Abstract
Apelin is a vasoactive peptide and is an endogenous ligand for APJ receptors, which are widely expressed in blood vessels, heart, and cardiovascular regulatory regions of the brain. A growing body of evidence now demonstrates a regulatory role for the apelin/APJ receptor system in cardiovascular physiology and pathophysiology, thus making it a potential target for cardiovascular drug discovery and development. Indeed, ongoing studies are investigating the potential benefits of apelin and apelin-mimetics for disorders such as heart failure and pulmonary arterial hypertension. Apelin causes relaxation of isolated arteries, and systemic administration of apelin typically results in a reduction in systolic and diastolic blood pressure and an increase in blood flow. Nonetheless, vasopressor responses and contraction of vascular smooth muscle in response to apelin have also been observed under certain conditions. The goal of the current review is to summarize major findings regarding the apelin/APJ receptor system in blood vessels, with an emphasis on regulation of vascular tone, and to identify areas of investigation that may provide guidance for the development of novel therapeutic agents that target this system.
Keywords: Apelin, APJ receptors, Elabela, vasodilation, vasoconstriction
1. Introduction
Apelin and its receptor, APJ, are widely expressed throughout the cardiovascular system (Kleinz & Davenport, 2004; Kleinz, et al., 2005). This expression pattern has prompted considerable interest in the roles of apelin and APJ receptors in cardiovascular health and disease (Kalea & Batlle, 2010). Indeed, several clinical trials are evaluating the potential benefits of apelin and novel APJ receptor agonists in treating various cardiovascular disorders. A growing body of knowledge continues to shed new light on the apelinergic system, including the discovery of novel endogenous APJ receptor ligands (e.g. Elabela/Toddler) (Chng, et al., 2013; Pauli, et al., 2014), synthetic analogs (e.g. E339-3D6, ML-233, MM07) (Brame, et al., 2015; Iturrioz, et al., 2010; Khan, et al., 2010) and receptor antagonists (e.g. F13A, ML221) (Lee, et al., 2005; Maloney, et al., 2012). With regard to the apelinergic system in blood vessels, there are several features involved in apelin-APJ signaling that make it a potential, but challenging target for drug discovery: (1) apelin has multiple effects on vasomotor tone (Salcedo, et al., 2007; Maguire, et al., 2009; Mughal, et al., 2018), (2) APJ receptors are expressed in the intimal and medial layers of the blood vessel wall, and possibly on cells in the adventitia as well (Kleinz, et al., 2005; Pope, et al., 2012; Mughal, et al., 2018), (3) APJ receptors are associated with multiple G-protein subunits (Hashimoto, et al., 2006; Kang, et al., 2013; Szokodi, et al., 2002), and (4) apelin may act via central nervous system mechanisms to regulate peripheral vascular function (Kagiyama, et al., 2005; Zhang, et al., 2009). The present review summarizes the current state of knowledge regarding the vascular effects of apelin, with an emphasis on the regulation of vasomotor tone, as well as novel pharmacologic agents that interact with APJ receptors in blood vessels.
2. Apelin
Discovered in 1998, apelin was initially identified as the sole endogenous ligand for the APJ receptor (Tatemoto, et al., 1998). Apelin-77 (pre-pro-apelin) is the precursor for various pharmacologically active apelin isoforms (e.g. apelin-12, -13, -17 and -36), and it shares 75–95% sequence homology among various species including rat, mice and human (Lee, et al., 2000). N-terminal residues of apelin-77 are post-translationally modified by endopeptidase to form pro-apelin-55, which is further cleaved to apelin-36, -17, -13 and -12. Each of these fragments has a conserved C-terminal region, which is essential for APJ receptor binding and functional activity (Pitkin, et al., 2010a). Post-translational modification of the N-terminal residue of apelin-13 with cyclized glutamine forms [Pyr]-apelin-13, which evades enzymatic degradation and results in a longer biological duration of action (Zhen, et al., 2013). The different apelin isoforms (i.e. apelin-12, -13, -17 and -36) have variable potency, but apelin-13 and [Pyr]-apelin-13 are identified as the more predominant and potent isoforms in the cardiovascular system (Tatemoto, et al., 1998; Maguire, et al., 2009). More recently, another endogenous APJ receptor-activating peptide, known as Elabela/Toddler (referred to as Elabela) and encoded by the APELA gene, has been discovered (Chng, et al., 2013; Pauli, et al., 2014).
Apelin has potent vascular effects but it is highly prone to proteolytic degradation, thereby limiting its duration of action (Japp, et al., 2008). Studies have shown that angiotensin converting enzyme type-2 (ACE-2), a zinc-containing carboxy monopeptidase, is partially responsible for apelin metabolism (Vickers, et al., 2002; Wang, et al., 2016; Yang, et al., 2017a). [Pyr]-apelin-13 is more susceptible to ACE-2 degradation than is apelin-17, and results in metabolites, i.e. [Pyr]-apelin-12 and apelin-16, respectively, that have reduced functional activity (Wang, et al., 2016). Another recent report showed that ACE-2 metabolizes [Pyr1]-apelin-13 to biologically active [Pyr1]-apelin-13 (1–12), suggesting a beneficial effect of increased ACE-2 expression during cardiovascular diseases (Yang, et al., 2017a). The metalloprotease, neprilysin, has also been shown to be involved in the degradation of apelin. Neprilysin, by truncating the RPRL (Arg2- Leu5) region of apelin, forms a peptide that is devoid of the ability to activate APJ receptors (McKinnie, et al., 2016).
The apelin gene is located on band q25–26.1 of chromosome X and is expressed in vascular endothelial cells of both conduit and resistant arteries (Kawamata, et al., 2001; Medhurst, et al., 2003; Pitkin, et al., 2010b; Pope, et al., 2012). Kleinz et al. reported that the apelin gene is localized to endothelial cells, but not to vascular smooth muscle cells or adipocytes in human saphenous vein, and coronary, pulmonary, and mammary arteries; however, intrarenal arterial endothelial cells lack apelin-like immunoreactivity (Kleinz & Davenport, 2004). It is worth noting that although the apelin peptide is usually absent in vascular smooth muscle cells, it is expressed in smooth muscle cell positive atherosclerotic plaques (Pitkin, et al., 2010b). Moreover, atherosclerotic plaque apelin is co-localized with APJ receptors, suggesting the possibility of apelin-APJ signaling in coronary atherosclerosis.
Regulation of apelin gene expression and secretion of apelin from intracellular sources in the blood vessel wall (e.g. adipocytes, endothelial cells) is complex. Numerous factors and conditions increase apelin gene expression, including cytokines (tumor necrosis factor-alpha) (Daviaud, et al., 2006), lipopolysaccharides (Han, et al., 2008), hypoxia (Eyries, et al., 2008; Glassford, et al., 2007), dehydration (Reaux-Le Goazigo, et al., 2004), and insulin (Boucher, et al., 2005). In humans, hyperinsulinemia up-regulates apelin gene expression in adipose tissue and increases apelin release via phosphoinositide 3-kinase (PI3K) and protein kinase C (PKC)-dependent pathways (Castan-Laurell, et al., 2008). Hypoxia increases apelin gene expression in cultured human pulmonary arterial endothelial and smooth muscle cells via hypoxia inducible factor–1α (HIF-1α) dependent mechanism (Eyries, et al., 2008). Activation of α-retinoic acid receptors by all-trans retinoic acid increases apelin gene expression in HUVEC cells and in carotid arteries from balloon-injured rats (Shi, et al., 2017). Although these studies provide insight into several key pathways involved in apelin regulation, the association between apelin gene expression and release of apelin into the circulation and/or surrounding tissues remains poorly understood. Moreover, although administration of exogenous apelin has beneficial effects on cardiovascular diseases (Japp, et al., 2008; Tatemoto, et al., 2001), our knowledge of the role of endogenous apelin, as well as the stimuli for apelin release, in cardiovascular health and disease is limited.
Patients with cardiovascular disorders typically have lower plasma apelin levels than matched healthy controls. For example, plasma apelin levels are reduced in patients with coronary artery disease (CAD), and among CAD patients with unstable angina or acute myocardial infraction there is a further reduction in apelin levels as compared with asymptomatic CAD patients (Kadoglou, et al., 2010). Similarly, plasma apelin levels are reduced in patients with stable angina, essential hypertension and acute coronary syndrome (Li, et al., 2008b; Gupta, et al., 2016). In patients with heart failure, apelin levels vary depending on the stage and severity of the disease, as plasma apelin levels are elevated in early stage heart failure, but significantly reduced in patients with severe disease as compared to healthy controls (Chen, et al., 2003). Moreover, the concentration of apelin in myocardial tissue is reduced in patients with heart failure (Chandrasekaran, et al., 2010). In contrast to cardiovascular diseases, apelin levels are elevated during metabolic disorders. During obesity, plasma apelin levels are significantly higher than those observed in subjects with normal weight (Boucher, et al., 2005), and are further increased in patients with morbid obesity (Heinonen, et al., 2005). Similarly, plasma apelin levels are increased in patients with diabetes (Castan-Laurell, et al., 2011; Habchi, et al., 2014). It is not yet known whether these changes in apelin levels during cardiovascular and metabolic disorders are associated with protective or compensatory roles of the apelinergic system. For example, in some human and animal studies administration of apelin is shown to restore decreased cardiac function, consistent with a beneficial therapeutic effect of apelin (Koguchi, et al., 2012; Nagano, et al., 2013).
In 2013, a second endogenous ligand for APJ receptors was discovered. Elabela has a 32 amino acid sequence but does not share sequence homology with apelin (Chng, et al., 2013; Pauli, et al., 2014); however, the affinity of the peptide for APJ receptors is similar to that of apelin (Perjes, et al., 2016; O’Carroll, et al., 2017). Nonetheless, evidence suggests that the peptides may not bind in the same way to the orthostatic binding site. Structure-activity relationship studies reveal that the C-terminal moiety (Arg28, Val29, Pro30, Phe31, and Pro32) and His26 residues of Elabela are most important for receptor binding and signaling (Murza, et al., 2016). This contrasts markedly with apelin-13, in which the key pharmacophores (Arg2, Pro3, Arg4, Leu5) are primarily located at the N-terminal. Elabela was initially identified as an embryonic APJ receptor regulator, but recent studies suggest its role in adult tissues as well (Perjes, et al., 2016; Yang, et al., 2017b). Similar to apelin, Elabela is also expressed in vascular endothelial cells (Perjes, et al., 2016; O’Carroll, et al., 2017). An understanding of the functional role of Elabela in the vascular system is still emerging but recent evidence suggests that Elabela can compensate for down-regulated functions of apelin during various cardiovascular diseases. Administration of exogenous Elabela improves right ventricular systolic pressure and hypertrophy, as well as pulmonary vascular remodeling, in a rat model of pulmonary hypertension in which apelin signaling is downregulated (Yang, et al., 2017b). Infusion of Elabela, which was associated with an increase in cardiac output and reduced systemic vascular resistance, improves cardiovascular function and survival in a cecal ligation puncture model of sepsis (Coquerel, et al., 2017). On the other hand, reduced levels of Elabela may lead to cardiovascular abnormalities, inasmuch as pregnant Elabela knockout mice display preeclampsia-like symptoms and defective placental angiogenesis (Ho, et al., 2017).
3. APJ Receptor
The APJ receptor is a G-protein coupled receptor located on chromosome 11 and composed of an intron-less gene (APLNR), which is conserved in many species including human, monkey, chimpanzee, mouse and rat (Pitkin, et al., 2010a). APJ receptors were first discovered in 1993 during a search for vasopressin receptors (O’Dowd, et al., 1993). Immunocytochemical and autoradiographic studies have detected APJ receptors in endothelial and smooth muscle cells in numerous blood vessels, including human coronary, internal mammary, radial, and pulmonary arteries, human saphenous vein, and rat aorta, coronary, cerebral and pulmonary arteries (Katugampola, et al., 2001; Kleinz, et al., 2005; Pitkin, et al., 2010b; Mughal, et al., 2018).
