):1159–68. [PubMed: 25084208] [82]. Nguyen MN, Kiriazis H, Ruggiero D, Gao XM, Su Y, Jian A, Han LP, McMullen JR, Du XJ. Spontaneous ventricular tachyarrhythmias in beta2-adrenoceptor transgenic mice in relation to

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 14

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

cardiac interstitial fibrosis. American journal of physiology. Heart and circulatory physiology. 2015; 309(5):H946–57. [PubMed: 26116714] [83]. Ritchie RH, Leo CH, Qin C, Stephenson EJ, Bowden MA, Buxton KD, Lessard SJ, Rivas DA, Koch LG, Britton SL, Hawley JA, Woodman OL. Low intrinsic exercise capacity in rats predisposes to age-dependent cardiac remodeling independent of macrovascular function. American journal of physiology. Heart and circulatory physiology. 2013; 304(5):H729–39. [PubMed: 23262135] [84]. Ho D, Yan L, Iwatsubo K, Vatner DE, Vatner SF. Modulation of beta-adrenergic receptor signaling in heart failure and longevity: targeting adenylyl cyclase type 5. Heart failure reviews. 2010; 15(5):495–512. [PubMed: 20658186] [85]. Dorn GW 2nd, Tepe NM, Lorenz JN, Koch WJ, Liggett SB. Low- and high-level transgenic expression of beta2-adrenergic receptors differentially affect cardiac hypertrophy and function in Galphaq-overexpressing mice. Proceedings of the National Academy of Sciences of the United States of America. 1999; 96(11):6400–5. [PubMed: 10339599] [86]. Tevaearai HT, Eckhart AD, Walton GB, Keys JR, Wilson K, Koch WJ. Myocardial gene transfer and overexpression of beta2-adrenergic receptors potentiates the functional recovery of unloaded failing hearts. Circulation. 2002; 106(1):124–9. [PubMed: 12093781] [87]. 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):7059–64. [PubMed: 10359838] [88]. Du XJ, Gao XM, Wang B, Jennings GL, Woodcock EA, Dart AM. Age-dependent cardiomyopathy and heart failure phenotype in mice overexpressing beta(2)-adrenergic receptors in the heart. Cardiovascular research. 2000; 48(3):448–54. [PubMed: 11090840] [89]. Lymperopoulos, A.; Chowdhary, S.; Sankar, K.; Simon, I. Regulation of Catecholamine Production from the Adrenal Medulla. In: Santulli, G., editor. Adrenal Glands: From Pathophysiology to Clinical Evidence. Nova Science Publisher; New York, NY: 2015. [90]. Thireau J, Karam S, Roberge S, Roussel J, Aimond F, Cassan C, Gac A, Babuty D, Le Guennec JY, Lacampagne A, Fauconnier J, Richard S. Beta-adrenergic blockade combined with subcutaneous B-type natriuretic peptide: a promising approach to reduce ventricular arrhythmia in heart failure? Heart. 2014; 100(11):833–41. [PubMed: 24667281] [91]. Shiny KS, Kumar SH, Farvin KH, Anandan R, Devadasan K. Protective effect of taurine on myocardial antioxidant status in isoprenaline-induced myocardial infarction in rats. The Journal of pharmacy and pharmacology. 2005; 57(10):1313–7. [PubMed: 16259760] [92]. Huang CJ, Webb HE, Zourdos MC, Acevedo EO. Cardiovascular reactivity, stress, and physical activity. Frontiers in physiology. 2013; 4:314. [PubMed: 24223557] [93]. Weng TP, Fu TC, Wang CH, Hsu CC, Wang JS. Activation of lymphocyte autophagy/apoptosis reflects haemodynamic inefficiency and functional aerobic impairment in patients with heart failure. Clinical science. 2014; 127(10):589–602. [PubMed: 24863428] [94]. Berezin A, Kremzer A, Samura T, Martovitskaya Y. Apoptotic Microparticles to Progenitor Mononuclear Cells Ratio in Heart Failure: Relevance of Clinical Status and Outcomes. JCvD. 2014; 2(2):50–57. [95]. Ciccarelli M, Santulli G, Campanile A, Galasso G, Cervero P, Altobelli GG, Cimini V, Pastore L, Piscione F, Trimarco B, Iaccarino G. Endothelial alpha1-adrenoceptors regulate neoangiogenesis. Br J Pharmacol. 2008; 153(5):936–46. [PubMed: 18084315] [96]. Buroker N. ADRBK1 (GRK2) rSNPs, Transcriptional Factor Binding Sites and Cardiovascular Disease in the Black Population. JCvD. 2014; 2(2):62–67. [97]. Santulli G. Epidemiology of Cardiovascular Disease in the 21st Century: Updated Numbers and Updated Facts. JCvD. 2013; 1(1):1–2. [98]. Philipson LH. beta-Agonists and metabolism. The Journal of allergy and clinical immunology. 2002; 110(6 Suppl):S313–7. [PubMed: 12464941] [99]. Ngala RA, O’Dowd J, Wang SJ, Agarwal A, Stocker C, Cawthorne MA, Arch JR. Metabolic responses to BRL37344 and clenbuterol in soleus muscle and C2C12 cells via different atypical pharmacologies and beta2-adrenoceptor mechanisms. Br J Pharmacol. 2008; 155(3):395–406. [PubMed: 18552870]

