Skip to main content
Pharmacological Reviews logoLink to Pharmacological Reviews
. 2014 Jan;66(1):308–333. doi: 10.1124/pr.112.007203

Cardiac Alpha1-Adrenergic Receptors: Novel Aspects of Expression, Signaling Mechanisms, Physiologic Function, and Clinical Importance

Timothy D O’Connell 1, Brian C Jensen 1, Anthony J Baker 1, Paul C Simpson 1,
Editor: Paul A Insel
PMCID: PMC3880467  PMID: 24368739

Abstract

Adrenergic receptors (AR) are G-protein-coupled receptors (GPCRs) that have a crucial role in cardiac physiology in health and disease. Alpha1-ARs signal through Gαq, and signaling through Gq, for example, by endothelin and angiotensin receptors, is thought to be detrimental to the heart. In contrast, cardiac alpha1-ARs mediate important protective and adaptive functions in the heart, although alpha1-ARs are only a minor fraction of total cardiac ARs. Cardiac alpha1-ARs activate pleiotropic downstream signaling to prevent pathologic remodeling in heart failure. Mechanisms defined in animal and cell models include activation of adaptive hypertrophy, prevention of cardiac myocyte death, augmentation of contractility, and induction of ischemic preconditioning. Surprisingly, at the molecular level, alpha1-ARs localize to and signal at the nucleus in cardiac myocytes, and, unlike most GPCRs, activate “inside-out” signaling to cause cardioprotection. Contrary to past opinion, human cardiac alpha1-AR expression is similar to that in the mouse, where alpha1-AR effects are seen most convincingly in knockout models. Human clinical studies show that alpha1-blockade worsens heart failure in hypertension and does not improve outcomes in heart failure, implying a cardioprotective role for human alpha1-ARs. In summary, these findings identify novel functional and mechanistic aspects of cardiac alpha1-AR function and suggest that activation of cardiac alpha1-AR might be a viable therapeutic strategy in heart failure.

I. Introduction

Adrenergic receptors (ARs) bind to and are activated by the endogenous catecholamine hormones epinephrine and norepinephrine (NE). Epinephrine is primarily produced in and released to the circulation from the adrenal gland, whereas NE is synthesized in and released by sympathetic nerve terminals in the peripheral nervous system and brain. In the heart, the two main ARs are the β-ARs, which comprise roughly 90% of the total cardiac ARs, and α1-ARs, which account for approximately 10% (see section II).

In general, acute activation of cardiac β1-ARs, the predominant β-AR subtype (80% or more of total β-ARs in heart), induces positive inotropic and chronotropic responses, although in heart failure, where sympathetic activation and catecholamine levels are increased, long-term activation of β1-ARs exacerbates pathologic remodeling (Bristow, 2000; Naga Prasad et al., 2001; Lohse et al., 2003).

Less is known about cardiac α1-ARs, but studies from the last thirty years indicate that long-term activation of cardiac α1-ARs activates beneficial trophic signaling in the developing heart and that these α1-AR-mediated trophic effects in the adult, in many ways, counteract the negative effects of overstimulation of β1-ARs in heart failure. This review will focus on these trophic effects of cardiac α1-ARs and how activation of α1-ARs might be beneficial in heart failure.

There are three α1-AR subtypes, the α1A, α1B, and α1D, and all three are expressed in the heart in a cell-type specific manner (section II). All three α1-ARs are G-protein-coupled receptors (GPCR), and classic α1-AR signaling mechanisms involve coupling to the Gq/11 (Gαq) family of G-proteins and activation of phospholipase Cβ1 (PLCβ1) at the plasma membrane. Activation of PLCβ1 cleaves phosphatidylinositol (PI), increasing inositol trisphosphate (IP3) and diacylglycerol (DAG). IP3 binds to the IP3-receptor to release calcium from intracellular stores, and DAG activates protein kinase C (PKC).

Other Gq-coupled GPCRs that signal through Gαq, such as endothelin receptors (ETRs) and angiotensin receptors (ATRs), are believed to play an important role in the pathogenesis of heart failure. Hallmarks of cardiomyopathy with heart failure include contractile dysfunction (both systolic and diastolic), myocyte hypertrophy, fibrosis, and increased cardiac cell death (Anand and Florea, 2003), which can all be worsened by Gq-coupled receptors (Salazar et al., 2007).

However, it also needs to be recalled that the view that Gq-coupled receptor signaling is toxic is based in large part on a transgenic mouse model with Gq overexpression that markedly exceeds the 2-fold increase found in human heart failure (Adams et al., 1998; Ponicke et al., 1998; Sakata et al., 1998) and thus cannot be considered to simulate human pathophysiology. In human heart failure, the maximal increase in Gq abundance is 2-fold (Ponicke et al., 1998), and transgenic mice with 2-fold cardiomyocyte-specific Gq overexpression have no discernible cardiac phenotype (Adams et al., 1998; Sakata et al., 1998).

Furthermore, α1-ARs differ from other Gq-coupled receptors in several important ways, including expression limited to myocytes within the heart (section II) and localization and signaling at the nucleus, as discussed in section III.

Thus, unlike what can be seen with some Gq-coupled receptors, α1-ARs protect the heart by activating an adaptive or physiologic hypertrophy, preventing cardiac myocyte death, augmenting contractile function in heart failure and inducing preconditioning (section IV). Finally, clinical trials indicate that blockade of α1-ARs exacerbates heart failure (section V), which could be explained by the cardioprotective functions of α1-ARs identified in cell and animal models. This review summarizes these data, which span decades, and emphasizes recent findings from our laboratories.

II. α1-Adrenergic Receptor Expression in the Heart

A. α1-Adrenergic Receptor Expression in the Heart in Animal Models

In mice and rats, all three α1-AR subtype mRNAs, α1A, α1B, and α1D, are detected in the heart (Rokosh et al., 1994; Stewart et al., 1994; Cavalli et al., 1997; O'Connell et al., 2003). Interestingly, among most species, including mouse, guinea pig, rabbit, pig, and cow, heart α1-AR levels determined by ligand binding are relatively constant (mouse: mean of six studies, ∼12 fmol/mg protein) (Steinfath et al., 1992a; Cavalli et al., 1997; Yang et al., 1998; Lin et al., 2001; O'Connell et al., 2003; Rokosh and Simpson, 2002), with the exception of rat heart, in which α1-AR levels are approximately 10-fold higher (rat: mean of four studies, ∼114 fmol/mg) (Steinfath et al., 1992a; Michel et al., 1994; Noguchi et al., 1995; Stewart et al., 1994).

Determination of cell-type specific expression of α1-ARs in the heart, or any tissue, is hampered by the lack of validated, subtype-specific α1-AR antibodies (Jensen et al., 2009c), which is a general problem with antibodies for GPCRs, as reviewed (Michel et al., 2009). However, studies in α1-AR knockout mice demonstrate that cardiac myocytes express only the α1A- and α1B-subtypes, based on lack of [3H]prazosin binding or functional responses in hearts from α1AB-double knockout mice (α1ABKO) (McCloskey et al., 2003; O'Connell et al., 2003; Turnbull et al., 2003) as well as lack of binding to a fluorescent α1-AR antagonist or signaling in cardiac myocytes isolated from α1ABKO hearts (O'Connell et al., 2003; Wright et al., 2008).

Ligand binding studies further indicate that the α1B is predominant, with the α1A- and α1B-subtypes expressed in a 1:2–4 ratio in cardiac myocytes (Rokosh and Simpson, 2002; O'Connell et al., 2003). Despite the presence of α1D-subtype mRNA, rodent cardiac myocytes do not appear to express the α1D-subtype protein by binding (O'Connell et al., 2003). However, the α1D might be expressed in the coronary vasculature, based on studies demonstrating α1-AR mediated reductions in coronary flow in isolated α1-AR knockout hearts (Chalothorn et al., 2003; Turnbull et al., 2003). This idea is supported by human studies (below). Conversely, rodent cardiac fibroblasts do not express α1-ARs (Stewart et al., 1994; O'Connell et al., 2001), and α1-agonist infusion induces hypertrophy without fibrosis (Marino et al., 1991), suggesting that α1-AR activation does not exacerbate fibrosis associated with heart failure. In contrast with α1-ARs, most ATRs and ETBRs are in fibroblasts, not cardiac myocytes (Kim et al., 1995; Gray et al., 1998; Modesti et al., 1999).

Long-term activation of α1-ARs and other hypertrophic agonists increases the α1A-subtype, without desensitizing α1-mediated inositol phosphate (IP) turnover or growth, while decreasing α1B-subtype mRNA and protein levels in cultured neonatal rat cardiac myocytes (NRVM) and in rats subjected to aortic banding (Rokosh et al., 1996). Moreover, total α1-AR levels are not altered in vivo by hypertrophy or heart failure in rats (Rokosh et al., 1996; Sjaastad et al., 2003), and α1-AR inotropic effects are maintained or increased (Wang et al., 2010), in contrast to β-ARs that are desensitized and downregulated in heart failure (Bristow et al., 1982; Bristow et al., 1988).

Partial explanation for the differences in desensitization of α1-AR and β-ARs might reside in expression and regulation of G-protein receptor kinases (GRKs). GRK3 is found exclusively in myocytes, regulates α1-ARs, and is not upregulated in heart failure (Vinge et al., 2001, 2007; Aguero et al., 2012). In contrast, GRK2 and GRK5 that desensitize β-ARs but not α1-ARs are expressed in many myocardial cell types and are upregulated in heart failure (Rockman et al., 1996; Eckhart et al., 2000; Vinge et al., 2001, 2007; Aguero et al., 2012).

B. Unique Aspects of α1-Adrenergic Receptor Expression Profiles in Cardiac Myocytes

Recent studies provide unique information on the expression and distribution of α1-ARs in cardiac myocytes. First, both the α1A- and α1B-subtypes localize to and signal at the nuclear membrane, but not the plasma membrane, in adult mouse cardiac myocytes (Huang et al., 2007; Wright et al., 2008; Wright et al., 2012), as reviewed in section III. Second, α1A-subtype expression and function are graded in adult cardiac myocytes, from high levels to none, whereas the α1B-subtype is expressed in all cardiac myocytes (unpublished data).