APJ receptors have a 380 amino acid sequence with a characteristic G-protein structure, including seven transmembrane domains and post-translation modification sites for phosphorylation, palmitoylation and glycosylation along with association sites for β-arrestin (O’Dowd, et al., 1993). APJ receptor signaling involves multiple G-protein subunits. Initial work suggested the involvement of pertussis toxin sensitive Gαi/o subunits that inhibit forskolin-induced cyclic adenosine monophosphate (cAMP) formation in CHO cells (Tatemoto, et al., 1998). This signaling pathway is also present in blood vessels, inasmuch as apelin inhibits large conductance, calcium-activated potassium (BKCa) channel currents in cerebral vascular smooth muscle cells in a pertussis toxin-sensitive manner (Modgil, et al., 2013). Apelin was also found to increase Ca2+ mobilization in neuronal cells (Choe, et al., 2000), suggesting the involvement of other G-protein subunits. Subsequent studies demonstrated that apelin-induced cardiac and smooth muscle contractile responses were attenuated by inhibitors of phospholipase C and PKC, suggesting a role for Gαq/11 subunits in APJ receptor signaling (Hashimoto, et al., 2006; Szokodi, et al., 2002). Moreover, apelin was shown to increase phosphorylation of myosin light chain (MLC) in vascular smooth muscle cells via activation of Gαq/11 subunits and a PKC-dependent mechanism (Hashimoto, et al., 2006). In addition, a functional role for Gα13 subunits has been identified in apelin-induced cytoplasmic translocation of histone deacetylase during cardiac and vascular development (Kang, et al., 2013).
Various G-protein subunits, e.g. Gαi/o, Gαq and βγ dimers, can activate PI3K/Akt signaling (Murga, et al., 1998), which is indeed involved in apelin-induced vascular responses. For example, apelin inhibits calcification of vascular smooth muscle cells via a PI3K/Akt signaling mechanism (Shan, et al., 2011). Similarly, apelin attenuates apoptosis in vascular smooth muscle cells by activating PI3K/Akt and extracellular signal-regulated kinase (ERK) signaling (Cui, et al., 2010; Tang, et al., 2007). Apelin also inhibits BKCa channel currents by activation of PI3K/Akt pathways (Modgil, et al., 2013). Apelin-induced PI3K/Akt signaling can increase phosphorylation of endothelial nitric oxide synthase (eNOS) to modulate aortic vascular tone (Zhong, et al., 2007b). Likewise, Elabela improves self-renewal and regenerative ability of human progenitor stem cells via PI3K/Akt activation (Ho, et al., 2015). These latter actions of Elabela might be mediated by a cell surface receptor different from the APJ receptor in human embryonic stem cells (Ho, et al., 2015), suggesting the possible existence of another subtype or novel type of receptor; however, the bulk of evidence to date continues to support the view that the peptide is acting solely on the currently identified APJ receptor. Elabela also inhibits forskolin-induced cAMP formation by activating ERK1/2 signaling pathways in CHO cells (Wang, et al., 2015b).
In addition to these intracellular events, APJ receptors also have the ability to form homo- and heterodimers. APJ receptors are reported to hetero-dimerize with angiotensin (AT1) receptors (Siddiquee, et al., 2013), bradykinin (B1) receptors (Bai, et al., 2014b), neurotensin receptor-1 (NTSR1) (Bai, et al., 2014a) and κ-opioid receptors (KOR) (Li, et al., 2012). These receptors are involved in cardiovascular regulation and their ligands (apelin, des-Arg(9)-bradykinin, neurotensin and dynorphin) are sensitive to ACE-2 proteolysis (Vickers, et al., 2002), suggesting the importance of these heterodimers in cardiovascular pharmacology. For example, APJ receptors can allosterically modify the functional state of AT1-receptors to a low affinity state towards angiotensin II (Siddiquee, et al., 2013; Sun, et al., 2011) and these actions can be independent of its putative ligand, apelin (Siddiquee, et al., 2013). Similarly, constitutive heterodimers of APJ- and bradykinin (B1)-receptors are shown to generate higher intracellular Ca2+ and upregulate eNOS phosphorylation, possibly by strengthening the association of the dimer with Gαq subunits (Bai, et al., 2014b). APJ-NTSR1 or APJ-KOR heterodimers also cause significant increases in phosphorylated ERK1/2 levels by increasing intracellular Ca2+ (Li, et al., 2012; Bai, et al., 2014a). A recent report demonstrated the existence of homodimers-oligomers of the human APJ receptor, which could possibly mediate different signaling events in comparison to APJ monomers (Cai, et al., 2017).
APJ receptor expression is altered in various cardiovascular diseases, and both increases as well as decreases in APJ receptor expression have been reported. A decrease in mRNA and protein levels of APJ receptors was observed in patients with ischemic heart disease or idiopathic dilated cardiomyopathy (Földes, et al., 2003; Pitkin, et al., 2010b); however, APJ receptor protein levels were significantly increased in ischemic heart failure (Atluri, et al., 2007; Sheikh, et al., 2008). Hypoxia, one of the major consequences of ischemic injury, may lead to increased APJ receptor expression, as hypoxia-activated HIF-1α has been shown to cause aberrant increases in APJ receptor protein expression (Kerkela, et al., 2013; Kong, et al., 2015; Zhang, et al., 2015). APJ receptor mRNA levels are up-regulated in patients with aortic valve stenosis in comparison to controls (Peltonen, et al., 2009). Both mRNA and protein levels of APJ receptors are reduced in the heart, kidney and aorta of hypertensive rats (Najafipour, et al., 2012; Najafipour, et al., 2015), whereas in obese women, there is an increase in APJ receptor mRNA levels, which was attenuated by improving insulin resistance (Castan-Laurell, et al., 2008).
4. Vascular Effects of Apelin
Apelin has complex vasomotor effects as it can cause either vasodilation or vasoconstriction depending on the vascular bed and underlying conditions. These dual actions of apelin are attributed to the presence of APJ receptors in both the endothelial and smooth muscle cell layers of the blood vessel wall. Vasoactive agents may act directly on vascular smooth muscle cells to cause contraction or relaxation, or on endothelial cells, which can secrete substances that mediate vasodilation (e.g. NO, prostacyclin) as well as vasoconstriction (e.g. endothelin) (O’Rourke, et al., 2006).
4.1 Role of Apelinergic System in Vasodilation
A hypotensive effect of apelin was first reported in 2000, when it was shown that intraperitoneal injection of apelin reduces both systolic and diastolic pressure in anesthetized normotensive animals (Lee, et al., 2000). Shortly thereafter, intravenous administration of apelin was shown to produce hypotensive effects (Reaux, et al., 2001), as well as cause venodilation (Cheng, et al., 2003). The various apelin isoforms may have different potencies in different experimental models. For example, apelin-12 is more potent in reducing mean arterial pressure in comparison to apelin-13 and -36 in anesthetized normotensive rats (Tatemoto, et al., 2001), whereas apelin-13 has higher potency in comparison to apelin-12 in spontaneously hypertensive rats (Lee, et al., 2005). In addition, apelin-17 produces a greater depressor response than [Pyr]-apelin-13 and apelin-15 in normotensive rats (El Messari, et al., 2004).
A role for NO in the hypotensive effect of apelin was demonstrated in studies where the NO synthase inhibitor, N(G)-nitro-l-arginine methyl ester (L-NAME), abolished the reduction in mean arterial pressure produced by intravenous administration of apelin (Tatemoto, et al., 2001). Similarly, intravenous infusion of apelin-36 and [Pyr]-apelin-13 produced coronary vasodilation that was inhibited by an “NO clamp” (L-N(G)-monomethylarginine co-infused with sodium nitroprusside), but not by inhibition of cyclooxygenase with aspirin (Japp, et al., 2008). Experiments performed with isolated human and rat peripheral arteries also suggested a role for NO in apelin-induced relaxations. Relaxation of isolated human mesenteric, but not hepatic arteries was attenuated in the presence of L-NAME (Salcedo, et al., 2007). In isolated rat aorta, apelin caused concentration- and time-dependent increases in eNOS activity and NO formation (Jia, et al., 2007). Apelin produced endothelium-dependent relaxation of isolated rat coronary arteries by stimulating the release of NO, which activated BKCa channels without altering cGMP levels in the underlying smooth muscle cells of these arteries (Mughal, et al., in press). Activation of mechanosensor pathways by flow-mediated shear stress increases APJ receptor expression and apelin and NO formation in human endothelial cells (Busch, et al., 2015). A role for prostanoids in apelin-induced relaxation was established in human mammary arteries, where several apelin isoforms (apelin-13, -36 and [Pyr]-apelin-13) caused concentration-dependent relaxations that were inhibited in the presence of indomethacin, but not L-NAME (Maguire, et al., 2009). Moreover, in the isolated portal vein of the rat, apelin attenuated angiotensin II-induced contractions, but the effect of apelin was only partially inhibited by endothelial denudation, suggesting that apelin may act directly on vascular smooth muscle to cause relaxation (Gurzu, et al., 2006). By contrast to apelin-induced relaxation, Elabela-induced relaxation in isolated mouse aortae was unchanged in the presence of L-NAME, suggesting the involvement of NO-independent mechanism(s) in the relaxation response to Elabela (Wang, et al., 2015b).
Apelin has been shown to have beneficial effects in several vascular or metabolic disorders. In animal models of pulmonary hypertension and diabetes, apelin treatment lowered mean arterial pressure and reduced the levels of vasoconstrictor mediators such as angiotensin-II and endothelin-1 (Akcilar, et al., 2013; Falcao-Pires, et al., 2009). Similarly, intravenous bolus administration of apelin caused a significant reduction in mean pulmonary arterial pressure during acute pulmonary embolism (Feng, et al., 2010), and apelin signaling via APJ – KOR heterodimers caused a depressor response during renovascular hypertension (Yeganeh-Hajahmadi, et al., 2017; Rostamzadeh, et al., 2018). Moreover, by increasing eNOS phosphorylation, apelin reversed the impaired relaxation response to acetylcholine and abrogated abnormal Ang II-induced contractile tone in intrarenal arteries from diabetic mice (Zhong, et al., 2007a). In human subjects, apelin-induced vasodilation was preserved in patients with chronic heart failure whereas the vasodilator response to acetylcholine was significantly impaired (Japp, et al., 2010). In middle-aged and older adults, an exercise-associated increase in plasma apelin levels was positively correlated with a decrease in carotid arterial stiffness, which commonly occurs with aging (Fujie, et al., 2014).
Like apelin, the novel peptide, Elabela, also has the ability to cause vasodilation in coronary arteries (Perjes, et al., 2016). Elabela has been shown to reverse vasopressor responses during pulmonary arterial hypertension and angiotensin-II-induced hypertension (Yang, et al., 2017b; Sato, et al., 2017). Moreover, sustained Elabela gene therapy delayed blood pressure elevation in hypertensive rats (Schreiber, et al., 2017).
The renin-angiotensin system is a key pathway in the development and progression of hypertension, heart failure and other cardiovascular disorders. APJ and AT1-receptors share significant sequence homology and have a similar tissue distribution pattern (O’Dowd, et al., 1993); however, apelin does not bind AT1-receptors nor does angiotensin II (Ang II) bind to APJ receptors (O’Dowd, et al., 1993; Tatemoto, et al., 1998). Nonetheless, activation of the apelinergic system has an antagonistic effect toward AT1-receptor mediated responses, either by allosteric regulation of the receptor (Sun, et al., 2011; Siddiquee, et al., 2013) or by increasing NO-dependent signaling (Siddiquee, et al., 2011). Both acute and chronic apelin-induced vasodilation and depressor responses are preserved during renin-angiotensin system activation in healthy humans and in patients with heart failure (Barnes, et al., 2013). Moreover, apelin abrogated Ang II-induced atherosclerosis in ApoE-deficient mice by increasing NO formation to quench superoxide-induced changes in the vascular wall (Chun, et al., 2008). The antagonistic effects of apelin on Ang II signaling may be dependent on the underlying pathologic condition, since apelin attenuated Ang II-induced contractions in pulmonary arteries from normoxic animals, but not in arteries from animals exposed to chronic hypoxia (Andersen, et al., 2009). Apelin-APJ receptor signaling can also increase ACE-2 gene expression, which could increase conversion of Ang II to Angiotensin1–7 to modulate cardiovascular functions (Sato, et al., 2013).