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 15

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

[100]. Sakamoto K, Holman GD. Emerging role for AS160/TBC1D4 and TBC1D1 in the regulation of GLUT4 traffic. American journal of physiology. 2008; 295(1):E29–37. [PubMed: 18477703] [101]. Maarbjerg SJ, Sylow L, Richter EA. Current understanding of increased insulin sensitivity after exercise - emerging candidates. Acta physiologica (Oxford, England). 202(3):323–35. [102]. Perez-Schindler J, Philp A, Baar K, Hernandez-Cascales J. Regulation of contractility and metabolic signaling by the beta2-adrenergic receptor in rat ventricular muscle. Life sciences. 88(19-20):892–7. [PubMed: 21466811] [103]. Bruzzone A, Sauliere A, Finana F, Senard JM, Luthy I, Gales C. Dosage-dependent regulation of cell proliferation and adhesion through dual beta2-adrenergic receptor/cAMP signals. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2014; 28(3):1342–54. [PubMed: 24308976] [104]. Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nature reviews. 13(4):251–62. [105]. Nikolaidis LA, Poornima I, Parikh P, Magovern M, Shen YT, Shannon RP. The effects of combined versus selective adrenergic blockade on left ventricular and systemic hemodynamics, myocardial substrate preference, and regional perfusion in conscious dogs with dilated cardiomyopathy. Journal of the American College of Cardiology. 2006; 47(9):1871–81. [PubMed: 16682315] [106]. Shahid G, Hussain T. GRK2 negatively regulates glycogen synthesis in mouse liver FL83B cells. The Journal of biological chemistry. 2007; 282(28):20612–20. [PubMed: 17517892] [107]. Lupien JR, Hirshman MF, Horton ES. Effects of norepinephrine infusion on in vivo insulin sensitivity and responsiveness. The American journal of physiology. 1990; 259(2 Pt 1):E210–5. [PubMed: 2116726] [108]. Straznicky NE, Grima MT, Sari CI, Eikelis N, Lambert EA, Nestel PJ, Esler MD, Dixon JB, Chopra R, Tilbrook AJ, Schlaich MP, Lambert GW. Neuroadrenergic dysfunction along the diabetes continuum: a comparative study in obese metabolic syndrome subjects. Diabetes. 2012; 61(10):2506–16. [PubMed: 22664956] [109]. Spallone V, Ziegler D, Freeman R, Bernardi L, Frontoni S, Pop-Busui R, Stevens M, Kempler P, Hilsted J, Tesfaye S, Low P, Valensi P. N. on behalf of the Toronto Consensus Panel on Diabetic, Cardiovascular autonomic neuropathy in diabetes: clinical impact, assessment, diagnosis, and management. Diabetes/metabolism research and reviews. 2011 [110]. Quinones MJ, Nicholas SB, Lyon CJ. Insulin resistance and the endothelium. Current diabetes reports. 2005; 5(4):246–53. [PubMed: 16033673] [111]. Straznicky NE, Lambert EA, Nestel PJ, McGrane MT, Dawood T, Schlaich MP, Masuo K, Eikelis N, de Courten B, Mariani JA, Esler MD, Socratous F, Chopra R, Sari CI, Paul E, Lambert GW. Sympathetic neural adaptation to hypocaloric diet with or without exercise training in obese metabolic syndrome subjects. Diabetes. 2010; 59(1):71–9. [PubMed: 19833893] [112]. Oriente F, Iovino S, Cassese A, Romano C, Miele C, Troncone G, Balletta M, Perfetti A, Santulli G, Iaccarino G, Valentino R, Beguinot F, Formisano P. Overproduction of phosphoprotein enriched in diabetes (PED) induces mesangial expansion and upregulates protein kinase C-beta activity and TGF-beta1 expression. Diabetologia. 2009; 52(12):2642–52. [PubMed: 19789852] [113]. Schernthaner G. Pleiotropic effects of thiazolidinediones on traditional and non-traditional atherosclerotic risk factors. Int J Clin Pract. 2009; 63(6):912–29. [PubMed: 19490202] [114]. Fu Q, Xu B, Liu Y, Parikh D, Li J, Li Y, Zhang Y, Riehle C, Zhu Y, Rawlings T, Shi Q, Clark RB, Chen X, Abel ED, Xiang YK. Insulin inhibits cardiac contractility by inducing a Gi-biased beta2 adrenergic signaling in hearts. Diabetes. 2014 [115]. Wright JJ, Kim J, Buchanan J, Boudina S, Sena S, Bakirtzi K, Ilkun O, Theobald HA, Cooksey RC, Kandror KV, Abel ED. Mechanisms for increased myocardial fatty acid utilization following short-term high-fat feeding. Cardiovascular research. 2009; 82(2):351–60. [PubMed: 19147655] [116]. Azevedo PS, Minicucci MF, Santos PP, Paiva SA, Zornoff LA. Energy metabolism in cardiac remodeling and heart failure. Cardiology in review. 2013; 21(3):135–40. [PubMed: 22990373]