C. α1-Adrenergic Receptor Expression in Human Heart

In human heart, all three α1-AR subtype mRNAs are detected (Jensen et al., 2009a). Furthermore, α1-AR expression levels in human heart determined by ligand binding are similar to mouse and most other species (human: mean of 6 studies, ∼12 fmol/mg protein) (Bohm et al., 1988; Bristow et al., 1988; Vago et al., 1989; Steinfath et al., 1992b; Hwang et al., 1996; Jensen et al., 2009a). Human myocardium has the α1A- and α1B-subtypes, with the α1B predominant, similar to other species (Jensen et al., 2009a,b), and the α1A is functional in signaling (R. C. Thomas and P. C. Simpson, unpublished data). These data suggest that the mouse is a more appropriate model to approximate cardiac α1-AR function than the rat, which as mentioned above, has roughly 10-fold more α1-ARs.

Competition binding experiments do not detect the α1D-subtype in explanted human heart (Jensen et al., 2009a,b). However, the α1D-subtype is expressed and functional in coronary artery smooth muscle cells and might cause vasoconstriction (Jensen et al., 2009b). The α1B-subtype is expressed in coronary artery endothelial cells and might induce vasodilation and angiogenesis (Jensen et al., 2010).

D. α1-Adrenergic Receptor Levels Increase Proportionately in Human Heart Failure

In heart failure, β1-ARs are desensitized and downregulated. In contrast, radioligand-binding studies indicate that myocardial α1-AR levels are slightly increased in human heart failure (mean of six studies, increased from ∼12 to ∼19 fmol/mg protein) (Bohm et al., 1988; Bristow et al., 1988; Vago et al., 1989; Steinfath et al., 1992b; Hwang et al., 1996; Jensen et al., 2009a). This means that α1-AR levels in the heart, which normally represent approximately 11% of the total AR population at baseline (range 2–23%, mean of six studies), are proportionately increased to approximately 25% of the total AR population in heart failure (range 9–41%) (Bohm et al., 1988; Bristow et al., 1988; Vago et al., 1989; Steinfath et al., 1992b; Hwang et al., 1996; Jensen et al., 2009a). Given that sympathetic drive and catecholamine levels are increased in heart failure (Cohn et al., 1984), this could imply that α1-ARs sustain adrenergic function when β1-ARs are downregulated. In fact, α1-AR-induced positive inotropy, which at baseline is minimal, can be equal to β-AR-mediated inotropy in ventricular muscle strips isolated from human heart failure patients (Skomedal et al., 1997), as reviewed in more detail below.

E. Conclusions on α1-Adrenergic Receptor Heart Expression

In summary, α1-ARs constitute a minority of the total cardiac AR population in humans at baseline, and this seems to hold across species, with the exception of rats where α1-ARs levels are ∼10-fold higher than any other species. This should be considered when interpreting results from studies of α1-ARs in rats, particularly in cultured NRVMs, the most common cardiac myocyte culture model.

Cardiac myocytes of all species have all three α1-AR subtype mRNAs, but only the α1A- and α1B-subtype receptor proteins are detected. In humans, the α1D-subtype is present in coronary smooth muscle and might regulate coronary vasoconstriction, whereas the α1B-subtype is in coronary endothelial cells and might regulate vasodilation and angiogenesis. In contrast, α1-ARs are not expressed by cardiac fibroblasts.

In heart failure, α1-ARs are not downregulated as are β1-ARs and thus become a greater share (25%) of ARs in the heart. This increase in α1-ARs could suggest that α1-ARs have a compensatory or adaptive role in heart failure, as suggested by studies showing that α1-mediated inotropy can be similar to β-AR-mediated inotropy in heart failure (Skomedal et al., 1997). The idea that α1-ARs might have an adaptive and protective role in the heart is a central theme of this review and is discussed in following sections.

III. α1-AR Signaling in Cardiac Myocytes

The following sections review the conventional models of α1-AR localization and signaling at the plasma membrane, or “outside-in” signaling, and evidence for novel models, suggesting that α1-ARs and other GPCRs signal from the cardiac myocyte nucleus, or “inside-out” signaling.

A. Conventional Models of α1-Adrenergic Receptor Signaling

Conventional models of GPCR signaling describe receptor activation at the plasma membrane leading to initiation of downstream signaling within the cell, commonly referred to as “outside-in” signaling. Furthermore, classic models of GPCR function suggest that GPCRs are expressed on the cell membrane and are only internalized after receptor phosphorylation and subsequent desensitization (Drake et al., 2006). α1-ARs signal through the Gq/11 class of G-proteins, leading to activation of PLCβ1 and increases in IP3/calcium signaling and activation of PKC (Graham et al., 1996; Piascik and Perez, 2001). The α1B-subtype might also signal through Gi (Hu and Nattel, 1995; Steinberg et al., 1985; Akhter et al., 1997; Melien et al., 2000; Snabaitis et al., 2005). In NRVM, historically the primary cell model used to study cardiac α1-AR signaling, α1-AR-induced increases in IP3 are readily observed, but in adult cardiac myocytes this is controversial. The general consensus is that α1-ARs signal through the Gq/PLCβ1-IP3/PKC pathway, but downstream signaling pathways are diverse, as reviewed elsewhere (Hein and Michel, 2007; Cotecchia, 2010; Jensen et al., 2011). To date, over 70 downstream signaling molecules have been implicated in cardiac α1-AR signaling, using the NRVM model of α1-AR-stimulated cardiac myocyte hypertrophy (Jensen et al., 2011). Some data suggest interactions with β-arrestin (Pediani et al., 2005; Stanasila et al., 2008; Hennenberg et al., 2011) and Gβγ (Vettel et al., 2012).

B. New Model for General G-Protein-Coupled Receptor Signaling: G-Protein-Coupled Receptors at the Nucleus

It is now clear that several GPCRs localize to and signal at the nucleus, or “inside-out” signaling. Nuclear signaling is seen in several cell types, including neurons, hepatocytes, and cardiac myocytes, as reviewed previously (Gobeil et al., 2006; Boivin et al., 2008; Bkaily et al., 2009; Tadevosyan et al., 2012). The GPCRs include receptors for prostaglandin E2 in the brain (Gobeil et al., 2002), angiotensin II (AT1R) in the brain and in HEK and Chinese hamster ovary cells (Lu et al., 1998; Chen et al., 2000; Lee et al., 2004), platelet activating factor in the liver and brain (Marrache et al., 2002), apelin in the brain (Lee et al., 2004), bradykinin in HEK cells (Lee et al., 2004), and glutamate in neurons (O'Malley et al., 2003).

Several recent studies show that GPCRs localize to nuclei in binucleate adult cardiac myocytes, as reviewed previously (Tadevosyan et al., 2012). Specifically, ETRs are detected on nuclei isolated from adult cardiac myocytes, and endothelin stimulates nuclear calcium transients (Boivin et al., 2003). ATRs and β-ARs are also detected on nuclei isolated from adult cardiac myocytes and mediate increased RNA synthesis (Boivin et al., 2006; Tadevosyan et al., 2010; Vaniotis et al., 2011). These findings indicate that GPCR localization to the nucleus could regulate important physiologic functions in adult cardiac myocytes. However, the majority of these other receptors can localize also to the myocyte plasma membrane; for example, 95% of ETRs are on the sarcolemma (Boivin et al., 2003; Wright et al., 2012), so that the relative functional significance of nuclear versus surface localization is uncertain.

Despite the data reviewed above, the prevalent view is that GPCRs, including α1-ARs, are localized primarily to the plasma membrane in heart and myocytes. This impression is based predominantly on radioligand binding to membrane fractions, binding assays in whole cells (Filipeanu et al., 2006) and studies with α1-AR antibodies. Difficulties with these approaches are discussed in the next section.

C. α1-Adrenergic Receptors in the Nuclei in Cardiac Myocytes

Cellular localization of signaling molecules determines function, emphasizing the importance of α1-AR subcellular localization in cardiac myocytes. The following sections review the limitations and advantages of different approaches to detect α1-AR subcellular localization and the evidence that α1-ARs are in the cardiac myocyte nucleus, derived from studies of localization, agonist uptake, and signaling. Physiologic implications of nuclear α1-ARs are also suggested. This novel nuclear α1-AR signaling paradigm in cardiac myocytes is illustrated in Fig. 1.

Fig. 1.

Fig. 1.

Model for α1-AR signaling at the nuclear membrane. In adult cardiac myocytes, catecholamine α1-AR agonists (NE/PE) are actively transported into the myocyte via organic cation transporter 3 (OCT), which can be inhibited by corticosterone. The membrane-permeable α1-AR antagonist prazosin (and similar derivatives) can cross the plasma membrane to inhibit signaling, whereas the membrane impermeable α1-AR antagonist CGP12177A fails to inhibit signaling. The model suggests that active α1-ARs localize to the inner nuclear membrane with the ligand-binding domain facing the space between the outer and inner nuclear membranes (ONM and INM, respectively). On the basis of this orientation, binding of agonist to α1-ARs induces signaling inside the nucleus, possibly through Gαq, although downstream intranuclear signaling pathways remain to be defined. We propose that activation of nuclear α1-ARs can induce intranuclear hypertrophic signaling as well as extranuclear signaling, including activation of ERK in caveolae and survival signaling or phosphorylation of cardiac troponin I at the sarcomere and contractile function. HDAC, histone deacetylase; Ca Ch, calcium channel; RYR, ryanodine receptor; PTP, mitochondrial permeability transition pore; ER/SR, endoplasmic/ sarcoplasmic reticulum; NR, nucleoplasmic reticulum; NPC, nuclear pore complex.

1. Nuclear Localization of α1-Adrenergic Receptors in Cardiac Myocytes.

Limitations with the techniques used to detect α1-ARs, radioligand binding and α1-AR antibodies, might explain the conventional view that α1-ARs localize mainly to the plasma membrane. Ligand binding assays typically involve homogenization of heart tissue or cultured cells followed by a high-speed ultracentrifugation to isolate total membrane fractions. This high-speed ultracentrifugation pulls down all membranes, and if subcellular markers are not used, this technique does not distinguish between plasma, sarcoplasmic, and nuclear membranes (Lin et al., 2001; Rokosh and Simpson, 2002; O'Connell et al., 2003). Furthermore, most purified membrane preparations exclude over 65 to 85% of total heart α1-ARs that are found in “debris” and low-speed pellets discarded normally (Simpson, 2006). Whole-cell binding assays are limited by the lack of radioligands that do not enter the cell (Filipeanu et al., 2006).