4.2 Role of the Apelinergic System in Vasoconstriction
APJ receptors are expressed on vascular smooth muscle cells and their activation results in vasoconstriction (Katugampola, et al., 2001; Maguire, et al., 2009; Han, et al., 2013; Mughal, et al., 2018). Indeed, some studies have reported a blood pressure-elevating effect of systemic administration of apelin, consistent with a vasoconstrictor response to the peptide. For example, a biphasic hemodynamic response to an intravenous bolus of apelin was observed in conscious sheep, where there was an initial transient fall in arterial pressure followed by a rise in arterial pressure and peripheral vascular resistance (Charles, et al., 2006). In conscious rats, intravenous administration of apelin failed to produce a depressor response, but caused a dose-dependent increase in mean arterial pressure (Kagiyama, et al., 2005).
Though few studies have been performed with isolated blood vessels, apelin generally causes relaxation of isolated arteries with intact endothelium (as described above), whereas vasoconstriction has been observed in isolated vessels denuded of endothelium and under conditions where the endothelial cells are damaged or dysfunctional. [Pyr]-Apelin-13 causes vasoconstriction in endothelium-denuded human saphenous veins and mammary arteries (Katugampola, et al., 2001; Maguire, et al., 2009), and in coronary arteries obtained from atherosclerotic patients (Pitkin, et al., 2010b). Apelin may also permeate through endothelial cells damaged by asymmetric dimethylarginine (Wang, et al., 2011), and cause concentration-dependent vasoconstriction in isolated caudal arteries by increasing phosphorylation of MLC in vascular smooth muscle cells (Han, et al., 2013). That apelin passes through dysfunctional endothelial cells and activates APJ receptors on vascular smooth muscle is supported by in vivo studies in which apelin caused an increase in systolic blood pressure in mice treated with L-NAME, and in rats with vascular endothelium damaged by asymmetric dimethylarginine (Han, et al., 2013; Nagano, et al., 2013). In mouse thoracic aorta, apelin causes vasoconstriction by increasing myosin light chain phosphorylation via Gαi/o-dependent activation of PKC and Na+-Ca2+ exchanger-dependent pathways (Hashimoto, et al., 2006). In addition to MLC phosphorylation as a mechanism of vasoconstriction, apelin may also exert a net vasoconstrictor influence by inhibiting NO-induced relaxation, an effect of apelin that is mediated by inhibition of NO-induced activation of BKCa channels in cerebral arterial smooth muscle cells (Modgil, et al., 2013; Mughal, et al., 2018).
Vasopressor responses to apelin have also been observed following injection of the peptide into cardiovascular regulatory regions of the brain (Seyedabadi, et al., 2002; Kagiyama, et al., 2005). Moreover, overexpression of apelin in the rostral ventrolateral medulla of normotensive rats results in increased mean arterial blood pressure via NADPH oxidase-dependent formation of superoxide(s), and apelin gene expression in the rostral ventrolateral medulla is increased in spontaneously hypertensive rats (Yao, et al., 2011; Zhang, et al., 2009). The vasopressor response to centrally administered apelin is abolished in animals fed a high fat diet or subjected to chronic stress (Cudnoch-Jedrzejewska, et al., 2015; Gomolka, et al., 2015).
The role of apelin-APJ receptor signaling during cardiovascular disorders associated with endothelial dysfunction remains unclear. On one hand, increased apelin/APJ receptor expression and bioavailability produced beneficial vascular effects in conditions with endothelial dysfunction such as end-stage heart failure (Fukushima, et al., 2010), atherosclerosis (Chun, et al., 2008) and obesity (Schinzari, et al., 2017), while others have shown that apelinergic signaling may be a mediator for atherosclerosis (Hashimoto, et al., 2007). Indeed, up-regulated apelin genes are localized in atherosclerosis plaques (Pitkin, et al., 2010b) and stenotic aortic valves (Peltonen, et al., 2009). Thus, it will be important to decipher whether increased apelin-APJ receptor signaling has a role in disease progression or is involved in compensatory feedback mechanism(s).
4.3 Other Vascular Effects of Apelin
In addition to its direct regulatory role in controlling vascular tone, the apelin/APJ receptor signaling system also plays an important role in vascular stabilization, including angiogenesis, proliferation and permeability.
Cell Migration and Angiogenesis
Cell migration is an important feature of several physiological and pathological processes, including embryogenesis, would healing, inflammation, cardiovascular disease and cancer (Staff, 2001). High expression of APJ receptors during embryogenesis and postnatal retinal blood vessel formation provided initial support for the importance of apelinergic signaling during angiogenesis (Devic, et al., 1996; Saint-Geniez, et al., 2002). In addition, Elabela increases migration of mesendodermal cells during gastrulation in a tightly regulated fashion (Pauli, et al., 2014).
Angiogenesis can be either beneficially adaptive (e.g. ischemia, myocardial infarction) or potentially deleterious (e.g. tumor growth, retinopathy, nephropathy). Hence, the role of the apelinergic system during angiogenesis can have either beneficial or detrimental effects based on the underlying physiology or pathological conditions. For example, intra-myocardial delivery of apelin-overexpressing bone marrow cells to ischemic mice increased the homing of bone marrow derived progenitor cells in ischemic heart progressing to neovascularization (Li, et al., 2013b). Similarly, increased coronary collateral development is correlated with increased plasma apelin levels in patients with coronary artery disease, establishing the beneficial angiogenic potential of apelin (Akboga, et al., 2014). Moreover, loss of apelin-APJ signaling has been shown to impair differentiation of endothelial, hematopoietic and cardiac progenitor cells (Inui, et al., 2006; Zeng, et al., 2007; Wang, et al., 2013). Treatment with apelin also alleviates diabetic cardiomyopathy in mice by increasing angiogenesis via SirT3-dependent pathways (Zeng, et al., 2014). By contrast, apelin-induced angiogenesis of retinal and glomerular endothelial cells is associated with increased risk of retinopathy and nephropathy during type 2 diabetes mellitus (Zhang, et al., 2013; Du, et al., 2014). Furthermore, apelin-induced migration of vascular smooth muscle cells suggests the possible involvement of smooth muscle APJ receptor signaling in the development of atherosclerosis (Liu, et al., 2013; Wang, et al., 2015a).
Cell Proliferation
Cellular proliferation is a complex process and apelin is shown to have both proliferative and anti-proliferative actions. Apelin stimulates vascular smooth muscle cell proliferation via PI3K/Akt/ERK (Liu, et al., 2010) and Jagged-1/Notch3 (Li, et al., 2013a) signaling pathways with a common downstream mediator, cyclin D1, to accelerate cell cycle progression from G0/G1 to S phase (Li, et al., 2008a). In addition, hypoxia-induced apelin gene expression also increases proliferation and differentiation of stem cells and progenitor cells, suggesting the possible therapeutic potential of apelin in ischemic reperfusion injury (Zhang, et al., 2015; Hou, et al., 2017). By contrast, apelin treatment reduces pulmonary vascular smooth muscle cell proliferation in hypoxic conditions via PI3K/Akt/mTOR signaling pathways (Zhang, et al., 2014b). Likewise, down-regulation of apelin signaling during pulmonary hypertension is correlated with hyper-proliferation of pulmonary endothelial cells and smooth muscle cells (Kim, et al., 2013).
Cell Permeability
Cell adhesion molecules regulate cell-to-cell adhesion and paracellular permeability. By regulating the permeability of pericytes, apelin modulates vascular development and remodeling (Chen, et al., 2015). The putative beneficial effects of apelin during obesity and cancer are associated with improved vascular integrity and decreased permeability (Kidoya, et al., 2012; Sawane, et al., 2013). Similarly, overexpression of apelin-APJ signaling during acute lung injury and respiratory distress syndrome can reduce capillary-alveolar leakage to alleviate symptoms (Fan, et al., 2015). These beneficial actions of apelin suggest a protective role of apelin during inflammation and immunotoxicity. In this regard, apelin is shown to have protective actions during post-burn sepsis via PI3K signaling pathways (Luo, et al., 2015).
5. Apelin Mimetics and Inhibitors
At present, only a limited number of apelin-like agonists and APJ receptor antagonists have been discovered. There is still a great need for the development of potent and selective drugs that target the apelin/APJ receptor-signaling pathway, not only as potential therapeutic agents but also as pharmacologic tools for dissecting the role of apelin in vascular biology. Those agents most commonly being used at the present time are described below.
5.1 Apelin Mimetics
There are three general types of apelin-like agonists, i.e. non-peptide agonists, biased agonists, and peptide agonists. In addition, novel drug delivery approaches, such as PEGylation and nano-encapsulation, have been employed in order to increase the biological half-life and activity of native apelin isoforms (Narayanan, et al., 2015).
E339-3D6 was the first non-peptide APJ receptor agonist to be developed (Iturrioz, et al., 2010). E339-3D6 has a binding affinity of 90 nM towards APJ receptors and it relaxes precontracted rat thoracic aorta at lower concentrations than acetylcholine (pD2= 8.6 ± 0.4 vs. 6.8 ± 0.4 for E339-3D6 and acetylcholine; respectively). Moreover, E339-3D6 showed full agonist behavior with regard to β-arrestin-dependent APJ receptor internalization, but behaved as a partial agonist in inhibiting cAMP formation (Iturrioz, et al., 2010). E339-3D6 was later shown to be a mixture of polymethylated species, which were further purified to generate analogs with higher affinity and full agonist activity (Margathe, et al., 2014). Another synthetic non-peptide analog, ML-233, also reduced forskolin-induced cAMP formation, with maximum effects at 100 μM, and increased APJ receptor internalization by β-arrestin recruitment (Khan, et al., 2010). ML-233 has ~ 21-fold selectivity towards APJ receptors as compared to AT1-receptors. Further in vitro and in vivo characterization of these molecules is required in order to better understand their vasoactive therapeutic potential.
Target receptor internalization and β-arrestin dependent receptor silencing are major limitations for G-protein coupled receptors, including APJ receptor mediated signaling (Evans, et al., 2001; Masri, et al., 2006). Novel strategies are being employed to develop biased agonists with higher affinity towards the APJ receptor and with minimal effects on β-arrestin dependent pathways. Brame et al. designed the first cyclic biased APJ receptor agonist, MM07 (Brame, et al., 2015). MM07 has nanomolar potency towards the human APJ receptor (pD2: 9.54 ± 0.42) and higher circulatory life (~17 min), but significantly lower potency towards β-arrestin internalization (pD2: 6.16 ± 0.07) in comparison with [Pyr]-apelin-13. Repeated administration of MM07 caused an increase in forearm blood flow without desensitization of APJ receptors. MM07 also caused a significant increase in cardiac output with no evidence of hemodynamic instability (Brame, et al., 2015). This same group later designed another small molecule APJ receptor biased agonist, CMF-019. This molecule has longer half-life (~38 minutes) with nanomolar affinity (pD2=10.00 ± 0.13) towards APJ receptors, with lesser potency on receptor internalization (pD2=6.16 ± 0.21). Moreover, CMF-019 increased cardiac contractility with limited effects on the vasculature (Read, et al., 2016).
Two other synthetic analogs of apelin-17, P92 and LIT01-196, were designed by classic chemical substitution and fluorocarbon chain addition, respectively (Gerbier, et al., 2017). These analogs resulted in higher circulatory half-life (P92: 24 min and LIT01-196: >24 hr) with sub-nanomolar binding affinity (Ki= 0.09 ± 0.02 nM and 0.08 ± 0.01 nM for P92 and LIT01-196, respectively). Both P92 and LIT01-196 caused potent relaxation of rat thoracic aorta (EC50= 7 ± 5 nM and 2 ± 1 nM for P92 and LIT01-196, respectively), which was abolished in the presence of L-NAME. Moreover, both P92 and LIT01-196 caused a marked increase in the diameter of rat glomerular arterioles precontracted with angiotensin-II. Intravenous injection of LIT01-96 and P92 in normotensive anesthetized rats caused a potent and prolonged depressor response in comparison to apelin-17. Likewise, both compounds showed a potent inotropic and diuretic response suggesting their ability to activate multiple signaling mechanisms to maintain physiological homeostasis (Gerbier, et al., 2017).