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 16

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

[117]. Glatz JF, Luiken JJ, Bonen A. Membrane fatty acid transporters as regulators of lipid metabolism: implications for metabolic disease. Physiological reviews. 2010; 90(1):367–417. [PubMed: 20086080] [118]. Shimizu I, Minamino T, Toko H, Okada S, Ikeda H, Yasuda N, Tateno K, Moriya J, Yokoyama M, Nojima A, Koh GY, Akazawa H, Shiojima I, Kahn CR, Abel ED, Komuro I. Excessive cardiac insulin signaling exacerbates systolic dysfunction induced by pressure overload in rodents. The Journal of clinical investigation. 2010; 120(5):1506–14. [PubMed: 20407209] [119]. Avendano MS, Lucas E, Jurado-Pueyo M, Martinez-Revelles S, Vila-Bedmar R, Mayor F Jr. Salaices M, Briones AM, Murga C. Increased nitric oxide bioavailability in adult GRK2 hemizygous mice protects against angiotensin II-induced hypertension. Hypertension. 2014; 63(2):369–75. [PubMed: 24191280] [120]. Cipolletta E, Campanile A, Santulli G, Sanzari E, Leosco D, Campiglia P, Trimarco B, Iaccarino G. The G protein coupled receptor kinase 2 plays an essential role in beta-adrenergic receptorinduced insulin resistance. Cardiovascular research. 2009; 84(3):407–15. [PubMed: 19620130] [121]. Watari K, Nakaya M, Kurose H. Multiple functions of G protein-coupled receptor kinases. Journal of molecular signaling. 2014; 9(1):1. [PubMed: 24597858] [122]. Fusco A, Santulli G, Sorriento D, Cipolletta E, Garbi C, Dorn GW 2nd, Trimarco B, Feliciello A, Iaccarino G. Mitochondrial localization unveils a novel role for GRK2 in organelle biogenesis. Cell Signal. 2012; 24(2):468–75. [PubMed: 21983013] [123]. Chen M, Sato PY, Chuprun JK, Peroutka RJ, Otis NJ, Ibetti J, Pan S, Sheu SS, Gao E, Koch WJ. Prodeath signaling of G protein-coupled receptor kinase 2 in cardiac myocytes after ischemic stress occurs via extracellular signal-regulated kinase-dependent heat shock protein 90-mediated mitochondrial targeting. Circulation research. 2013; 112(8):1121–34. [PubMed: 23467820] [124]. Sorriento D, Ciccarelli M, Santulli G, Illario M, Trimarco B, Iaccarino G. Trafficking GRK2: Cellular and Metabolic consequences of GRK2 subcellular localization. Transl Med UniSa. 2014; 10:3–7. [PubMed: 25147759] [125]. Lin LY, Wu CK, Juang JM, Wang YC, Su MY, Lai LP, Hwang JJ, Chiang FT, Tseng WY, Lin JL. Myocardial Regional Interstitial Fibrosis is Associated With Left Intra-Ventricular Dyssynchrony in Patients With Heart Failure: A Cardiovascular Magnetic Resonance Study. Scientific reports. 2016; 6:20711. [PubMed: 26846306] [126]. Trial J, Entman ML, Cieslik KA. Mesenchymal stem cell-derived inflammatory fibroblasts mediate interstitial fibrosis in the aging heart. Journal of molecular and cellular cardiology. 2016; 91:28–34. [PubMed: 26718722] [127]. Schelbert EB, Piehler KM, Zareba KM, Moon JC, Ugander M, Messroghli DR, Valeti US, Chang CC, Shroff SG, Diez J, Miller CA, Schmitt M, Kellman P, Butler J, Gheorghiade M, Wong TC. Myocardial Fibrosis Quantified by Extracellular Volume Is Associated With Subsequent Hospitalization for Heart Failure, Death, or Both Across the Spectrum of Ejection Fraction and Heart Failure Stage. Journal of the American Heart Association. 