Immunochemical detection, either by immunoblot or cell/tissue staining, is another commonly used technique to detect α1-ARs. However, none of 10 commercial α1-AR antibodies are specific for α1-ARs in general or for any subtype, as documented by the fact that no antibody detects a band in wild-type (WT) tissue that is absent in tissue from α1-AR knockout (KO) mice (Jensen et al., 2009c). This nonspecificity of anti-GPCR antibodies is a general problem, reviewed recently, emphasizing that α1-AR antibodies need to be validated using KO tissue (Michel et al., 2009). Nonspecificity of α1-AR antibodies calls into question previous reports with these reagents, for example, work suggesting α1-AR localization to the plasma membrane and t-tubules in adult rat cardiac myocytes (O-Uchi et al., 2008) or a study using immunoprecipitation of α1-ARs with potential signaling partners (Fujita et al., 2001).

An antibody to the 1D4 epitope tag at the C terminus of the α1A detects surface membrane expression in heart sections of a transgenic mouse (Lin et al., 2001). However, it is problematic whether receptor localization with 170-fold overexpression simulates that of endogenous α1-ARs (Lin et al., 2001).

A few studies use membrane fractionation combined with the caveolar marker caveolin-3 (cav-3) to detect α1-AR binding in caveolae in NRVMs (Fujita et al., 2001; Lanzafame et al., 2006). In NRVMs, a caveolar fraction defined in this way contains most or all α1-mediated IP turnover (Morris et al., 2006) and 27% of total α1-AR binding, both α1A and α1B (Lanzafame et al., 2006). The value of 27% α1-AR binding in caveolae in NRVMs agrees well with a more recent study finding 20% of total α1-ARs in adult myocyte membranes defined by high levels of cav-3 (Wright et al., 2008).

The contrary notion that α1-ARs localize primarily to the nucleus arises from three main lines of evidence. First, 80% of total α1-AR binding in adult mouse cardiac myocytes is found in nuclear membranes defined by the marker LAP2 (Wright et al., 2008, 2012). In NRVMs, nuclear α1-AR binding is also observed (Buu et al., 1993), and 73% of total α1-ARs are in noncaveolar membranes that might be nuclear (Lanzafame et al., 2006), in good agreement with the results in adult myocytes.

Second, BODIPY-prazosin is a fluorescent analog of the α1-AR antagonist prazosin that binds all three α1-AR subtypes with equal affinity and fluoresces only when bound to receptor (Daly et al., 1998; Mackenzie et al., 2000; Pediani et al., 2005). BODIPY-prazosin staining of living adult cardiac myocytes identifies endogenous α1-ARs on the nuclear membrane but does not detect receptors at the plasma membrane (Wright et al., 2008). Nuclei isolated from adult cardiac myocytes confirm positive BODIPY-prazosin staining of endogenous nuclear α1-ARs (Wright et al., 2012).

Third, a reconstitution system, in which α1-AR-GFP fluorescent fusion proteins are expressed in cultured adult α1ABKO cardiac myocytes, recapitulates the nuclear localization of the endogenous α1-ARs (Huang et al., 2007; Wright et al., 2008, 2012).

Studies in other cells provide some support for the results in cardiac myocytes. In recombinant cells expressing α1-ARs, for example, HEK293 cells, all α1-AR subtypes show some intracellular localization (Daly et al., 1998; Mackenzie et al., 2000; Chalothorn et al., 2002). In primary cultures of smooth muscle cells, endogenous α1-ARs are also found on both the plasma membrane and intracellular, using a fluorescent ligand, BODIPY-FL prazosin (Mackenzie et al., 2000).

2. Mechanism of α1-Adrenergic Receptor Nuclear Localization.

The mechanism for nuclear targeting involves nuclear localization sequences embedded in the protein. These nuclear localization sequences typically consist of mono- or bi-partite basic residues, usually lysines and arginines or glycine-arginine repeats (Dono et al., 1998; Hock et al., 1998; Lu et al., 1998). Nuclear localization sequences are recognized by a class of proteins known as importins that bind these sequences and facilitate transport of the target protein to the nucleus. This importin-mediated nuclear localization not only occurs for proteins that target to the nucleoplasm but for proteins that target the inner nuclear membrane as well (King et al., 2006; Cook et al., 2007; Lusk et al., 2007). Importin-mediated nuclear localization was previously described for the type 1 parathyroid hormone receptor (Pickard et al., 2006, 2007) and more recently for the gonadotropin-releasing hormone type 1 receptor (Re et al., 2010). Recent experiments identify nuclear localization sequences in the α1A- and α1B-subtypes, and mutation of these sequences results in loss of nuclear localization for each subtype in adult mouse cardiac myocytes (Wright et al., 2012).

3. Receptor Orientation in the Inner Nuclear Membrane.

As described above, nuclear membrane proteins are targeted to the inner nuclear membrane through nuclear localization sequences, similar to proteins in the nucleoplasm. Also important is the orientation of inner nuclear membrane proteins, which could affect how they signal. For GPCRs, such as α1-ARs, if the ligand-binding domain faces the inside of the nucleus, the ligand would have to enter the nucleus and signaling would be initiated on the cytoplasmic side in the space between the inner and outer nuclear membranes. Conversely, if the ligand-binding domain faces the space between the outer and inner nuclear membranes, then signaling would be activated inside the nucleus.

Recent studies with nuclear GPCRs detect signaling in isolated nuclei, implying that nuclear receptors are likely oriented with the ligand-binding domain facing outward and the C terminus facing the nucleoplasm. ETRs induce calcium transients in isolated nuclei (Boivin et al., 2003), and β-ARs and ATRs induce transcriptional responses in isolated nuclei (Tadevosyan et al., 2010; Vaniotis et al., 2011). These studies suggest that nuclear GPCR signaling is activated inside the nucleus, thereby indicating an orientation in the inner nuclear membrane similar to GPCRs at the plasma membrane where the C terminus faces the cytoplasm.

4. Catecholamine Uptake in Cardiac Myocytes.

A prerequisite for nuclear α1-AR signaling is that NE and other α1-AR ligands must traverse the plasma membrane, transit to the nucleus, and bind to and activate receptors in a time course consistent with signaling. In nonneuronal cells, this process is known as NE “uptake-2” (Obst et al., 1996) and is facilitated by extraneuronal monoamine transporter/organic cation transporter 3 (EMT/OCT3) (Zwart et al., 2001; Schomig et al., 2006). EMT/OCT3 is expressed most abundantly in heart (Zwart et al., 2001), where it is present on both the plasma and nuclear membranes in adult cardiac myocytes (Wright et al., 2008). In neonatal myocytes, uptake of [3H]NE is observed, but the time scale of nearly an hour before NE is detected in the nucleus is not sufficiently rapid to account for α1-AR signaling (Buu et al., 1993).

However, a more sensitive fluorescent-based catecholamine uptake assay shows that catecholamines are taken up very rapidly in cultured adult mouse cardiac myocytes. In this system, catecholamine uptake begins within seconds, is clearly increased by 5 minutes, peaks at 30 minutes, and is antagonized by addition of unlabeled NE 15 minutes prior to catecholamine uptake measurement, indicating specificity (Wright et al., 2008).

Further consistent with rapid uptake, the intrinsic uptake kinetics of OCT3, which is the rate at which one transporter moves a cation, show that OCT3-mediated cation transport is in the time frame of seconds. Thus, the uptake kinetics of catecholamines by recombinant OCT3 expressed in HEK293 cells is a Vmax ∼30,000 pmol/mg protein/min and a Km ∼900 μM for NE, and a Vmax ∼13,000 pmol/mg protein/min and a Km ∼500 μM for epinephrine (Duan and Wang, 2010). Catecholamine uptake is observed in seconds, with half-maximum response seen in ∼2 minute (Duan and Wang, 2010). It is likely that the kinetic properties of the transporter are relatively consistent from cell to cell and that expression level will dictate the absolute amount of uptake and OCT3 expression is highest in heart (Zwart et al., 2001).

In mice, OCT3-mediated heart uptake of the neurotoxin cation methyl-4-phenylpyridinium acetate is observed within minutes of infusion (∼4000 ng/g tissue 5 minutes after infusion), and this uptake is inhibited by 75% in OCT3KO mice (Zwart et al., 2001), indicating a rapid and robust uptake system. The phenotype of OCT3KO mice is further interesting. Thus, OCT3KO mice have a trend toward reduced heart size in males [WT heart weight (HW) 160 mg, OCT3KO HW 145 mg, n = 7, P = 0.138, a 10% reduction] (Zwart et al., 2001), reminiscent of the small heart phenotype seen in male α1ABKO mice (WT HW 147 mg, α1ABKO 122 mg, n = 33–27, P < 0.05, a 17% reduction) (O'Connell et al., 2003), but the number of OCT3KO mice analyzed was small (n = 7) (Zwart et al., 2001).

Thus, the kinetics of catecholamine uptake in myocytes (Wright et al., 2008) and the biochemistry and biology of OCT3 (Zwart et al., 2001; Duan and Wang, 2010) are consistent with α1-AR responses initiated by agonist activation of nuclear receptors.

In agreement with the idea that α1-AR agonist must be transported into the myocyte for signaling, there is a long latency of α1-AR responses after agonist addition, in contrast to the rapid onset for β-AR agonism. Specifically, the latency for contractile or calcium responses to α1-agonism in isolated myocytes is 2 to 5 minutes in nine studies (Tohse et al., 1990; Terzic et al., 1992; Gambassi et al., 1998; Zhang et al., 1998; Woo and Lee, 1999; Ross et al., 2003; O-Uchi et al., 2005; Luo et al., 2007; Ichishima et al., 2010), rather than seconds as would be expected for a receptor at the sarcolemma.