Considerable efforts have been made to structurally modify apelin in order to improve its biological half-life. One approach has been to design molecules that are resistant to enzymatic degradation by neprilysin. Recently, three metabolically stable analogs of apelin-17 have been synthesized by modifying neprilysin-sensitive Leu-Arg (RPRL) residues. These analogs include modified N-methyl Leu9 analogs and aza-analogs (azaArg and azaLeu), which demonstrated sub-nanomolar binding affinity (Ki= 0.14 – 0.15 nM) with beneficial cardiovascular effects, including vasodepressor and inotropic effects (McKinnie, et al., 2017). Another approach has been to structurally modify apelin by PEGylation of the N-terminal residue. Conjugation of a 40 kDa PEG moiety to apelin-36 resulted in a longer circulatory half-life with minimal effect on the binding constant (Ki= 0.3 nM vs. 0.05 nM with and without PEG-conjugation)(Jia, et al., 2012). PEG-apelin-36 demonstrated an extended effect on cardiac ejection fraction, but had no effect on aortic pressure, similar to that observed with apelin-36 (Jia, et al., 2012). It is not yet known if developing similar conjugates with the more vasoactive isoforms of apelin, i.e. apelin-13 and/or [Pyr]-apelin-13, will further prolong and enhance their vasoactive effects.
5.2 Apelin Inhibitors
The first antagonist of the cardiovascular actions of apelin is known as F13A, which was synthesized by chemical substitution of the C-terminal phenylalanine (Phe13) residue of the native apelin peptide with an alanine residue (Lee, et al., 2005). F13A demonstrated dose-dependent inhibition of apelin-induced hypotensive effects, with no effect on Ang II-induced hypertensive effects, indicating selectivity of F13A toward apelin-induced responses (Lee, et al., 2005). F13A also attenuated apelin-induced depressor responses during renovascular hypertension (Rostamzadeh, et al., 2018) and, in isolated rat cerebral arteries, F13A abolished the inhibitory effects of apelin on BKCa channel function and NO-induced relaxation (Mughal, et al., 2018). Moreover, pressor responses to centrally administered apelin in both normotensive and hypertensive rats are significantly attenuated by pre-treatment with F13A (Zhang, et al., 2014a; Griffiths, et al., 2017).
A second peptide APJ receptor antagonist was designed using a bivalent ligand approach (Macaluso, et al., 2011). The cyclic APJ receptor antagonist, [cyclo(1–6)CRPRLC-KH-cyclo(9–14)CRPRLC], demonstrated a Ki value of 82 nM in CHO cells transfected with human APJ receptors and a KD value of 3.2 μM in competition binding studies in human left ventricle (Macaluso, et al., 2011). This cyclic receptor antagonist has yet to be evaluated in in vivo or in vitro studies designed to determine its antagonist activity versus apelin-induced vasoactive effects.
ML-221 is the first non-peptide APJ receptor antagonist to be developed (Maloney, et al., 2012). ML-221 has micromolar potency in cell-based assays (IC50: 0.70 μM in the cAMP assay, and 1.75 μM in the β-arrestin assay) and > 37-fold selectivity towards APJ receptors as compared to AT1-receptors. However, ML-221 has low aqueous solubility, moderate stability, limited membrane permeability, and high protein binding, all of which can limit its utility as an in vitro or in vivo pharmacologic tool (Maloney, et al., 2012). Nonetheless, it was recently reported that ML-221 caused a reduction in apelin-induced microvascular endothelial cell proliferation and that intraperitoneal administration of ML-221 reduced the retinal angiogenesis commonly observed during ischemic retinopathy (Ishimaru, et al., 2017). ML-221 has also been shown to attenuate Elabela-induced APJ receptor activation and signaling (Yang, et al., 2017b). The effects of ML-221 on the vasomotor actions of apelin are not yet known.
6. Conclusions and Future Directions
Targeting the apelinergic system provides exciting opportunities for the treatment of cardiovascular disease. Apelin/APJ receptor signaling has beneficial effects in several cardiovascular and metabolic disorders, including hypertension, heart failure, pulmonary hypertension, and obesity. Promising apelin-mimetics are in development, but several challenges remain. A key limitation at present is that our knowledge of the vasomotor effects of apelin in specific vascular beds (e.g. coronary, cerebral, renal) is limited. As there is potential for apelin to cause both vasodilation and vasoconstriction, there is a need to improve our understanding of apelin signaling and how it relates to these opposing functional effects. Other challenges associated with the therapeutic use of apelin and its isoforms include their short biological half-lives, complex intracellular signaling pathways, and the possible existence of more than one APJ receptor subtype or another cell surface receptor that has affinity for both apelin and Elabela. Overcoming these challenges will provide guidance on how to optimize opportunities to target the beneficial effects of apelin and improve the development of novel therapeutic agents for treating cardiovascular diseases.
Acknowledgments
Funding Source
Research in the authors’ laboratory is supported by a grant to STO from the National Institutes of Health National Heart, Lung, and Blood Institute (Grant HL124338).
Abbreviations
- APJ
apelin receptor
- ACE-2
angiotensin converting enzyme type-2
- AT-1
angiotensin-1
- BKCa
large conductance calcium-activated potassium
- CAD
coronary artery disease
- cAMP
cyclic adenosine monophosphate
- cGMP
cyclic guanosine monophosphate
- ERK
extracellular signal-regulated kinase
- HIF-1α
hypoxia inducible factor - 1α
- KOR
κ-opioid receptors
- L-NAME
N(G)-nitro-l-arginine methyl ester
- MLC
myosin light chain
- NO
nitric oxide
- PI3K
phosphoinositide 3-kinase
- PKC
protein kinase C
Footnotes
Conflict of Interest Statement
The authors declare that there are no conflicts of interest.
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.
References
- Akboga MK, Akyel A, Sahinarslan A, Demirtas CY, Yayla C, Boyaci B, Yalcin R. Relationship between plasma apelin level and coronary collateral circulation. Atherosclerosis. 2014;235:289–294. doi: 10.1016/j.atherosclerosis.2014.04.029. [DOI] [PubMed] [Google Scholar]
- Akcilar R, Turgut S, Caner V, Akcilar A, Ayada C, Elmas L, Ozcan TO. Apelin effects on blood pressure and RAS in DOCA-salt-induced hypertensive rats. Clinical and Experimental Hypertension. 2013;35:550–557. doi: 10.3109/10641963.2013.764889. [DOI] [PubMed] [Google Scholar]
- Andersen CU, Markvardsen LH, Hilberg O, Simonsen U. Pulmonary apelin levels and effects in rats with hypoxic pulmonary hypertension. Respiratory Medicine. 2009;103:1663–1671. doi: 10.1016/j.rmed.2009.05.011. [DOI] [PubMed] [Google Scholar]
- Atluri P, Morine KJ, Liao GP, Panlilio CM, Berry MF, Hsu VM, Hiesinger W, Cohen JE, Joseph Woo Y. Ischemic heart failure enhances endogenous myocardial apelin and APJ receptor expression. Cellular & Molecular Biology Letters. 2007;12:127–138. doi: 10.2478/s11658-006-0058-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai B, Cai X, Jiang Y, Karteris E, Chen J. Heterodimerization of apelin receptor and neurotensin receptor 1 induces phosphorylation of ERK(1/2) and cell proliferation via Gαq-mediated mechanism. Journal of Cellular and Molecular Medicine. 2014a;18:2071–2081. doi: 10.1111/jcmm.12404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai B, Liu L, Zhang N, Wang C, Jiang Y, Chen J. Heterodimerization of human apelin and bradykinin 1 receptors: novel signal transduction characteristics. Cellular Signalling. 2014b;26:1549–1559. doi: 10.1016/j.cellsig.2014.03.022. [DOI] [PubMed] [Google Scholar]
- Barnes GD, Alam S, Carter G, Pedersen CM, Lee KM, Hubbard TJ, Veitch S, Jeong H, White A, Cruden NL, Huson L, Japp AG, Newby DE. Sustained cardiovascular actions of APJ agonism during renin-angiotensin system activation and in patients with heart failure. Circulation Heart Failure. 2013;6:482–491. doi: 10.1161/CIRCHEARTFAILURE.111.000077. [DOI] [PubMed] [Google Scholar]
- Boucher J, Masri B, Daviaud D, Gesta S, Guigne C, Mazzucotelli A, Castan-Laurell I, Tack I, Knibiehler B, Carpene C, Audigier Y, Saulnier-Blache JS, Valet P. Apelin, a newly identified adipokine up-regulated by insulin and obesity. Endocrinology. 2005;146:1764–1771. doi: 10.1210/en.2004-1427. [DOI] [PubMed] [Google Scholar]
- Brame AL, Maguire JJ, Yang P, Dyson A, Torella R, Cheriyan J, Singer M, Glen RC, Wilkinson IB, Davenport AP. Design, characterization, and first-in-human study of the vascular actions of a novel biased apelin receptor agonist. Hypertension. 2015;65:834–840. doi: 10.1161/HYPERTENSIONAHA.114.05099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Busch R, Strohbach A, Pennewitz M, Lorenz F, Bahls M, Busch MC, Felix SB. Regulation of the endothelial apelin/APJ system by hemodynamic fluid flow. Cellular Signalling. 2015;27:1286–1296. doi: 10.1016/j.cellsig.2015.03.011. [DOI] [PubMed] [Google Scholar]
- Cai X, Bai B, Zhang R, Wang C, Chen J. Apelin receptor homodimer-oligomers revealed by single-molecule imaging and novel G protein-dependent signaling. Scientific Reports. 2017;7:40335. doi: 10.1038/srep40335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castan-Laurell I, Dray C, Attané C, Duparc T, Knauf C, Valet P. Apelin, diabetes, and obesity. Endocrine. 2011;40:1–9. doi: 10.1007/s12020-011-9507-9. [DOI] [PubMed] [Google Scholar]
- Castan-Laurell I, Vítkova M, Daviaud D, Dray C, Kováciková M, Kovacova Z, Hejnova J, Stich V, Valet P. Effect of hypocaloric diet-induced weight loss in obese women on plasma apelin and adipose tissue expression of apelin and APJ. European Journal of Endocrinology. 2008;158:905–910. doi: 10.1530/EJE-08-0039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chandrasekaran B, Kalra PR, Donovan J, Hooper J, Clague JR, McDonagh TA. Myocardial apelin production is reduced in humans with left ventricular systolic dysfunction. Journal of Cardiac Failure. 2010;16:556–561. doi: 10.1016/j.cardfail.2010.02.004. [DOI] [PubMed] [Google Scholar]
- Charles CJ, Rademaker MT, Richards AM. Apelin-13 induces a biphasic haemodynamic response and hormonal activation in normal conscious sheep. Journal of Endocrinology. 2006;189:701–710. doi: 10.1677/joe.1.06804. [DOI] [PubMed] [Google Scholar]