2015; 4(12) [128]. Dai DF, Rabinovitch PS. Cardiac aging in mice and humans: the role of mitochondrial oxidative stress. Trends in cardiovascular medicine. 2009; 19(7):213–20. [PubMed: 20382344] [129]. Horn MA, Trafford AW. Aging and the cardiac collagen matrix: Novel mediators of fibrotic remodelling. Journal of molecular and cellular cardiology. 2015 [130]. Iwanaga Y, Aoyama T, Kihara Y, Onozawa Y, Yoneda T, Sasayama S. Excessive activation of matrix metalloproteinases coincides with left ventricular remodeling during transition from hypertrophy to heart failure in hypertensive rats. Journal of the American College of Cardiology. 2002; 39(8):1384–91. [PubMed: 11955860] [131]. Seeland U, Selejan S, Engelhardt S, Muller P, Lohse MJ, Bohm M. Interstitial remodeling in beta1-adrenergic receptor transgenic mice. Basic research in cardiology. 2007; 102(2):183–93. [PubMed: 17122889] [132]. Kato S, Spinale FG, Tanaka R, Johnson W, Cooper G.t. Zile MR. Inhibition of collagen crosslinking: effects on fibrillar collagen and ventricular diastolic function. The American journal of physiology. 1995; 269(3 Pt 2):H863–8. [PubMed: 7573529]

Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 17

Author Manuscript

[133]. Communal C, Singh M, Menon B, Xie Z, Colucci WS, Singh K. beta1 integrins expression in adult rat ventricular myocytes and its role in the regulation of beta-adrenergic receptor-stimulated apoptosis. Journal of cellular biochemistry. 2003; 89(2):381–8. [PubMed: 12704801] [134]. Banfi C, Cavalca V, Veglia F, Brioschi M, Barcella S, Mussoni L, Boccotti L, Tremoli E, Biglioli P, Agostoni P. Neurohormonal activation is associated with increased levels of plasma matrix metalloproteinase-2 in human heart failure. European heart journal. 2005; 26(5):481–8. [PubMed: 15618033]

Author Manuscript Author Manuscript Author Manuscript Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 18

Author Manuscript

Highlights ○

Adrenergic receptors are functionally involved in numerous processes underlying both aging and cardiovascular disease, in particular heart failure;



Aging causes a series of rearrangements in the sympathetic nervous system;



This article reviews the pathophysiological role of the sympathetic nervous system in cardiovascular aging.

Author Manuscript Author Manuscript Author Manuscript Maturitas. Author manuscript; available in PMC 2017 November 01.

Santulli and Iaccarino

Page 19

Author Manuscript Author Manuscript Author Manuscript

Figure 1.

Common features in cardiovascular aging and heart failure.

Author Manuscript Maturitas. Author manuscript; available in PMC 2017 November 01.

Adrenergic signaling in heart failure and cardiovascular aging.

Both cardiovascular disease and aging are associated with changes in the sympathetic nervous system. Indeed, mounting evidence indicates that adrenerg...
547KB Sizes 5 Downloads 12 Views