Finally, there is functional evidence that agonist uptake is required for α1-AR signaling. Inhibition of EMT/OCT3-mediated catecholamine uptake with corticosterone, an EMT/OCT3 antagonist, prevents α1-AR activation of ERK in cultured adult mouse cardiac myocytes (Wright et al., 2008).

In summary, the kinetics of agonist uptake in myocytes, EMT/OCT3 biochemistry and biology, including kinetics, heart expression, and inhibition by corticosterone, and the latency of α1-AR physiologic responses are all consistent with agonist uptake and activation of nuclear α1-ARs.

5. Functional Evidence for Nuclear α1-Adrenergic Receptor Signaling.

α1-ARs have numerous signaling effects in the cytosol, as reviewed later. Thus, if α1-ARs signal in the nucleus, then that signal must be transduced out of the nucleus to reach these cytosolic targets, defining an inside-out (nuclear-to-cytoplasmic) signaling mechanism. Three sets of data support the idea that α1-signaling is initiated in the nucleus. First, CGP-12177A [4-[3-[(1,1-dimethylethyl)amino]2-hydroxypropoxy]-1,3-dihydro-2H-benzimidazol-2-one hydrochloride], an α1-antagonist that does not cross membranes (Staehelin et al., 1983; Levin et al., 2002; Brahmadevara et al., 2003, 2004), does not block α1-AR-ERK signaling in cultured adult mouse cardiac myocytes, whereas the prototypical α1-AR antagonist prazosin, which freely crosses the plasma membrane, does block α1-AR-ERK signaling (Wright et al., 2008). Second, mislocalization mutants of both the α1A- and α1B-subtype, in which the nuclear localization sequences are mutated, do not activate ERK in cultured adult mouse cardiac myocytes (Wright et al., 2012). These mislocalization mutants are not redirected to the plasma membrane, which would provide a more crucial test of the requirement for nuclear localization, but the mutants do show that nuclear localization is required for α1-AR signaling in adult cardiac myocytes. Finally, activation of nuclear α1-ARs leads to the activation of ERK in caveolae at the plasma membrane (Wright et al., 2008), and the nuclear export inhibitor leptomycin B blocks α1-AR-mediated activation of ERK, suggesting that α1-AR signaling to ERK at caveolae must originate in the nucleus (Wright et al., 2012). How signals are transported from the nucleus to cytosolic targets is uncertain. However, α1-ARs activate PKC, a molecule known to translocate upon activation, suggesting a possible mechanism to transmit a signal out of the nucleus. Taken together, these studies provide functional evidence for α1-AR signaling initiated in the nucleus.

6. Localization of Signaling Partners with α1-Adrenergic Receptors in Cardiac Myocyte Nuclei.

To effect nuclear α1-AR signaling in cardiac myocytes, α1-ARs must colocalize with downstream signaling partners in the nuclear membrane. However, their identities so far remain unclear.

A fraction of Gαq colocalizes with α1-ARs at the nucleus, based on immunocytochemistry in α1ABKO cardiac myocytes expressing α1-AR-GFP fluorescent fusion proteins, and on subcellular fractionation of WT adult cardiac myocytes (Wright et al., 2008). In NRVMs, approximately 56% of Gαq is in caveolar membranes defined by cav-3, and the remainder is in noncaveolar membranes, some of which might be nuclear (Morris et al., 2006).

A role in nuclear signaling is uncertain for PLCβ1, the classic α1-coupled PLC. PLCβ1 is detected in the nuclei of adult cardiac myocytes using the G12 PLCβ1 antibody from Santa Cruz Biotechnology (Santa Cruz, CA) (Wright et al., 2008), but this and other commercial PLCβ1 antibodies are not proven specific for PLCβ1 using KO tissues. In NRVMs, 91% of PLCβ1 is detected in a caveolar fraction using a Santa Cruz antibody (Lanzafame et al., 2006), and forced expression of PLCβ1b with an N-terminal enhanced GFP tag detects localization on the sarcolemma but not the nucleus (Grubb et al., 2008). Also in NRVMs, expression of a C-terminal peptide from PLCβ1b blocks α1- and Gq-mediated IP turnover and aspects of hypertrophy (Grubb et al., 2008; Filtz et al., 2009). Finally, the substrate for PLCβ1, phosphatidylinositol 4,5-bisphosphate, is not detected in nuclear membranes (Zhang et al., 2013). Taken together, these data suggest that PLCβ1 might not be involved in α1-AR nuclear signaling.

An interesting alternate mediator of nuclear α1-AR signaling is PLCε, which might be regulated by small GTPases (Rho, Ras, Rap) and Gβγ subunits, but not by Gαq (Lopez et al., 2001). PLCε in NRVMs and heart is scaffolded to muscle-specific A kinase-anchoring protein at the nuclear envelope with PKD, one key hypertrophic signaling molecule (Zhang et al., 2011, 2013). Knockdown of PLCε inhibits α1-and Gαq-stimulated hypertrophy in NRVMs, but has no effect on IP turnover, and expression of PLCε causes hypertrophy, an effect that requires PLCε catalytic activity (Zhang et al., 2011, 2013). Myocyte-specific PLCε KO also inhibits hypertrophy with pressure overload in vivo (Zhang et al., 2013). Extensive evidence suggests that PLCε mediates hypertrophy by hydrolysis of phosphatidylinositol 4-phosphate (PI4P) at the nuclear envelope, with generation of DAG and activation of PKD, but upstream mechanisms are uncertain (Zhang et al., 2013). Clearly, definition of nuclear α1-AR signaling partners is a promising area for the future.

7. Nuclear α1-Adrenergic Receptor Localization in Vivo.

It is possible that localization observed in isolated or cultured cardiac myocytes does not reflect the true localization of α1-ARs in vivo. As mentioned, overexpressed α1A-ARs in transgenic mice localize to the plasma membrane based on immunohistochemical staining for an epitope tag (Lin et al., 2001). However, very high receptor levels, about 170-fold over basal, might cause artifactual localization, and the lack of validated α1-AR antibodies (Jensen et al., 2009c) make conventional immunohistochemical approaches problematic.

Conversely, in a different α1A-subtype transgenic model, in which an α1A-subtype GFP fusion protein is expressed at a much lower level, approximately 5-fold over basal, α1-ARs are detected at the nuclei in ventricular tissue sections with a GFP antibody (Wright et al., 2008). This result with the α1A-GFP transgenic mice suggests that α1-AR nuclear localization observed in cultured cardiac myocytes can represent α1-AR localization in vivo.

8. Pathophysiologic Implications of Nuclear α1-Adrenergic Receptor Signaling.

ETRs, ATRs, and β-ARs signal in isolated nuclei from adult cardiac myocytes (Boivin et al., 2003; Tadevosyan et al., 2010; Vaniotis et al., 2011). However, it is difficult to assign a functional significance to nuclear signaling by these GPCRs in cardiac myocytes, because the majority of ETRs, ATRs, and β-ARs localize to the plasma membrane (although quantitative ligand binding in subcellular fractions for ATRs is not possible due to low level of expression). Conversely, approximately 80% of α1-ARs localize to the nuclei in adult mouse cardiac myocytes. Interestingly, in pathologic settings, α1-AR signaling is clearly protective (sections IV and V), whereas ETR and ATR signaling can exacerbate pathologic remodeling (Harada et al., 1999; Yang et al., 2004). This raises the possibility that differences in receptor localization might lead to differences between physiologic and pathologic signaling. In other words, nuclear receptors, like α1-ARs, might be protective, whereas ETRs and ATRs at the plasma membrane might induce pathologic signaling (Wright et al., 2012). Although these ideas remain to be tested, differential localization of Gq-coupled receptors could have significant implications for their physiologic functions and for therapeutic targeting of Gq-coupled receptors in heart disease.

9. Summary of α1-Adrenergic Receptor Nuclear Localization.

Overall, the majority of current data supports the idea that α1-ARs localize to and signal from the nuclei in adult cardiac myocytes in vitro and in vivo. Identification of functional nuclear localization sequences in each α1-subtype provides a mechanistic basis to support nuclear α1-AR localization, oriented with the C-terminal tail in the nucleoplasm. Ligand uptake into the cell via EMT/OCT3 provides a mechanism for receptor activation. The signaling mechanisms of nuclear α1-ARs remain unclear, as do the physiologic implications of nuclear versus sarcolemmal signaling by α1-ARs and other Gq-coupled receptors.

IV. α1-Adrenergic Receptor Physiologic Function in the Heart

In the heart, AR physiology is largely focused on acute β-AR mediated regulation of contractile function, whereas chronic β-AR signaling is maladaptive and β-AR antagonists are now standard therapy in heart failure. Short-term α1-AR signaling can increase contractility, as reviewed below, but this has not been studied in detail in vivo. On the other hand, many studies now indicate that chronic α1-AR signaling is adaptive, protecting the heart from pathologic stress through activation of physiologic hypertrophy, survival signaling, augmentation of contractility, and ischemic preconditioning. These data are described below.

A. α1-Adrenergic Receptors Activate Physiologic or Adaptive Hypertrophy

Cardiac myocyte hypertrophy is the most common cellular response in the heart to pathologic stress, but hypertrophy is not always maladaptive (Frey and Olson, 2003). Cardiac hypertrophy occurs during normal physiologic development and in response to exercise and also as an adaptive response to pathologic stress. Physiologic or adaptive hypertrophy is characterized by an increase in heart and cardiac myocyte size without fibrosis and an overall improvement in function.

In contrast, pathologic or maladaptive hypertrophy is characterized by an increase in heart and cardiac myocyte size accompanied by combinations of cardiac cell death, fibrosis, vessel loss, reduced innervation, and, most importantly, declining function. Clinically, cardiac hypertrophy in Framingham adults is correlated with a significantly increased risk of heart failure and sudden death (Levy et al., 1990). Thirty years of research from cell culture to genetically modified mice, summarized below, demonstrates clearly that α1-ARs can mediate a physiologic or adaptive form of cardiac hypertrophy that offsets pathologic remodeling in heart failure.

1. α1-Adrenergic Receptor-Mediated Hypertrophy in Cell Culture Models.