- Chen L, Tao Y, Feng J, Jiang YR. Apelin protects primary rat retinal pericytes from chemical hypoxia-induced apoptosis. Journal of Ophthalmology. 2015;2015:186946. doi: 10.1155/2015/186946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen MM, Ashley EA, Deng DX, Tsalenko A, Deng A, Tabibiazar R, Ben-Dor A, Fenster B, Yang E, King JY, Fowler M, Robbins R, Johnson FL, Bruhn L, McDonagh T, Dargie H, Yakhini Z, Tsao PS, Quertermous T. Novel role for the potent endogenous inotrope apelin in human cardiac dysfunction. Circulation. 2003;108:1432–1439. doi: 10.1161/01.CIR.0000091235.94914.75. [DOI] [PubMed] [Google Scholar]
- Cheng X, Cheng XS, Pang CC. Venous dilator effect of apelin, an endogenous peptide ligand for the orphan APJ receptor, in conscious rats. European Journal of Pharmacology. 2003;470:171–175. doi: 10.1016/s0014-2999(03)01821-1. [DOI] [PubMed] [Google Scholar]
- Chng SC, Ho L, Tian J, Reversade B. ELABELA: a hormone essential for heart development signals via the apelin receptor. Developmental Cell. 2013;27:672–680. doi: 10.1016/j.devcel.2013.11.002. [DOI] [PubMed] [Google Scholar]
- Choe W, Albright A, Sulcove J, Jaffer S, Hesselgesser J, Lavi E, Crino P, Kolson DL. Functional expression of the seven-transmembrane HIV-1 co-receptor APJ in neural cells. Journal of Neurovirology. 2000;6(Suppl 1):S61–69. [PubMed] [Google Scholar]
- Chun HJ, Ali ZA, Kojima Y, Kundu RK, Sheikh AY, Agrawal R, Zheng L, Leeper NJ, Pearl NE, Patterson AJ, Anderson JP, Tsao PS, Lenardo MJ, Ashley EA, Quertermous T. Apelin signaling antagonizes Ang II effects in mouse models of atherosclerosis. Journal of Clinical Investigation. 2008;118:3343–3354. doi: 10.1172/JCI34871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coquerel D, Chagnon F, Sainsily X, Dumont L, Murza A, Cote J, Dumaine R, Sarret P, Marsault E, Salvail D, Auger-Messier M, Lesur O. ELABELA improves cardio-renal outcome in fatal experimental septic shock. Critical Care Medicine. 2017;45:e1139–e1148. doi: 10.1097/CCM.0000000000002639. [DOI] [PubMed] [Google Scholar]
- Cudnoch-Jedrzejewska A, Gomolka R, Szczepanska-Sadowska E, Czarzasta K, Wrzesien R, Koperski L, Puchalska L, Wsol A. High-fat diet and chronic stress reduce central pressor and tachycardic effects of apelin in Sprague-Dawley rats. Clinical and Experimental Pharmacology and Physiology. 2015;42:52–62. doi: 10.1111/1440-1681.12324. [DOI] [PubMed] [Google Scholar]
- Cui RR, Mao DA, Yi L, Wang C, Zhang XX, Xie H, Wu XP, Liao XB, Zhou H, Meng JC, Yuan LQ, Liao EY. Apelin suppresses apoptosis of human vascular smooth muscle cells via APJ/PI3-K/Akt signaling pathways. Amino Acids. 2010;39:1193–1200. doi: 10.1007/s00726-010-0555-x. [DOI] [PubMed] [Google Scholar]
- Daviaud D, Boucher J, Gesta S, Dray C, Guigne C, Quilliot D, Ayav A, Ziegler O, Carpene C, Saulnier-Blache JS, Valet P, Castan-Laurell I. TNF alpha up-regulates apelin expression in human and mouse adipose tissue. FASEB Journal. 2006;20:1528–1530. doi: 10.1096/fj.05-5243fje. [DOI] [PubMed] [Google Scholar]
- Devic E, Paquereau L, Vernier P, Knibiehler B, Audigier Y. Expression of a new G protein-coupled receptor X-msr is associated with an endothelial lineage in Xenopus laevis. Mechanisms of Development. 1996;59:129–140. doi: 10.1016/0925-4773(96)00585-0. [DOI] [PubMed] [Google Scholar]
- Du JH, Li X, Li R, Xu L, Ma RR, Liu SF, Zhang Z, Sun HZ. Elevation of serum apelin-13 associated with proliferative diabetic retinopathy in type 2 diabetic patients. International Journal of Ophthalmology. 2014;7:968–973. doi: 10.3980/j.issn.2222-3959.2014.06.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El Messari S, Iturrioz X, Fassot C, De Mota N, Roesch D, Llorens-Cortes C. Functional dissociation of apelin receptor signaling and endocytosis: implications for the effects of apelin on arterial blood pressure. Journal of Neurochemistry. 2004;90:1290–1301. doi: 10.1111/j.1471-4159.2004.02591.x. [DOI] [PubMed] [Google Scholar]
- Evans NA, Groarke DA, Warrack J, Greenwood CJ, Dodgson K, Milligan G, Wilson S. Visualizing differences in ligand-induced beta-arrestin-GFP interactions and trafficking between three recently characterized G protein-coupled receptors. Journal of Neurochemistry. 2001;77:476–485. doi: 10.1046/j.1471-4159.2001.00269.x. [DOI] [PubMed] [Google Scholar]
- Eyries M, Siegfried G, Ciumas M, Montagne K, Agrapart M, Lebrin F, Soubrier F. Hypoxia-induced apelin expression regulates endothelial cell proliferation and regenerative angiogenesis. Circulation Research. 2008;103:432–440. doi: 10.1161/CIRCRESAHA.108.179333. [DOI] [PubMed] [Google Scholar]
- Falcao-Pires I, Goncalves N, Henriques-Coelho T, Moreira-Goncalves D, Roncon-Albuquerque R, Jr, Leite-Moreira AF. Apelin decreases myocardial injury and improves right ventricular function in monocrotaline-induced pulmonary hypertension. American Journal of Physiology: Heart and Circulatory Physiology. 2009;296:H2007–2014. doi: 10.1152/ajpheart.00089.2009. [DOI] [PubMed] [Google Scholar]
- Fan XF, Xue F, Zhang YQ, Xing XP, Liu H, Mao SZ, Kong XX, Gao YQ, Liu SF, Gong YS. The Apelin-APJ axis is an endogenous counterinjury mechanism in experimental acute lung injury. Chest. 2015;147:969–978. doi: 10.1378/chest.14-1426. [DOI] [PubMed] [Google Scholar]
- Feng JH, Li WM, Wu XP, Tan XY, Gao YH, Han CL, Li SQ, Xie HN. Hemodynamic effect of apelin in a canine model of acute pulmonary thromboembolism. Peptides. 2010;31:1772–1778. doi: 10.1016/j.peptides.2010.06.004. [DOI] [PubMed] [Google Scholar]
- Földes G, Horkay F, Szokodi I, Vuolteenaho O, Ilves M, Lindstedt KA, Mäyränpää M, Sármán B, Seres L, Skoumal R, Lakó-Futó Z, deChâtel R, Ruskoaho H, Tóth M. Circulating and cardiac levels of apelin, the novel ligand of the orphan receptor APJ, in patients with heart failure. Biochemical and Biophysical Research Communications. 2003;308:480–485. doi: 10.1016/s0006-291x(03)01424-4. [DOI] [PubMed] [Google Scholar]
- Fujie S, Sato K, Miyamoto-Mikami E, Hasegawa N, Fujita S, Sanada K, Hamaoka T, Iemitsu M. Reduction of arterial stiffness by exercise training is associated with increasing plasma apelin level in middle-aged and older adults. PLoS One. 2014;9:e93545. doi: 10.1371/journal.pone.0093545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fukushima H, Kobayashi N, Takeshima H, Koguchi W, Ishimitsu T. Effects of olmesartan on Apelin/APJ and Akt/endothelial nitric oxide synthase pathway in Dahl rats with end-stage heart failure. Journal of Cardiovascular Pharmacology. 2010;55:83–88. doi: 10.1097/FJC.0b013e3181c87a82. [DOI] [PubMed] [Google Scholar]
- Gerbier R, Alvear-Perez R, Margathe JF, Flahault A, Couvineau P, Gao J, De Mota N, Dabire H, Li B, Ceraudo E, Hus-Citharel A, Esteoulle L, Bisoo C, Hibert M, Berdeaux A, Iturrioz X, Bonnet D, Llorens-Cortes C. Development of original metabolically stable apelin-17 analogs with diuretic and cardiovascular effects. FASEB Journal. 2017;31:687–700. doi: 10.1096/fj.201600784R. [DOI] [PubMed] [Google Scholar]
- Glassford AJ, Yue P, Sheikh AY, Chun HJ, Zarafshar S, Chan DA, Reaven GM, Quertermous T, Tsao PS. HIF-1 regulates hypoxia- and insulin-induced expression of apelin in adipocytes. American Journal of Physiology Endocrinology and Metabolism. 2007;293:E1590–1596. doi: 10.1152/ajpendo.00490.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomolka RS, Cudnoch-Jedrzejewska A, Czarzasta K, Szczepanska-Sadowska E. Reduction of pressor response to stress by centrally acting apelin in spontaneously hypertensive rats. Journal of Basic and Clinical Physiology and Pharmacology. 2015;26:233–236. doi: 10.1515/jbcpp-2014-0066. [DOI] [PubMed] [Google Scholar]
- Griffiths PR, Lolait SJ, Harris LE, Paton JFR, O’Carroll AM. Vasopressin V1a receptors mediate the hypertensive effects of [Pyr(1)]apelin-13 in the rat rostral ventrolateral medulla. The Journal of Physiology. 2017;595:3303–3318. doi: 10.1113/JP274178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gupta MD, Girish MP, Shah D, Rain M, Mehta V, Tyagi S, Trehan V, Pasha Q. Biochemical and genetic role of apelin in essential hypertension and acute coronary syndrome. International Journal of Cardiology. 2016;223:374–378. doi: 10.1016/j.ijcard.2016.07.242. [DOI] [PubMed] [Google Scholar]
- Gurzu B, Petrescu BC, Costuleanu M, Petrescu G. Interactions between apelin and angiotensin II on rat portal vein. Journal of the Renin-Angiotensin-Aldosterone System. 2006;7:212–216. doi: 10.3317/jraas.2006.040. [DOI] [PubMed] [Google Scholar]
- Habchi M, Duvillard L, Cottet V, Brindisi MC, Bouillet B, Beacco M, Crevisy E, Buffier P, Baillot-Rudoni S, Verges B, Petit JM. Circulating apelin is increased in patients with type 1 or type 2 diabetes and is associated with better glycaemic control. Clinical Endocrinology. 2014;81:696–701. doi: 10.1111/cen.12404. [DOI] [PubMed] [Google Scholar]
- Han S, Wang G, Qi X, Englander EW, Greeley GH., Jr Involvement of a Stat3 binding site in inflammation-induced enteric apelin expression. American Journal of Physiology Gastrointestinal and Liver Physiology. 2008;295:G1068–1078. doi: 10.1152/ajpgi.90493.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han X, Zhang DL, Yin DX, Zhang QD, Liu WH. Apelin-13 deteriorates hypertension in rats after damage of the vascular endothelium by ADMA. Canadian Journal of Physiology and Pharmacology. 2013;91:708–714. doi: 10.1139/cjpp-2013-0046. [DOI] [PubMed] [Google Scholar]
- Hashimoto T, Kihara M, Imai N, Yoshida S, Shimoyamada H, Yasuzaki H, Ishida J, Toya Y, Kiuchi Y, Hirawa N, Tamura K, Yazawa T, Kitamura H, Fukamizu A, Umemura S. Requirement of apelin-apelin receptor system for oxidative stress-linked atherosclerosis. American Journal of Pathology. 2007;171:1705–1712. doi: 10.2353/ajpath.2007.070471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hashimoto T, Kihara M, Ishida J, Imai N, Yoshida S, Toya Y, Fukamizu A, Kitamura H, Umemura S. Apelin stimulates myosin light chain phosphorylation in vascular smooth muscle cells. Arteriosclerosis, Thrombosis, and Vascular Biology. 2006;26:1267–1272. doi: 10.1161/01.ATV.0000218841.39828.91. [DOI] [PubMed] [Google Scholar]
- Heinonen MV, Purhonen AK, Miettinen P, Paakkonen M, Pirinen E, Alhava E, Akerman K, Herzig KH. Apelin, orexin-A and leptin plasma levels in morbid obesity and effect of gastric banding. Regulatory Peptides. 2005;130:7–13. doi: 10.1016/j.regpep.2005.05.003. [DOI] [PubMed] [Google Scholar]