Primary cultures of NRVMs are the most common cell culture model used to examine signaling in cardiac myocytes. The original and now classic experiments (Glembotski, 2013) demonstrated that catecholamines acting through α1-ARs produce a direct trophic response in NRVMs (Simpson et al., 1982; Simpson, 1983, 1985). At the time, there was debate as to whether catecholamines induced hypertrophy through increasing blood pressure or through a direct action on cardiac myocytes, that is, whether myocyte hypertrophy was regulated in some way only by “load” or whether growth factors and their receptors were involved, as in other types of cells. These papers were the first demonstration that catecholamines induce cardiac myocyte hypertrophy directly. This finding was later confirmed in cultured adult rat and cat cardiac myocytes (Simpson, 1988; Fuller et al., 1990; Ikeda et al., 1991; Volz et al., 1991; Clark et al., 1993).

Cardiac hypertrophy is clearly linked to induction of gene transcription, and in NRVMs, α1-ARs induce a pattern of hypertrophic gene transcription characterized by re-expression of genes normally expressed only in the fetal heart (Simpson et al., 1989). Early studies identified a group of these “fetal genes” induced by α1-ARs, including c-myc (Starksen et al., 1986), atrial natriuretic factor (Knowlton et al., 1991, 1993), α-skeletal actin (αSkAct) (Bishopric et al., 1987; Long et al., 1989; Karns et al., 1995), and β-myosin heavy chain (βMyHC) (Waspe et al., 1990; Kariya et al., 1993, 1994).

A crucial study of endogenous transcription in intact NRVMs proved that α1-ARs stimulate transcription not only of a fetal gene (αSkAct), but also all RNA species, including the mRNA for an adult gene (cardiac actin), and total RNA (ribosomal and transfer) (Long et al., 1989). Several subsequent studies focusing on α1-AR-mediated transcriptional regulation delineated a host of transcriptional factors and modifiers activated by α1-ARs in cardiac myocytes, including TEF-1 (Kariya et al., 1993, 1994; Karns et al., 1995; McLean et al., 2003), GATA-4 (Morimoto et al., 2000; Liang et al., 2001a,b), Egr-1 (Jin et al., 2000), Elk 1 (McWhinney et al., 2000), Vgl-4 (Chen et al., 2004), Rlf (Post et al., 2002), CREB (Markou et al., 2004), Zfp260 (Debrus et al., 2005), and class 2 histone deacetylase (Vega et al., 2004; Liu et al., 2009).

Mechanistically, the α1A-subtype is implicated in α1-AR mediated hypertrophy in NRVMs through the use of α1-AR subtype-specific pharmacologic agents (Autelitano and Woodcock, 1998). Several mechanisms for α1-AR-mediated hypertrophic signaling are proposed, and a multitude of signal transducers are implicated (Jensen et al., 2011). Certain molecules, based on frequency in the literature, might be considered essential or “core” molecules required for α1-AR-mediated hypertrophic signaling, including PLC (Filtz et al., 2009; Zhang et al., 2013), PKC (α, δ, and ε, three main isotypes activated by α1-ARs) (Henrich and Simpson, 1988; Kariya et al., 1991, 1993, 1994; Karns et al., 1995; Haworth et al., 2000; Rohde et al., 2000; Braz et al., 2002, 2004; Vega et al., 2004; Carnegie et al., 2008), PKD (Haworth et al., 2000; Vega et al., 2004; Harrison et al., 2006; Avkiran et al., 2008; Bossuyt et al., 2008, 2011; Carnegie et al., 2008; Liu et al., 2009), ERK (Bueno et al., 2000; Xiao et al., 2001; Barron et al., 2003; O'Connell et al., 2003), and class 2 histone deacetylase (Vega et al., 2004; Backs et al., 2006, 2008; Harrison et al., 2006; Liu et al., 2009). Evidence also exists that transactivation of the EGFR is involved in α1-mediated hypertrophy (Morris et al., 2004; Guo et al., 2009; Li et al., 2011; Papay et al., 2013).

In total, α1-mediated hypertrophy in NRVMs is characterized by α1A-subtype-mediated activation of the “fetal gene program” along with general increases in transcription of all RNA species and protein synthesis (Simpson, 1985; Long et al., 1989). Because the fetal gene program is often associated with pathologic hypertrophy, it was believed originally that α1-ARs induce a pathologic hypertrophy, similar to high levels of Gq overexpression (Dorn and Brown, 1999). This idea proved to be incorrect.

2. α1-Adrenergic Receptor-Mediated Hypertrophy in Animal Models.

Early studies in mice, cats, and dogs showed that long-term catecholamine infusion in various species and at doses that do not increase blood pressure produces cardiac hypertrophy in vivo, which is “physiological,” in that cardiac function is normal or improved and there is no fibrosis (Laks et al., 1973; King et al., 1987; Marino et al., 1991; Patel et al., 1991; Stewart et al., 1992; Vecchione et al., 2002). Whereas these studies suggest clearly that activation of ARs induces cardiac hypertrophy directly, which was debated at the time, the lack of subtype-specific AR pharmacologic agents limits mechanistic insight. However, in preliminary experiments, infusion of a subpressor dose of an α1A agonist in mice can increase fetal gene expression (unpublished data).

The advent of transgenic mouse technology provided a platform to address AR subtype-specific function in vivo, and transgenic gain-of function and gene deletion loss-of-function models have mostly confirmed cell culture studies indicating that α1-ARs regulate hypertrophy, with some prominent exceptions.

Cardiac myocyte-specific transgenic overexpression of the WT α1A-subtype, even at very high levels (148- to 170-fold), does not alter heart size, although α1A transgenic mice eventually develop dilated cardiomyopathy and die prematurely (Lin et al., 2001; Chaulet et al., 2006). On the other hand, transgenic overexpression of constitutively active mutant (CAM) α1A with the endogenous α1A-promoter induces cardiac hypertrophy without an effect on systemic blood pressure (Papay et al., 2013).

Similarly, the α1B-subtype shows a variable ability to induce hypertrophy when overexpressed, depending on the model. Overexpression of a CAM of the α1B-subtype with the α-myosin heavy chain (αMyHC) promoter at low levels (2- to 3-fold) induces hypertrophy (Milano et al., 1994) and exacerbates pathologic remodeling after aortic constriction (Wang et al., 2000). Likewise, systemic overexpression of a CAM α1B with the endogenous α1B-promoter also induces cardiac hypertrophy, along with hypotension, and a decreased pressor response, clearly dissociating hypertrophy from blood pressure (Zuscik et al., 2001). In the same study, overexpression of a wild-type (WT) α1B with the endogenous α1B-promoter shows a lesser degree of hypertrophy (Zuscik et al., 2001). More recently, a different result was found in that overexpression of the CAM α1B caused hypertrophy, but only in older mice, and hypertrophy was associated with fibrosis (Papay et al., 2013). Furthermore, hypertrophy seen in both the CAM α1A and α1B mice was not observed when the mice were interbred to derive a systemic CAM α1AB transgenic mouse (Papay et al., 2013). In contrast with these results, relatively high-level overexpression of the WT α1B with the αMyHC promoter (>40-fold) does not induce hypertrophy but results in dilated cardiomyopathy and death (Akhter et al., 1997; Grupp et al., 1998; Iaccarino et al., 2001; Lemire et al., 2001).

α1-AR gene-deletion models are reviewed in detail (Simpson, 2006). In brief, heart size is not different in the α1A-knockout on a mixed FVB/129SvJ background (α1AKO) (Rokosh and Simpson, 2002) or in the α1B-knockout on a mixed C57Bl/6/129SvJ background (α1BKO) (Cavalli et al., 1997). Knockout of the α1D-subtype, which is not expressed in cardiac myocytes, also has no effect on heart size (Tanoue et al., 2002; Chalothorn et al., 2003; Hosoda et al., 2005).

However, double knockout of both the α1A- and α1B-subtypes, which eliminates α1-AR binding in the heart, on a congenic C57Bl/6J background (α1ABKO) causes a 15% reduction in heart and cardiac myocyte size during normal postnatal development (O'Connell et al., 2003). Mechanistically, ERK activity is reduced 30% in α1ABKO hearts, and α1-mediated activation of ERK is absent in cultured α1ABKO cardiac myocytes, suggesting that α1-AR-ERK signaling might regulate hypertrophic growth during postnatal development (O'Connell et al., 2003). Importantly, α1ABKO mice have normal basal blood pressure, normal body and organ weights, normal home cage locomotor activity, and normal overall health (O'Connell et al., 2003, 2006). These data show that any effects of the α1A- and α1B-subtypes on maintenance of basal blood pressure can be compensated by other receptors, but that heart growth requires the α1A and/or α1B. Maximal contractile responses to phenylephrine in α1ABKO isolated arteries are reduced by 35% in carotid and by 77% in mesenteric, with little or no changes in pEC50, indicating compensation by α1D (Methven et al., 2009). We have not tested α1-mediated pressor response in the intact α1ABKO, but they are reduced in both the α1AKO and α1BKO (Cavalli et al., 1997; Rokosh and Simpson, 2002).

Interestingly, a preliminary re-examination of the α1-AR single knockouts on a congenic C57Bl/6J background reveals that α1BKO mice have small hearts, similar to the α1ABKO, suggesting that the α1B-subtype alone is required for physiologic postnatal growth of the heart (unpublished data). In support of this, subpressor catecholamine infusion does not cause hypertrophy in an α1BKO on a mixed genetic background (Vecchione et al., 2002).

Aortic constriction in the α1ABKO mice results in a worse dilated cardiomyopathy, with fibrosis, apoptosis, decreased contractility, and increased mortality contrasted to WT mice (O'Connell et al., 2006). Interestingly, the final degree of hypertrophy in α1ABKO hearts after aortic constriction is similar to that in WT hearts, but induction of the fetal gene program is lost (O'Connell et al., 2006). This shows that the absence of α1-ARs exacerbates pathologic hypertrophic responses and that induction of the fetal-gene program can be uncoupled from pathologic hypertrophy.