- Ho L, Tan SY, Wee S, Wu Y, Tan SJ, Ramakrishna NB, Chng SC, Nama S, Szczerbinska I, Chan YS, Avery S, Tsuneyoshi N, Ng HH, Gunaratne J, Dunn NR, Reversade B. ELABELA is an endogenous growth factor that sustains hESC self-renewal via the PI3K/AKT pathway. Cell Stem Cell. 2015;17:435–447. doi: 10.1016/j.stem.2015.08.010. [DOI] [PubMed] [Google Scholar]
- Ho L, van Dijk M, Chye STJ, Messerschmidt DM, Chng SC, Ong S, Yi LK, Boussata S, Goh GH, Afink GB, Lim CY, Dunn NR, Solter D, Knowles BB, Reversade B. ELABELA deficiency promotes preeclampsia and cardiovascular malformations in mice. Science. 2017;357:707–713. doi: 10.1126/science.aam6607. [DOI] [PubMed] [Google Scholar]
- Hou J, Wang L, Long H, Wu H, Wu Q, Zhong T, Chen X, Zhou C, Guo T, Wang T. Hypoxia preconditioning promotes cardiac stem cell survival and cardiogenic differentiation in vitro involving activation of the HIF-1alpha/apelin/APJ axis. Stem Cell Research & Therapy. 2017;8:215. doi: 10.1186/s13287-017-0673-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Inui M, Fukui A, Ito Y, Asashima M. Xapelin and Xmsr are required for cardiovascular development in Xenopus laevis. Developmental Biology. 2006;298:188–200. doi: 10.1016/j.ydbio.2006.06.028. [DOI] [PubMed] [Google Scholar]
- Ishimaru Y, Shibagaki F, Yamamuro A, Yoshioka Y, Maeda S. An apelin receptor antagonist prevents pathological retinal angiogenesis with ischemic retinopathy in mice. Scientific Reports. 2017;7:15062. doi: 10.1038/s41598-017-15602-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iturrioz X, Alvear-Perez R, De Mota N, Franchet C, Guillier F, Leroux V, Dabire H, Le Jouan M, Chabane H, Gerbier R, Bonnet D, Berdeaux A, Maigret B, Galzi JL, Hibert M, Llorens-Cortes C. Identification and pharmacological properties of E339-3D6, the first nonpeptidic apelin receptor agonist. FASEB Journal. 2010;24:1506–1517. doi: 10.1096/fj.09-140715. [DOI] [PubMed] [Google Scholar]
- Japp AG, Cruden NL, Amer DA, Li VK, Goudie EB, Johnston NR, Sharma S, Neilson I, Webb DJ, Megson IL, Flapan AD, Newby DE. Vascular effects of apelin in vivo in man. Journal of the American College of Cardiology. 2008;52:908–913. doi: 10.1016/j.jacc.2008.06.013. [DOI] [PubMed] [Google Scholar]
- Japp AG, Cruden NL, Barnes G, van Gemeren N, Mathews J, Adamson J, Johnston NR, Denvir MA, Megson IL, Flapan AD, Newby DE. Acute cardiovascular effects of apelin in humans: potential role in patients with chronic heart failure. Circulation. 2010;121:1818–1827. doi: 10.1161/CIRCULATIONAHA.109.911339. [DOI] [PubMed] [Google Scholar]
- Jia YX, Lu ZF, Zhang J, Pan CS, Yang JH, Zhao J, Yu F, Duan XH, Tang CS, Qi YF. Apelin activates L-arginine/nitric oxide synthase/nitric oxide pathway in rat aortas. Peptides. 2007;28:2023–2029. doi: 10.1016/j.peptides.2007.07.016. [DOI] [PubMed] [Google Scholar]
- Jia ZQ, Hou L, Leger A, Wu I, Kudej AB, Stefano J, Jiang C, Pan CQ, Akita GY. Cardiovascular effects of a PEGylated apelin. Peptides. 2012;38:181–188. doi: 10.1016/j.peptides.2012.09.003. [DOI] [PubMed] [Google Scholar]
- Kadoglou NP, Lampropoulos S, Kapelouzou A, Gkontopoulos A, Theofilogiannakos EK, Fotiadis G, Kottas G. Serum levels of apelin and ghrelin in patients with acute coronary syndromes and established coronary artery disease--KOZANI STUDY. Translational Research: The Journal of Laboratory and Clinical Medicine. 2010;155:238–246. doi: 10.1016/j.trsl.2010.01.004. [DOI] [PubMed] [Google Scholar]
- Kagiyama S, Fukuhara M, Matsumura K, Lin Y, Fujii K, Iida M. Central and peripheral cardiovascular actions of apelin in conscious rats. Regulatory Peptides. 2005;125:55–59. doi: 10.1016/j.regpep.2004.07.033. [DOI] [PubMed] [Google Scholar]
- Kalea AZ, Batlle D. Apelin and ACE2 in cardiovascular disease. Current Opinion in Investigational Drugs. 2010;11:273–282. [PubMed] [Google Scholar]
- Kang Y, Kim J, Anderson JP, Wu J, Gleim SR, Kundu RK, McLean DL, Kim JD, Park H, Jin SW, Hwa J, Quertermous T, Chun HJ. Apelin-APJ signaling is a critical regulator of endothelial MEF2 activation in cardiovascular development. Circulation Research. 2013;113:22–31. doi: 10.1161/CIRCRESAHA.113.301324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katugampola SD, Maguire JJ, Matthewson SR, Davenport AP. [(125)I]-(Pyr(1))Apelin-13 is a novel radioligand for localizing the APJ orphan receptor in human and rat tissues with evidence for a vasoconstrictor role in man. British Journal of Pharmacology. 2001;132:1255–1260. doi: 10.1038/sj.bjp.0703939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kawamata Y, Habata Y, Fukusumi S, Hosoya M, Fujii R, Hinuma S, Nishizawa N, Kitada C, Onda H, Nishimura O, Fujino M. Molecular properties of apelin: tissue distribution and receptor binding. Biochimica et Biophysica Acta: Protein Structure and Molecular Enzymology. 2001;1538:162–171. doi: 10.1016/s0167-4889(00)00143-9. [DOI] [PubMed] [Google Scholar]
- Kerkela R, Karsikas S, Szabo Z, Serpi R, Magga J, Gao E, Alitalo K, Anisimov A, Sormunen R, Pietila I, Vainio L, Koch WJ, Kivirikko KI, Myllyharju J, Koivunen P. Activation of hypoxia response in endothelial cells contributes to ischemic cardioprotection. Molecular and Cellular Biology. 2013;33:3321–3329. doi: 10.1128/MCB.00432-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khan P, Maloney PR, Hedrick M, Gosalia P, Milewski M, Li L, Roth GP, Sergienko E, Suyama E, Sugarman E, Nguyen K, Mehta A, Vasile S, Su Y, Shi S, Stonich D, Nguyen H, Zeng FY, Novo AM, Vicchiarelli M, Diwan J, Chung TDY, Pinkerton AB, Smith LH. Probe Reports from the NIH Molecular Libraries Program. Bethesda (MD): 2010. Functional agonists of the apelin (APJ) receptor. [PubMed] [Google Scholar]
- Kidoya H, Kunii N, Naito H, Muramatsu F, Okamoto Y, Nakayama T, Takakura N. The apelin/APJ system induces maturation of the tumor vasculature and improves the efficiency of immune therapy. Oncogene. 2012;31:3254–3264. doi: 10.1038/onc.2011.489. [DOI] [PubMed] [Google Scholar]
- Kim J, Kang Y, Kojima Y, Lighthouse JK, Hu X, Aldred MA, McLean DL, Park H, Comhair SA, Greif DM, Erzurum SC, Chun HJ. An endothelial apelin-FGF link mediated by miR-424 and miR-503 is disrupted in pulmonary arterial hypertension. Nature Medicine. 2013;19:74–82. doi: 10.1038/nm.3040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kleinz MJ, Davenport AP. Immunocytochemical localization of the endogenous vasoactive peptide apelin to human vascular and endocardial endothelial cells. Regulatory Peptides. 2004;118:119–125. doi: 10.1016/j.regpep.2003.11.002. [DOI] [PubMed] [Google Scholar]
- Kleinz MJ, Skepper JN, Davenport AP. Immunocytochemical localisation of the apelin receptor, APJ, to human cardiomyocytes, vascular smooth muscle and endothelial cells. Regulatory Peptides. 2005;126:233–240. doi: 10.1016/j.regpep.2004.10.019. [DOI] [PubMed] [Google Scholar]
- Koguchi W, Kobayashi N, Takeshima H, Ishikawa M, Sugiyama F, Ishimitsu T. Cardioprotective effect of apelin-13 on cardiac performance and remodeling in end-stage heart failure. Circulation Journal. 2012;76:137–144. doi: 10.1253/circj.cj-11-0689. [DOI] [PubMed] [Google Scholar]
- Kong HL, Li ZQ, Zhao SM, Yuan L, Miao ZL, Liu Y, Guan RM. Apelin-APJ effects of ginsenoside-Rb1 depending on hypoxia-induced factor 1alpha in hypoxia neonatal cardiomyocytes. Chinese Journal of Integrative Medicine. 2015;21:139–146. doi: 10.1007/s11655-014-1774-2. [DOI] [PubMed] [Google Scholar]
- Lee DK, Cheng R, Nguyen T, Fan T, Kariyawasam AP, Liu Y, Osmond DH, George SR, O’Dowd BF. Characterization of apelin, the ligand for the APJ receptor. Journal of Neurochemistry. 2000;74:34–41. doi: 10.1046/j.1471-4159.2000.0740034.x. [DOI] [PubMed] [Google Scholar]
- Lee DK, Saldivia VR, Nguyen T, Cheng R, George SR, O’Dowd BF. Modification of the terminal residue of apelin-13 antagonizes its hypotensive action. Endocrinology. 2005;146:231–236. doi: 10.1210/en.2004-0359. [DOI] [PubMed] [Google Scholar]
- Li F, Li L, Qin X, Pan W, Feng F, Chen F, Zhu B, Liao D, Tanowitz H, Albanese C, Chen L. Apelin-induced vascular smooth muscle cell proliferation: the regulation of cyclin D1. Frontiers in Bioscience. 2008a;13:3786–3792. doi: 10.2741/2967. [DOI] [PubMed] [Google Scholar]
- Li L, Li L, Xie F, Zhang Z, Guo Y, Tang G, Lv D, Lu Q, Chen L, Li J. Jagged-1/Notch3 signaling transduction pathway is involved in apelin-13-induced vascular smooth muscle cells proliferation. Acta Biochimica Biophysica Sinica (Shanghai) 2013a;45:875–881. doi: 10.1093/abbs/gmt085. [DOI] [PubMed] [Google Scholar]
- Li L, Zeng H, Hou X, He X, Chen JX. Myocardial injection of apelin-overexpressing bone marrow cells improves cardiac repair via upregulation of Sirt3 after myocardial infarction. PLoS One. 2013b;8:e71041. doi: 10.1371/journal.pone.0071041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Chen J, Bai B, Du H, Liu Y, Liu H. Heterodimerization of human apelin and kappa opioid receptors: roles in signal transduction. Cellular Signalling. 2012;24:991–1001. doi: 10.1016/j.cellsig.2011.12.012. [DOI] [PubMed] [Google Scholar]
- Li Z, Bai Y, Hu J. Reduced apelin levels in stable angina. Internal Medicine. 2008b;47:1951–1955. doi: 10.2169/internalmedicine.47.1287. [DOI] [PubMed] [Google Scholar]
- Liu C, Su T, Li F, Li L, Qin X, Pan W, Feng F, Chen F, Liao D, Chen L. PI3K/Akt signaling transduction pathway is involved in rat vascular smooth muscle cell proliferation induced by apelin-13. Acta Biochimica Biophysica Sinica (Shanghai) 2010;42:396–402. doi: 10.1093/abbs/gmq035. [DOI] [PubMed] [Google Scholar]
- Liu QF, Yu HW, You L, Liu MX, Li KY, Tao GZ. Apelin-13-induced proliferation and migration induced of rat vascular smooth muscle cells is mediated by the upregulation of Egr-1. Biochemical and Biophysical Research Communications. 2013;439:235–240. doi: 10.1016/j.bbrc.2013.08.051. [DOI] [PubMed] [Google Scholar]
- Luo K, Long H, Xu B, Luo Y. Apelin attenuates postburn sepsis via a phosphatidylinositol 3-kinase/protein kinase B dependent mechanism: A randomized animal study. International Journal of Surgery. 2015;21:22–27. doi: 10.1016/j.ijsu.2015.06.072. [DOI] [PubMed] [Google Scholar]
- Macaluso NJ, Pitkin SL, Maguire JJ, Davenport AP, Glen RC. Discovery of a competitive apelin receptor (APJ) antagonist. ChemMedChem. 2011;6:1017–1023. doi: 10.1002/cmdc.201100069. [DOI] [PubMed] [Google Scholar]
- Maguire JJ, Kleinz MJ, Pitkin SL, Davenport AP. [Pyr1]apelin-13 identified as the predominant apelin isoform in the human heart: vasoactive mechanisms and inotropic action in disease. Hypertension. 2009;54:598–604. doi: 10.1161/HYPERTENSIONAHA.109.134619. [DOI] [PubMed] [Google Scholar]