3. Summary of α1-Adrenergic Receptor in Hypertrophy.

Early studies in NRVMs demonstrated that the α1A-subtype induces hypertrophy with activation of the fetal-gene program and overall RNA and protein synthesis. In vivo, infusions of subpressor doses of catecholamines cause a physiologic hypertrophy but generally do not pinpoint which α1-subtype might be responsible. Although studies from α1-AR transgenic mice provide inconsistent results, studies from α1-AR knockout mice suggest that the α1B-subtype is required for hypertrophic growth during postnatal cardiac development, a period of physiologic heart growth, and that α1-ARs are not required for pathologic hypertrophy after aortic constriction. In fact, pathologic hypertrophy is worse in the absence of α1-ARs.

Therefore, in vivo studies, either with catecholamine infusion or α1-AR knockout mice, collectively suggest that α1-ARs stimulate an adaptive or physiologic hypertrophy, with no decrease in contractile function. Interestingly, findings from NRVMs suggesting that α1-ARs induce pathologic hypertrophy based on activation of the fetal gene program are not supported by findings in α1ABKO mice. In α1ABKO mice, activation of the fetal gene program is absent despite significant hypertrophy and worse cardiomyopathy after aortic constriction.

A new consideration with regard to the fetal gene program is that a classic fetal gene, β-MyHC, is re-expressed only in a minor subpopulation of cardiac myocytes in the mouse heart after aortic constriction, and the myocytes with β-MyHC are smaller than the cells with α-MyHC, not larger (Lopez et al., 2011). These data question whether fetal genes are even markers of hypertrophy or pathology (Lopez et al., 2011).

B. α1-Adrenergic Receptors Prevent Cardiac Myocyte Death

Cell death, either apoptotic, necrotic, or autophagic, plays a significant role in the development of heart failure (Guerra et al., 1999; Kostin et al., 2003; Wencker et al., 2003; Baines et al., 2005; Foo et al., 2005; Nakayama et al., 2007; Nishida and Otsu, 2008; Baines, 2010; Whelan et al., 2010; Nemchenko et al., 2011). Whereas substantial evidence indicates that β1-ARs induce cell death, a growing body of research summarized below indicates that α1-ARs prevent cardiac myocyte cell death in direct opposition to β1-ARs.

1. α1-Adrenergic Receptor-Mediated Myocyte Survival Signaling in Cell Culture Models.

In cultured cardiac myocytes, NE stimulates apoptotic cell death through activation of β1-ARs, whereas β2-ARs are believed to be cytoprotective (Mann et al., 1992; Xiao et al., 2004). Interestingly, several studies indicate that α1-ARs are also cytoprotective and act antithetically to β1-ARs. In NRVM, the α1-AR agonist phenylephrine inhibits apoptosis induced by the β-agonist isoproterenol (Iwai-Kanai et al., 1999; Zhu et al., 2000), nonhydrolyzable cAMP analogs (Iwai-Kanai et al., 1999; Zhu et al., 2000), hypoxia (Zhu et al., 2000), serum starvation (Zhu et al., 2000), 2-deoxyglucose (Valks et al., 2002), and doxorubicin (Aries et al., 2004). Similar results are observed in cultured adult rat cardiac myocytes, where NE-induced apoptosis is abolished by the β-AR antagonist propranolol, but not the α1-AR antagonist prazosin (Communal et al., 1998; O'Connell et al., 2006).

Mechanistically, a variety of pathways are implicated in α1-AR-mediated survival signaling in cardiac myocytes. α1-AR survival signaling requires activation of ERK and subsequent regulation of Bcl-2 family members to stabilize the mitochondrial membrane (Iwai-Kanai et al., 1999; Zhu et al., 2000; Valks et al., 2002; Communal et al., 2003; Huang et al., 2007; Wright et al., 2008, 2012). In NRVM, phenylephrine inhibits apoptosis induced by serum starvation and hypoxia by preventing downregulation of Bcl-2 and Bcl-X mRNA and protein levels (Zhu et al., 2000). In addition, phenylephrine induces the phosphorylation of Bcl-2 family member Bad at Ser112 and Ser155, preventing 2-deoxyglucose-induced apoptosis (Valks et al., 2002). Interestingly, cAMP-dependent protein kinase (PKA), known to be downstream of β1-ARs, also stimulates phosphorylation of Bad at Ser136 (Valks et al., 2002). This finding might suggest that both β1-AR and α1-AR signaling converge on a single molecule to modulate cell survival, implying that the balance between β1- and α1-AR signaling could control cell fate. However, how the interplay between α1-AR and β-AR phosphorylation of Bad impacts cardiac myocyte survival is unclear.

Other studies propose a role for ERK as a regulator of α1-AR survival signaling. ERK mediates cytoprotective signaling in cardiac myocytes (Lips et al., 2004), and α1-AR mediated activation of ERK is a well-characterized signaling pathway involved in hypertrophy (Bueno et al., 2000; Xiao et al., 2001; Barron et al., 2003; O'Connell et al., 2003). In NRVM, the MEK-1 inhibitor PD098059 [2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one] negates phenylephrine-mediated survival signaling (Iwai-Kanai et al., 1999). Furthermore, in cultured adult rat cardiac myocytes, NE-mediated activation of ERK upregulates β1-integrin and protects cells against β1-AR-mediated apoptosis (Communal et al., 2003).

The most convincing evidence for ERK in α1-mediated survival signaling comes from α1ABKO myocytes. Cultured α1ABKO mouse myocytes have markedly increased necrosis and apoptosis with toxic stimuli, including hydrogen peroxide, doxorubicin, and β-AR stimulation (O'Connell et al., 2006; Huang et al., 2007). This sensitivity to death stimuli in α1ABKO myocytes is rescued by expression of the α1A-subtype but not by the α1B, indicating that the α1A is necessary and sufficient for myocyte survival (Huang et al., 2007). Furthermore, rescue is mimicked by expression of constitutively activated MEK, which increases ERK activity, and rescue by the α1A is prevented by dominant negative MEK, which inhibits ERK (Huang et al., 2007). Together, these experiments define an α1A-ERK pathway for myocyte survival (Huang et al., 2007).

Potential mediators downstream of ERK include p90Rsk, a potential kinase for α1-AR-mediated phosphorylation of Ser112 in Bad (Valks et al., 2002), and the transcription factors GATA4 and nuclear factor of activated T cells (NFAT) (Pu et al., 2003; Aries et al., 2004).

Overall, the data from cultured cardiac myocytes indicate that α1-ARs mediate survival signaling, potentially through α1A activation of ERK, leading to regulation of Bcl-2 family members and stabilization of the mitochondrial membrane, and/or by induction of GATA4 and NFAT.

2. α1-Adrenergic Receptor-Mediated Myocyte Survival Signaling in Animal Models.

In mice, cats, and dogs, long-term infusion of subpressor doses of the mixed α1/β-AR agonist NE produces an adaptive hypertrophy without increased cell death or fibrosis (Laks et al., 1973; King et al., 1987; Marino et al., 1991; Patel et al., 1991; Stewart et al., 1992; Vecchione et al., 2002). Furthermore, α1-AR stimulation in isolated, perfused hearts prevents ischemia-reperfusion-induced cell apoptosis and necrosis in mice (Tejero-Taldo et al., 2002), rats (Banerjee et al., 1993; Mitchell et al., 1995; Tosaki et al., 1995; Meng et al., 1996a,b, 1999; Meldrum et al., 1997; Imani et al., 2008), rabbits (Bankwala et al., 1994; Tsuchida et al., 1994; Cope et al., 1997; Baghelai et al., 1999a,b), and dogs (Kitakaze et al., 1987, 1991, 1994; Node et al., 1997).

However, gain-of-function α1-AR transgenic models are inconsistent in demonstrating that α1-ARs protect against cardiac cell death. Cardiac myocyte-specific transgenic overexpression of the α1A-subtype at high levels (66-fold) protects against pathologic stress from pressure overload induced by aortic constriction and ischemic injury induced by coronary artery ligation, although the mechanism is linked to a basal hypercontractile phenotype rather than prevention of cardiac myocyte death (Du et al., 2004, 2006). Moreover, by 1 year, ventricles from these α1A-transgenic mice show increased fibrosis and apoptotic labeling, and the mice die prematurely, indicating that long-term, very high-level α1A-subtype overexpression can be associated with cell death rather than survival signaling (Chaulet et al., 2006).

Although transgenic overexpression of the α1B-subtype shows a variable ability to induce hypertrophy, prolonged overexpression of the α1B-subtype in some models, although not directly linked to increased cell death, induces a pathologic remodeling (Grupp et al., 1998; Iaccarino et al., 2001; Lemire et al., 2001; Wang et al., 2000). The failure of these gain-of-function models to recapitulate the findings in NRVM or in other animal models might be linked to the high levels of overexpression in most of these models or failure of overexpressed receptors to recapitulate signaling by ligand-activated endogenous receptors.

Conversely, loss-of-function models clearly indicate that α1-ARs prevent cardiac myocyte cell death. In α1ABKO mice, which lack the two α1-AR subtypes expressed in cardiac myocytes, aortic constriction induces a worse dilated cardiomyopathy, accompanied by a significant increase in cardiac cell apoptosis and fibrosis, leading to decreased function and increased mortality compared with WT. Cultured α1ABKO cardiac myocytes have increased susceptibility to several pro-death agonists, as reviewed in the preceding section (O'Connell et al., 2006; Huang et al., 2007, 2008), and this is rescued by adenoviral mediated reconstitution of the α1A-subtype, but not the α1B-subtype, in a pathway that requires ERK (Huang et al., 2007). The absence of this α1A-subtype ERK survival signaling might explain, at least partially, the negative outcome in α1ABKO mice subjected to aortic constriction (O'Connell et al., 2006).

In support of the finding that the α1A-subtype is both sufficient and necessary to prevent cardiac myocyte death, long-term infusion of a subpressor concentration of the α1A-subtype-specific agonist A61603 [N-[5-(4,5-dihydro-1H-imidazol-2yl)-2-hydroxy-5,6,7,8-tetrahydronaphthalen-1-yl]methanesulphonamide hydrobromide] prevents cell death and pathologic remodeling associated with doxorubicin-induced cardiotoxicity (Chan et al., 2008; Dash et al., 2011).

3. Summary of α1-Adrenergic Receptor-Mediated Myocyte Survival Signaling.

Studies in cultured neonatal and adult cardiac myocytes show that α1-ARs mediate survival signaling, most likely through activation of ERK and subsequent regulation of Bcl-2 family members to preserve mitochondrial membrane stability, as well as induction of protective transcription factors, such as GATA4 and NFAT, with their own downstream effectors. These findings generally support early studies in which catecholamine infusion in several animal models stimulated an adaptive hypertrophy without cell death or fibrosis. However, gain-of-function studies in α1A- and α1B-transgenic mouse models are inconsistent with regard to survival signaling, possibly due to massive levels of overexpression and/or aberrant signaling by constitutively activated receptors. More importantly, loss-of-function models, particularly α1ABKO mice, support the notion that α1-ARs mediate survival signaling. Furthermore, studies in cultured α1ABKO cardiac myocytes with reconstitution of the α1A-subtype define an α1A-subtype ERK survival signaling pathway, the absence of which could at least partially explain the maladaptive responses to pathologic stress in α1ABKO mice.

C. α1-Adrenergic Receptors Augment Contractile Function

Contractile dysfunction is a major component of and a causative factor in heart failure progression. Furthermore, as β-AR-mediated inotropy declines in heart failure due to receptor desensitization and downregulation, α1-AR mediated inotropy, which contributes little to basal contractile function, might function in a compensatory role to preserve contractile function in the failing heart.

1. α1-Adrenergic Receptor Activation of Contraction in In Vitro Models.

In many species, α1-ARs induce a positive inotropic response in left ventricular myocytes, trabeculae, and the perfused heart (Endoh and Blinks, 1988; Terzic et al., 1992; Turnbull et al., 2003). However, in mice, the α1-AR inotropic response can be negative in a few left ventricular preparations, including isolated papillary muscles and a minority of isolated cardiac myocytes (Hirano et al., 2006; Chu et al., 2013). Interestingly, populations of cardiac myocytes from the right and left ventricle have a fraction of myocytes that have a positive inotropic response to phenylephrine and an increased Ca2+ transient, prominent in the left ventricle, and a fraction of myocytes that have a negative inotropic response to phenylephrine and a decreased Ca2+ transient, mainly in the right ventricle (Chu et al., 2013).

Another unexpected finding in mouse heart is that α1-ARs mediate negative inotropy in myocardium from the normal right ventricle but positive inotropy in left ventricular myocardium (Wang et al., 2010). Surprisingly, in heart failure caused by myocardial infarction, α1-AR inotropy in right ventricular myocardium switches from negative to positive, and α1-AR positive inotropy in left ventricular myocardium is preserved (Litwin et al., 1995; Wang et al., 2010). These two aspects of α1-AR-mediated positive inotropy in heart failure after myocardial infarction can be interpreted as adaptive, that is, undiminished inotropy in the left ventricle (Litwin et al., 1995; Wang et al., 2010) and the appearance of positive inotropy in the right ventricle (Wang et al., 2010). Notably, right ventricular failure predicts worse outcomes in patients with left ventricular failure (Bleasdale and Frenneaux, 2002).

Interestingly, some find that the α1A- and α1B-subtypes differentially regulate contraction, with the α1A-subtype mediating a positive response and the α1B-subtype a negative response (Gambassi et al., 1998; Lin et al., 2001; Ross et al., 2003; O-Uchi et al., 2008).

A multitude of mechanisms are proposed to explain α1-AR-mediated positive or negative inotropy. Proposed mechanisms include inhibition of outward K+ currents and increased action potential duration (Apkon and Nerbonne, 1988; Fedida et al., 1989, 1990, 1991; Ravens et al., 1989; Tohse et al., 1990, 1992; Wang et al., 1991, 2001; Braun et al., 1992; Sato and Koumi, 1995; Gaughan et al., 1998 ); inhibition of L-type Ca2+ channel current (Chen et al., 1996; Gaughan et al., 1998; Belevych et al., 2001) or activation (Zhang et al., 1998; Mohl et al., 2011; Chu et al., 2013); activation of the Na+/H+ exchanger and intracellular acidification (Gambassi et al., 1992; Terzic et al., 1992); and regulation of myofilament Ca2+ sensitivity through phosphorylation of myosin light chain and/or cardiac troponin I (Hartmann et al., 1995; Andersen et al., 2002; McCloskey et al., 2003; MacGowan et al., 2005; Wang et al., 2006, 2010).

2. α1-Adrenergic Receptor-Mediated Contraction in Transgenic and Gene-Deletion Mouse Models.

Cardiac myocyte-specific transgenic overexpression of the α1A-subtype at high levels (148- to 170-fold) increases basal contractile function (Lin et al., 2001) and limits pathologic remodeling from pressure overload and ischemic injury (Du et al., 2004, 2006). These results suggest that the α1A-subtype mediates positive inotropic responses, in agreement with recent in vitro studies (Mohl et al., 2011; Chu et al., 2013). Conversely, transgenic overexpression of the α1B-subtype can be associated with depressed contractile function and pathologic remodeling in the heart (Grupp et al., 1998; Wang et al., 2000; Iaccarino et al., 2001; Lemire et al., 2001).

Loss-of-function models suggest that α1-AR-mediated inotropic responses are not required for basal contractile function, but might prevent contractile decline in response to pathologic stress. Basal contractile function by echocardiography is normal in both α1AKO and α1BKO mice (Rokosh and Simpson, 2002; Vecchione et al., 2002). In a similar fashion, basal contractile function assessed by echocardiography in α1ABKO mice is similar to WT, although cardiac output is decreased, due to a slight bradycardia and reduced left-ventricular volume, and exercise performance is impaired, presumably for the same reasons (O'Connell et al., 2003). Interestingly, calcium sensitivity is increased and maximal force is decreased in isolated muscle strips from α1ABKO mice, suggesting subtle abnormalities in basal contractile function with long-term absence of α1-ARs (McCloskey et al., 2003). More strikingly, contractile function is impaired significantly by aortic constriction in α1ABKO mice (O'Connell et al., 2006). This contractile dysfunction might be caused by both the loss of α1-AR-mediated positive inotropy and the absence of α1-AR-mediated survival signaling and adaptive hypertrophic effects, such as increased myosin synthesis. The exaggerated contractile dysfunction in the α1ABKO confirms that α1-AR-mediated inotropy could play an important protective role in response to pathologic stress in the heart.

3. α1-Adrenergic Receptor Activation of Contraction in Humans.

In healthy young women, systemic infusion of the α1-AR agonist methoxamine increases contractility determined noninvasively (Curiel et al., 1989). In both healthy patients and patients with New York Heart Association (NYHA) II–IV heart failure, infusion of the α1-AR agonist phenylephrine into the left main coronary artery increases contractility measured as dp/dt, demonstrating α1-AR inotropy in both healthy and heart failure patients (Landzberg et al., 1991). Interestingly, in the same patients, the α1-AR antagonist phentolamine shows no effect on baseline contractile function, suggesting that there is little contribution of α1-AR inotropy to basal contractile function in humans (Landzberg et al., 1991).

Surprisingly, α1-AR-mediated inotropy can equal β-AR-mediated inotropy in trabeculae isolated from failing human hearts (Skomedal et al., 1997). This suggests that in human heart failure, as β-ARs are desensitized and downregulated, α1-AR mediated contractility might act in a compensatory role to maintain contractile function. Similarly, inhalation of the α1-AR agonist methoxamine improves exercise performance in patients with significant left ventricular contractile dysfunction (Cabanes et al., 1992). Furthermore, in a small cohort of patients with hypotension due to end stage heart failure, the use of the α1-AR agonist midodrine is associated with increased contractile function (Zakir et al., 2009). The authors attributed the benefit to a modest increase in blood pressure that resulted from activation of vascular α1-ARs, permitting up-titration of recommended heart failure medications (Zakir et al., 2009). However, it is possible that direct activation of cardiac α1-ARs contributed to this beneficial effect.

4. Summary of α1-Adrenergic Receptor Activation of Contraction.

Studies with isolated myocytes, muscle strips, and perfused hearts show that α1-ARs can induce either positive or negative inotropic responses by altering K+ and Ca2+ currents, intracellular pH, and myofilament Ca2+ sensitivity. Mouse models confirm that the α1A-subtype can induce positive inotropic responses in vitro and in vivo and that α1A-subtype-mediated inotropy might protect the heart from pathologic stress. Importantly, α1-AR-mediated positive inotropy is documented in intact humans and can be equal to β-AR inotropy in isolated trabeculae from human heart failure patients, identifying a clinically relevant adaptive function for α1-ARs.

D. α1-Adrenergic Receptors Induce Ischemic Preconditioning

Ischemic preconditioning is an intrinsic protective mechanism in the heart whereby transient periods of ischemia protect the myocardium from damage due to longer bouts of ischemia, and protection can be observed both early (minutes to hours after ischemia) and late (hours to days). Pharmacologic agents can also induce preconditioning, and α1-ARs are among the most effective (Jensen et al., 2011).

1. α1-Adrenergic Receptor-Mediated Preconditioning in Animal Models.

In several animal models, including dog (Kitakaze et al., 1987, 1991, 1994; Node et al., 1997), rabbit (Bankwala et al., 1994; Tsuchida et al., 1994; Cope et al., 1997; Baghelai et al., 1999a,b), rats (Banerjee et al., 1993; Mitchell et al., 1995; Tosaki et al., 1995; Meng et al., 1996a,b, 1999; Meldrum et al., 1997; Imani et al., 2008) and mice (Tejero-Taldo et al., 2002), α1-ARs induce both early and late preconditioning, through a variety of mechanisms including adenosine release (Kitakaze et al., 1991), activation of 5′-nuleotidase activity (Kitakaze et al., 1994; Node et al., 1997), activation of PKC (Tsuchida et al., 1994; Mitchell et al., 1995; Node et al., 1997; Meng et al., 1999), regulation of Bcl2 family members (Baghelai et al., 1999a), induction of heat-shock proteins and protein synthesis (Meng et al., 1996a,b), activation of mitochondrial K-ATP channels (Imani et al., 2008), and induction of iNOS (Tejero-Taldo et al., 2002; Zhao et al., 2012). Once again though, poor specificity of antagonists for the α1-subtypes has made it difficult to define the α1-AR subtype(s) responsible for ischemic preconditioning.