- Maloney PR, Khan P, Hedrick M, Gosalia P, Milewski M, Li L, Roth GP, Sergienko E, Suyama E, Sugarman E, Nguyen K, Mehta A, Vasile S, Su Y, Stonich D, Nguyen H, Zeng FY, Novo AM, Vicchiarelli M, Diwan J, Chung TD, Smith LH, Pinkerton AB. Discovery of 4-oxo-6-((pyrimidin-2-ylthio)methyl)-4H-pyran-3-yl 4-nitrobenzoate (ML221) as a functional antagonist of the apelin (APJ) receptor. Bioorganic and Medicinal Chemistry Letters. 2012;22:6656–6660. doi: 10.1016/j.bmcl.2012.08.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Margathe JF, Iturrioz X, Alvear-Perez R, Marsol C, Riche S, Chabane H, Tounsi N, Kuhry M, Heissler D, Hibert M, Llorens-Cortes C, Bonnet D. Structure-activity relationship studies toward the discovery of selective apelin receptor agonists. Journal of Medicinal Chemistry. 2014;57:2908–2919. doi: 10.1021/jm401789v. [DOI] [PubMed] [Google Scholar]
- Masri B, Morin N, Pedebernade L, Knibiehler B, Audigier Y. The apelin receptor is coupled to Gi1 or Gi2 protein and is differentially desensitized by apelin fragments. Journal of Biological Chemistry. 2006;281:18317–18326. doi: 10.1074/jbc.M600606200. [DOI] [PubMed] [Google Scholar]
- McKinnie SM, Fischer C, Tran KM, Wang W, Mosquera F, Oudit GY, Vederas JC. The metalloprotease neprilysin degrades and inactivates apelin peptides. ChemBioChem. 2016;17:1495–1498. doi: 10.1002/cbic.201600244. [DOI] [PubMed] [Google Scholar]
- McKinnie SMK, Wang W, Fischer C, McDonald T, Kalin KR, Iturrioz X, Llorens-Cortes C, Oudit GY, Vederas JC. Synthetic modification within the “RPRL” region of apelin peptides: impact on cardiovascular activity and stability to neprilysin and plasma degradation. Journal of Medicinal Chemistry. 2017;60:6408–6427. doi: 10.1021/acs.jmedchem.7b00723. [DOI] [PubMed] [Google Scholar]
- Medhurst AD, Jennings CA, Robbins MJ, Davis RP, Ellis C, Winborn KY, Lawrie KW, Hervieu G, Riley G, Bolaky JE, Herrity NC, Murdock P, Darker JG. Pharmacological and immunohistochemical characterization of the APJ receptor and its endogenous ligand apelin. Journal of Neurochemistry. 2003;84:1162–1172. doi: 10.1046/j.1471-4159.2003.01587.x. [DOI] [PubMed] [Google Scholar]
- Modgil A, Guo L, O’Rourke ST, Sun C. Apelin-13 inhibits large-conductance Ca2+-activated K+ channels in cerebral artery smooth muscle cells via a PI3-kinase dependent mechanism. PLoS One. 2013;8:e83051. doi: 10.1371/journal.pone.0083051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mughal A, Sun C, O’Rourke ST. Apelin reduces nitric oxide-induced relaxation of cerebral arteries by inhibiting activation of large-conductance, calcium-activated K Channels. Journal of Cardiovascular Pharmacology. 2018;71:223–232. doi: 10.1097/FJC.0000000000000563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mughal A, Sun C, O’Rourke ST. Activation of large conductance, calcium-activated K channels by nitric oxide mediates apelin-induced relaxation of isolated rat coronary arteries. Journal of Pharmacology and Experimental Therapeutics. doi: 10.1124/jpet.118.248682. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murga C, Laguinge L, Wetzker R, Cuadrado A, Gutkind JS. Activation of Akt/protein kinase B by G protein-coupled receptors. A role for alpha and beta gamma subunits of heterotrimeric G proteins acting through phosphatidylinositol-3-OH kinasegamma. Journal of Biological Chemistry. 1998;273:19080–19085. doi: 10.1074/jbc.273.30.19080. [DOI] [PubMed] [Google Scholar]
- Murza A, Sainsily X, Coquerel D, Cote J, Marx P, Besserer-Offroy E, Longpre JM, Laine J, Reversade B, Salvail D, Leduc R, Dumaine R, Lesur O, Auger-Messier M, Sarret P, Marsault E. Discovery and structure-activity relationship of a bioactive fragment of ELABELA that modulates vascular and cardiac functions. Journal of Medicinal Chemistry. 2016;59:2962–2972. doi: 10.1021/acs.jmedchem.5b01549. [DOI] [PubMed] [Google Scholar]
- Nagano K, Ishida J, Unno M, Matsukura T, Fukamizu A. Apelin elevates blood pressure in ICR mice with L-NAME induced endothelial dysfunction. Molecular Medicine Reports. 2013;7:1371–1375. doi: 10.3892/mmr.2013.1378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Najafipour H, Soltani Hekmat A, Nekooian AA, Esmaeili-Mahani S. Apelin receptor expression in ischemic and non- ischemic kidneys and cardiovascular responses to apelin in chronic two-kidney-one-clip hypertension in rats. Regulatory Peptides. 2012;178:43–50. doi: 10.1016/j.regpep.2012.06.006. [DOI] [PubMed] [Google Scholar]
- Najafipour H, Vakili A, Shahouzehi B, Soltani Hekmat A, Masoomi Y, Yeganeh Hajahmadi M, Esmaeli-Mahani S. Investigation of changes in apelin receptor mRNA and protein expression in the myocardium and aorta of rats with two-kidney, one-clip (2K1C) Goldblatt hypertension. Journal of Physiology and Biochemistry. 2015;71:165–175. doi: 10.1007/s13105-015-0394-z. [DOI] [PubMed] [Google Scholar]
- Narayanan S, Harris DL, Maitra R, Runyon SP. Regulation of the apelinergic system and its potential in cardiovascular disease: peptides and small molecules as tools for discovery. Journal of Medicinal Chemistry. 2015;58:7913–7927. doi: 10.1021/acs.jmedchem.5b00527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Carroll AM, Salih S, Griffiths PR, Bijabhai A, Knepper MA, Lolait SJ. Expression and functional implications of the renal apelinergic system in rodents. PLoS One. 2017;12:e0183094. doi: 10.1371/journal.pone.0183094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Dowd BF, Heiber M, Chan A, Heng HH, Tsui LC, Kennedy JL, Shi X, Petronis A, George SR, Nguyen T. A human gene that shows identity with the gene encoding the angiotensin receptor is located on chromosome 11. Gene. 1993;136:355–360. doi: 10.1016/0378-1119(93)90495-o. [DOI] [PubMed] [Google Scholar]
- O’Rourke ST, Vanhoutte PM, Miller VM. Biology of blood vessels: vascular pharmacology. In: Creager MA, Dzau VJ, Loscalzo J, editors. Vascular Medicine. 3. Philadelphia: Saunders/Elsevier; 2006. pp. 71–100. [Google Scholar]
- Pauli A, Norris ML, Valen E, Chew GL, Gagnon JA, Zimmerman S, Mitchell A, Ma J, Dubrulle J, Reyon D, Tsai SQ, Joung JK, Saghatelian A, Schier AF. Toddler: an embryonic signal that promotes cell movement via Apelin receptors. Science. 2014;343:1248636. doi: 10.1126/science.1248636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peltonen T, Napankangas J, Vuolteenaho O, Ohtonen P, Soini Y, Juvonen T, Satta J, Ruskoaho H, Taskinen P. Apelin and its receptor APJ in human aortic valve stenosis. Journal of Heart Valve Disease. 2009;18:644–652. [PubMed] [Google Scholar]
- Perjes A, Kilpio T, Ulvila J, Magga J, Alakoski T, Szabo Z, Vainio L, Halmetoja E, Vuolteenaho O, Petaja-Repo U, Szokodi I, Kerkela R. Characterization of apela, a novel endogenous ligand of apelin receptor, in the adult heart. Basic Research in Cardiology. 2016;111:2. doi: 10.1007/s00395-015-0521-6. [DOI] [PubMed] [Google Scholar]
- Pitkin SL, Maguire JJ, Bonner TI, Davenport AP. International Union of Basic and Clinical Pharmacology. LXXIV. Apelin receptor nomenclature, distribution, pharmacology, and function. Pharmacological Reviews. 2010a;62:331–342. doi: 10.1124/pr.110.002949. [DOI] [PubMed] [Google Scholar]
- Pitkin SL, Maguire JJ, Kuc RE, Davenport AP. Modulation of the apelin/APJ system in heart failure and atherosclerosis in man. British Journal of Pharmacology. 2010b;160:1785–1795. doi: 10.1111/j.1476-5381.2010.00821.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pope GR, Roberts EM, Lolait SJ, O’Carroll AM. Central and peripheral apelin receptor distribution in the mouse: species differences with rat. Peptides. 2012;33:139–148. doi: 10.1016/j.peptides.2011.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Read C, Fitzpatrick CM, Yang P, Kuc RE, Maguire JJ, Glen RC, Foster RE, Davenport AP. Cardiac action of the first G protein biased small molecule apelin agonist. Biochemical Pharmacology. 2016;116:63–72. doi: 10.1016/j.bcp.2016.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reaux-Le Goazigo A, Morinville A, Burlet A, Llorens-Cortes C, Beaudet A. Dehydration-induced cross-regulation of apelin and vasopressin immunoreactivity levels in magnocellular hypothalamic neurons. Endocrinology. 2004;145:4392–4400. doi: 10.1210/en.2004-0384. [DOI] [PubMed] [Google Scholar]
- Reaux A, De Mota N, Skultetyova I, Lenkei Z, El Messari S, Gallatz K, Corvol P, Palkovits M, Llorens-Cortès C. Physiological role of a novel neuropeptide, apelin, and its receptor in the rat brain. Journal of Neurochemistry. 2001;77:1085–1096. doi: 10.1046/j.1471-4159.2001.00320.x. [DOI] [PubMed] [Google Scholar]
- Rostamzadeh F, Najafipour H, Yeganeh-Hajahmadi M, Joukar S. Opioid receptors mediate inotropic and depressor effects of apelin in rats with 2K1C-induced chronic renovascular hypertension. Clinical and Experimental Pharmacology and Physiology. 2018;45:187–197. doi: 10.1111/1440-1681.12860. [DOI] [PubMed] [Google Scholar]
- Saint-Geniez M, Masri B, Malecaze F, Knibiehler B, Audigier Y. Expression of the murine msr/apj receptor and its ligand apelin is upregulated during formation of the retinal vessels. Mechanisms of Development. 2002;110:183–186. doi: 10.1016/s0925-4773(01)00558-5. [DOI] [PubMed] [Google Scholar]
- Salcedo A, Garijo J, Monge L, Fernández N, Luis García-Villalón A, Sánchez Turrión V, Cuervas-Mons V, Diéguez G. Apelin effects in human splanchnic arteries. Role of nitric oxide and prostanoids. Regulatory Peptides. 2007;144:50–55. doi: 10.1016/j.regpep.2007.06.005. [DOI] [PubMed] [Google Scholar]
- Sato T, Sato C, Kadowaki A, Watanabe H, Ho L, Ishida J, Yamaguchi T, Kimura A, Fukamizu A, Penninger JM, Reversade B, Ito H, Imai Y, Kuba K. ELABELA-APJ axis protects from pressure overload heart failure and angiotensin II-induced cardiac damage. Cardiovascular Research. 2017;113:760–769. doi: 10.1093/cvr/cvx061. [DOI] [PubMed] [Google Scholar]
- Sato T, Suzuki T, Watanabe H, Kadowaki A, Fukamizu A, Liu PP, Kimura A, Ito H, Penninger JM, Imai Y, Kuba K. Apelin is a positive regulator of ACE2 in failing hearts. Journal of Clinical Investigation. 2013;123:5203–5211. doi: 10.1172/JCI69608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sawane M, Kajiya K, Kidoya H, Takagi M, Muramatsu F, Takakura N. Apelin inhibits diet-induced obesity by enhancing lymphatic and blood vessel integrity. Diabetes. 2013;62:1970–1980. doi: 10.2337/db12-0604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schinzari F, Veneziani A, Mores N, Barini A, Di Daniele N, Cardillo C, Tesauro M. Beneficial effects of apelin on vascular function in patients with central obesity. Hypertension. 2017;69:942–949. doi: 10.1161/HYPERTENSIONAHA.116.08916. [DOI] [PubMed] [Google Scholar]
- Schreiber CA, Holditch SJ, Generous A, Ikeda Y. Sustained ELABELA gene therapy in high-salt diet-induced hypertensive rats. Current Gene Therapy. 2017;16:349–360. doi: 10.2174/1566523217666161121111906. [DOI] [PubMed] [Google Scholar]
- Seyedabadi M, Goodchild AK, Pilowsky PM. Site-specific effects of apelin-13 in the rat medulla oblongata on arterial pressure and respiration. Autonomic Neuroscience. 2002;101:32–38. doi: 10.1016/s1566-0702(02)00178-9. [DOI] [PubMed] [Google Scholar]
- Shan PF, Lu Y, Cui RR, Jiang Y, Yuan LQ, Liao EY. Apelin attenuates the osteoblastic differentiation of vascular smooth muscle cells. PLoS One. 2011;6:e17938. doi: 10.1371/journal.pone.0017938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheikh AY, Chun HJ, Glassford AJ, Kundu RK, Kutschka I, Ardigo D, Hendry SL, Wagner RA, Chen MM, Ali ZA, Yue P, Huynh DT, Connolly AJ, Pelletier MP, Tsao PS, Robbins RC, Quertermous T. In vivo genetic profiling and cellular localization of apelin reveals a hypoxia-sensitive, endothelial-centered pathway activated in ischemic heart failure. American Journal of Physiology: Heart and Circulatory Physiology. 2008;294:H88–98. doi: 10.1152/ajpheart.00935.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi H, Yuan L, Yang H, Zang A. The mechanism of all-trans retinoic acid in the regulation of apelin expression in vascular endothelial cells. Bioscience Reports. 2017:37. doi: 10.1042/BSR20170684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siddiquee K, Hampton J, Khan S, Zadory D, Gleaves L, Vaughan DE, Smith LH. Apelin protects against angiotensin II-induced cardiovascular fibrosis and decreases plasminogen activator inhibitor type-1 production. Journal of Hypertension. 2011;29:724–731. doi: 10.1097/HJH.0b013e32834347de. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siddiquee K, Hampton J, McAnally D, May L, Smith L. The apelin receptor inhibits the angiotensin II type 1 receptor via allosteric trans-inhibition. British Journal of Pharmacology. 2013;168:1104–1117. doi: 10.1111/j.1476-5381.2012.02192.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Staff AC. An introduction to cell migration and invasion. Scandinavian Journal of Clinical and Laboratory Investigation. 2001;61:257–268. doi: 10.1080/00365510152378978. [DOI] [PubMed] [Google Scholar]
- Sun X, Iida S, Yoshikawa A, Senbonmatsu R, Imanaka K, Maruyama K, Nishimura S, Inagami T, Senbonmatsu T. Non-activated APJ suppresses the angiotensin II type 1 receptor, whereas apelin-activated APJ acts conversely. Hypertension Research. 2011;34:701–706. doi: 10.1038/hr.2011.19. [DOI] [PubMed] [Google Scholar]
- Szokodi I, Tavi P, Foldes G, Voutilainen-Myllyla S, Ilves M, Tokola H, Pikkarainen S, Piuhola J, Rysa J, Toth M, Ruskoaho H. Apelin, the novel endogenous ligand of the orphan receptor APJ, regulates cardiac contractility. Circulation Research. 2002;91:434–440. doi: 10.1161/01.res.0000033522.37861.69. [DOI] [PubMed] [Google Scholar]
- Tang SY, Xie H, Yuan LQ, Luo XH, Huang J, Cui RR, Zhou HD, Wu XP, Liao EY. Apelin stimulates proliferation and suppresses apoptosis of mouse osteoblastic cell line MC3T3-E1 via JNK and PI3-K/Akt signaling pathways. Peptides. 2007;28:708–718. doi: 10.1016/j.peptides.2006.10.005. [DOI] [PubMed] [Google Scholar]
- Tatemoto K, Hosoya M, Habata Y, Fujii R, Kakegawa T, Zou MX, Kawamata Y, Fukusumi S, Hinuma S, Kitada C, Kurokawa T, Onda H, Fujino M. Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochemical and Biophysical Research Communications. 1998;251:471–476. doi: 10.1006/bbrc.1998.9489. [DOI] [PubMed] [Google Scholar]
- Tatemoto K, Takayama K, Zou MX, Kumaki I, Zhang W, Kumano K, Fujimiya M. The novel peptide apelin lowers blood pressure via a nitric oxide-dependent mechanism. Regulatory Peptides. 2001;99:87–92. doi: 10.1016/s0167-0115(01)00236-1. [DOI] [PubMed] [Google Scholar]
- Vickers C, Hales P, Kaushik V, Dick L, Gavin J, Tang J, Godbout K, Parsons T, Baronas E, Hsieh F, Acton S, Patane M, Nichols A, Tummino P. Hydrolysis of biological peptides by human angiotensin-converting enzyme-related carboxypeptidase. Journal of Biological Chemistry. 2002;277:14838–14843. doi: 10.1074/jbc.M200581200. [DOI] [PubMed] [Google Scholar]
- Wang C, Wen J, Zhou Y, Li L, Cui X, Wang J, Pan L, Ye Z, Liu P, Wu L. Apelin induces vascular smooth muscle cells migration via a PI3K/Akt/FoxO3a/MMP-2 pathway. International Journal of Biochemistry and Cell Biology. 2015a;69:173–182. doi: 10.1016/j.biocel.2015.10.015. [DOI] [PubMed] [Google Scholar]
- Wang LY, Zhang DL, Zheng JF, Zhang Y, Zhang QD, Liu WH. Apelin-13 passes through the ADMA-damaged endothelial barrier and acts on vascular smooth muscle cells. Peptides. 2011;32:2436–2443. doi: 10.1016/j.peptides.2011.10.001. [DOI] [PubMed] [Google Scholar]
- Wang W, McKinnie SM, Farhan M, Paul M, McDonald T, McLean B, Llorens-Cortes C, Hazra S, Murray AG, Vederas JC, Oudit GY. Angiotensin-converting enzyme 2 metabolizes and partially inactivates pyr-apelin-13 and apelin-17: physiological effects in the cardiovascular system. Hypertension. 2016;68:365–377. doi: 10.1161/HYPERTENSIONAHA.115.06892. [DOI] [PubMed] [Google Scholar]
- Wang W, McKinnie SM, Patel VB, Haddad G, Wang Z, Zhabyeyev P, Das SK, Basu R, McLean B, Kandalam V, Penninger JM, Kassiri Z, Vederas JC, Murray AG, Oudit GY. Loss of Apelin exacerbates myocardial infarction adverse remodeling and ischemia-reperfusion injury: therapeutic potential of synthetic Apelin analogues. Journal of American Heart Association. 2013;2:e000249. doi: 10.1161/JAHA.113.000249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z, Yu D, Wang M, Wang Q, Kouznetsova J, Yang R, Qian K, Wu W, Shuldiner A, Sztalryd C, Zou M, Zheng W, Gong DW. Elabela-apelin receptor signaling pathway is functional in mammalian systems. Scientific Reports. 2015b;5:8170. doi: 10.1038/srep08170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang P, Kuc RE, Brame AL, Dyson A, Singer M, Glen RC, Cheriyan J, Wilkinson IB, Davenport AP, Maguire JJ. [Pyr1]Apelin-13(1–12) is a biologically active ACE2 metabolite of the endogenous cardiovascular peptide [Pyr1]Apelin-13. Frontiers in Neuroscience. 2017a;11:92. doi: 10.3389/fnins.2017.00092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang P, Read C, Kuc RE, Buonincontri G, Southwood M, Torella R, Upton PD, Crosby A, Sawiak SJ, Carpenter TA, Glen RC, Morrell NW, Maguire JJ, Davenport AP. Elabela/Toddler is an endogenous agonist of the apelin APJ receptor in the adultcardiovascular system, and exogenous administration of the peptide compensates for the downregulation of its expression in pulmonary arterial hypertension. Circulation. 2017b;135:1160–1173. doi: 10.1161/CIRCULATIONAHA.116.023218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao F, Modgil A, Zhang Q, Pingili A, Singh N, O’Rourke ST, Sun C. Pressor effect of apelin-13 in the rostral ventrolateral medulla: role of NAD(P)H oxidase-derived superoxide. Journal of Pharmacology and Experimental Therapeutics. 2011;336:372–380. doi: 10.1124/jpet.110.174102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yeganeh-Hajahmadi M, Najafipour H, Rostamzadeh F. The differential effects of low and high doses of apelin through opioid receptors on the blood pressure of rats with renovascular hypertension. Hypertension Research. 2017;40:732–737. doi: 10.1038/hr.2017.28. [DOI] [PubMed] [Google Scholar]
- Zeng H, He X, Hou X, Li L, Chen JX. Apelin gene therapy increases myocardial vascular density and ameliorates diabetic cardiomyopathy via upregulation of sirtuin 3. American Journal of Physiology: Heart and Circulatory Physiology. 2014;306:H585–597. doi: 10.1152/ajpheart.00821.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeng XX, Wilm TP, Sepich DS, Solnica-Krezel L. Apelin and its receptor control heart field formation during zebrafish gastrulation. Developmental Cell. 2007;12:391–402. doi: 10.1016/j.devcel.2007.01.011. [DOI] [PubMed] [Google Scholar]
- Zhang BH, Wang W, Wang H, Yin J, Zeng XJ. Promoting effects of the adipokine, apelin, on diabetic nephropathy. PLoS One. 2013;8:e60457. doi: 10.1371/journal.pone.0060457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang F, Sun HJ, Xiong XQ, Chen Q, Li YH, Kang YM, Wang JJ, Gao XY, Zhu GQ. Apelin-13 and APJ in paraventricular nucleus contribute to hypertension via sympathetic activation and vasopressin release in spontaneously hypertensive rats. Acta Physiologica (Oxford) 2014a;212:17–27. doi: 10.1111/apha.12342. [DOI] [PubMed] [Google Scholar]
- Zhang H, Gong Y, Wang Z, Jiang L, Chen R, Fan X, Zhu H, Han L, Li X, Xiao J, Kong X. Apelin inhibits the proliferation and migration of rat PASMCs via the activation of PI3K/Akt/mTOR signal and the inhibition of autophagy under hypoxia. Journal of Cellular and Molecular Medicine. 2014b;18:542–553. doi: 10.1111/jcmm.12208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Liu Q, Hu X, Fang Z, Huang F, Tang L, Zhou S. Apelin/APJ signaling promotes hypoxia-induced proliferation of endothelial progenitor cells via phosphoinositide-3 kinase/Akt signaling. Molecular Medicine Reports. 2015;12:3829–3834. doi: 10.3892/mmr.2015.3866. [DOI] [PubMed] [Google Scholar]
- Zhang Q, Yao F, Raizada MK, O’Rourke ST, Sun C. Apelin gene transfer into the rostral ventrolateral medulla induces chronic blood pressure elevation in normotensive rats. Circulation Research. 2009;104:1421–1428. doi: 10.1161/CIRCRESAHA.108.192302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhen EY, Higgs RE, Gutierrez JA. Pyroglutamyl apelin-13 identified as the major apelin isoform in human plasma. Analytical Biochemistry. 2013;442:1–9. doi: 10.1016/j.ab.2013.07.006. [DOI] [PubMed] [Google Scholar]
- Zhong JC, Huang Y, Yung LM, Lau CW, Leung FP, Wong WT, Lin SG, Yu XY. The novel peptide apelin regulates intrarenal artery tone in diabetic mice. Regulatory Peptides. 2007a;144:109–114. doi: 10.1016/j.regpep.2007.06.010. [DOI] [PubMed] [Google Scholar]
- Zhong JC, Yu XY, Huang Y, Yung LM, Lau CW, Lin SG. Apelin modulates aortic vascular tone via endothelial nitric oxide synthase phosphorylation pathway in diabetic mice. Cardiovascular Research. 2007b;74:388–395. doi: 10.1016/j.cardiores.2007.02.002. [DOI] [PubMed] [Google Scholar]