However, transgenic overexpression of constitutively active mutants of the α1A- and α1B-subtypes reveals that the α1A-subtype, but not the α1B, mediates ischemic preconditioning that might not involve PKC (Rorabaugh et al., 2005). Preconditioning by the α1A in a rat transgenic model could be mediated by MEK/ERK phosphorylation and iNOS (Zhao et al., 2012). Similarly, cardiac myocyte-specific transgenic overexpression of the α1A-subtype suppresses ischemia-reperfusion-induced IP3 generation in isolated, perfused hearts (Amirahmadi et al., 2008). Conversely, cardiac myocyte-specific transgenic overexpression of the α1B-subtype does not prevent ischemic preconditioning or prevent ischemic-reperfusion injury (Gao et al., 2000).

2. α1-Adrenergic Receptor-Mediated Preconditioning in Humans.

In human atrial and ventricular muscle strips, α1-ARs mediate ischemic preconditioning through activation of PKC, p38 MAPK, and opening of mitochondrial K-ATP channels (Cleveland et al., 1996, 1997; Loubani and Galinanes, 2001, 2002).

3. Summary of α1-Adrenergic Receptor-Mediated Preconditioning.

In short, the data indicate that α1-ARs, most likely the α1A-subtype based on studies in mouse and rat, induce ischemic preconditioning and prevent cardiac myocyte death from ischemic injury.

E. Conclusions: α1-Adrenergic Receptors Are Cardioprotective

Three fundamental conclusions emerge based on 30 years of studies examining the physiologic function of α1-ARs in the heart that tend to contradict the conventional wisdom regarding cardiac α1-ARs.

1. α1-Adrenergic Receptors are Cardioprotective and Prevent Pathologic Remodeling in Heart Failure Unlike Other Gq-Coupled Receptors.

Specifically, the α1A-subtype induces cardioprotection and positive inotropy, whereas the α1B-subtype might be required for an adaptive, physiologic hypertrophy. Cardiac remodeling mediated by Gq-coupled receptors, such as α1-ARs, ETRs, and ATRs, is arguably the most significant physiologic function of these receptors in the heart. Currently, it is widely believed that all Gq-coupled receptors mediate a pathologic hypertrophic response. The idea that Gq-signaling is pathologic is based primarily on three lines of evidence, as reviewed previously (Dorn and Brown, 1999; Adams and Brown, 2001). First, Gq-agonists, such as phenylephrine (an α1-AR agonist), endothelin, and angiotensin, induce hypertrophy with expression of the “fetal genes” in cultured NRVM, and re-expression of the “fetal genes” is classically associated with pathologic ventricular remodeling, as reviewed previously (Dorn and Brown, 1999). Second, mouse models targeting overexpression of ETRs or ATRs suggest that these receptors generally induce pathologic remodeling (Ainscough et al., 2009; Paradis et al., 2000; Yang et al., 2004). Clinically, ATR blockers are used to treat heart failure (Chrysant, 2008), although current ETR antagonists have no proven efficacy for heart failure (Mylona and Cleland, 1999; McMurray et al., 2007). Third, cardiac-specific transgenic overexpression of Gαq in mice induces pathologic remodeling with increased cardiac myocyte apoptosis, as reviewed (Dorn and Brown, 1999; Dorn, 2005). However, high-level Gαq-overexpression is required for pathology, as reviewed previously (Jensen et al., 2011). Furthermore, the data summarized in this section clearly challenge the dogma that all Gq-coupled receptor signaling is pathologic. Instead, the data indicate α1-AR signaling can be cardioprotective.

2. α1-Adrenergic Receptor-Mediated Cardioprotective Signaling Can Explain the Worsening of Heart Failure with α1-Blockers Observed in Clinical Trials.

As detailed in the next section (section V), α1-blockers exacerbate heart failure and are associated with worse outcomes in patients with hypertension (ALLHAT), heart failure (V-HeFT), and benign prostatic hyperplasia (Cohn, 1993; Cohn et al., 1986; ALLHAT, 2000, 2003; Dhaliwal et al., 2009). Hallmarks of ventricular remodeling in heart failure include contractile dysfunction (both systolic and diastolic), pathologic hypertrophy, and increased cardiac cell death and fibrosis (Anand and Florea, 2003). Importantly, α1ABKO mice by virtue of their lack of cardiac myocyte α1-ARs approximate the use of α1-blockers (O'Connell et al., 2003). In the α1ABKO mouse model, pathologic stress from aortic constriction causes hypertrophy with failed gene transcription, increased cardiac cell death, increased fibrosis, and worsened contractile function, leading to dilated cardiomyopathy, heart failure, and ultimately 50% mortality (O'Connell et al., 2003, 2006). Follow-up studies in cultured α1ABKO cardiac myocytes define a cardioprotective α1A-subtype signaling pathway, identifying a direct requirement for protective α1-AR signaling in cardiac myocytes, the absence of which could at least partially explain the negative outcomes in α1ABKO mice (Huang et al., 2007, 2008). Support for the assertion that α1-ARs are cardioprotective can also be drawn from studies in gain-of-function models, where α1A-subtype overexpression protects against pathologic stress (Du et al., 2004, 2006; Lin et al., 2001). In combination, these studies indicate that α1-ARs are both sufficient to induce and required for cardioprotective signaling. In summary, these data provide a mechanistic basis to explain the negative outcomes in clinical trials with α1-blockers.

3. α1-Agonist Therapies Might Improve Heart Failure Outcomes.

On the basis of the accumulated evidence, α1-AR activation of adaptive hypertrophy, prevention of cardiac myocyte death, augmentation of contractility, and induction of ischemic preconditioning could prevent worsened outcomes from systolic heart failure. This provides the foundation of the argument for α1-AR agonist therapy in heart failure. However, several counterarguments exist (Jensen et al., 2011). First, in transgenic models, the α1B-subtype can worsen function and induce dilated cardiomyopathy (Grupp et al., 1998; Wang et al., 2000; Iaccarino et al., 2001; Lemire et al., 2001). However, pharmacology and knockouts argue against the results seen in certain cardiac transgenics. Specifically, studies with α1-agonists in mouse (Chan et al., 2008; Dash et al., 2011) and human (Cleveland et al., 1996, 1997; Loubani and Galinanes, 2001, 2002), as well as in loss-of-function mouse models (O'Connell et al., 2003, 2006; Huang et al., 2007) as reviewed above, support α1-mediated cardioprotective effects.

Second, α1-ARs induce vasoconstriction, which is contraindicated in heart failure. However, in mice, subpressor doses of an α1A-subtype-specific ligand prevent doxorubicin cardiotoxicity (Chan et al., 2008; Dash et al., 2011); in humans, some small trials indicate α1-ARs agonists might improve function in heart failure (Cabanes et al., 1992; Zakir et al., 2009); and numerous studies identify adaptive hypertrophy with subpressor α1-agonist infusion, as reviewed in the section on hypertrophy. These data provide a preliminary proof-of-principle demonstration of the efficacy of α1-agonists in heart failure, and justify further study.

A third argument against α1-agonist therapy is that the mixed α1/β-blocker carvedilol has proven efficacy in heart failure. However, as discussed in the next section (section V), current evidence suggests that the α1-blocking properties of carvedilol are not sustained in long-term dosing (Kubo et al., 2001; Hryniewicz et al., 2003), and conversely carvedilol might potentiate α1-AR signaling (Van Tassell et al., 2008).

Fourth, α1-ARs are Gq-coupled receptors, and the conventional wisdom is that Gq-signaling exacerbates pathologic remodeling (Jensen et al., 2011). However, as mentioned already, α1-ARs clearly do not fit this paradigm. In summary, despite the caveats listed, α1-AR agonist therapy might present a novel effective treatment of heart failure.

V. α1-Adrenergic Receptors in Human Heart Disease

The classic physiologic function of α1-ARs is to increase vascular smooth muscle contractility and hence blood pressure. α1-ARs are found in vascular beds throughout the body, including smooth muscle cells in arteries of the heart, brain, kidneys, and gut, as well as in smooth muscle in the prostate and bladder (Michelotti et al., 2000). By virtue of their ability to block α1-AR-mediated smooth muscle contractions, α1-AR antagonists (α1-blockers) are used to treat hypertension (Lund-Johansen and Omvik, 1991; Frishman and Kotob, 1999; Sica, 2005) and benign prostatic hyperplasia (Caine et al., 1976, 1978; Schwinn and Roehrborn, 2008; Michel, 2010).

More recently, it has become clear that α1-ARs might play a significant role in preventing the clinical progression of heart failure, as reviewed above. Heart failure is a clinical syndrome of varied etiology in which the heart cannot pump enough blood to meet the body's needs. Heart failure is characterized by neurohormonal augmentation, leading to increased catecholamine levels, which are thought to play a causative role in pathologic ventricular remodeling through increased activation of ARs. Indeed, increased blood NE levels are a primary finding in heart failure patients and predict disease severity and mortality (Cohn et al., 1984). Whereas this does not establish a causal relationship between increased NE levels and induction of heart failure, it was part of the rationale to block NE activation of ARs for therapy in heart failure.

In fact, clinical trials with β-AR antagonists or β-blockers, such as Metoprolol CR/XL Randomized Intervention Trial in Congestive Heart Failure (metoprolol) (MERIT, 1999), Cardiac Insufficiency Bisoprolol Study II (bucindolol) (CIBIS-II, 1999) and Carvedilol Prospective Randomized Cumulative Survival Trial (carvedilol) (Packer et al., 2001), show significant reductions in mortality in patients with heart failure (Foody et al., 2002; Teerlink and Massie, 1999; Chatterjee et al., 2013). On the basis of the success of β-blockers in improving outcomes in heart failure, β-blockers are standard of care in heart failure therapy (Hunt et al., 2005). This success has led to the notion that blocking all AR signaling in heart failure would be beneficial, and tests of this idea are reviewed below. Currently, 5.7 million Americans have heart failure, 1 million are admitted to the hospital each year, and the 5-year survival rate is only 50% (Roger et al., 2011). Thus, new drugs to treat heart failure are needed (Simpson, 2011). Here, we review the recent clinical trials suggesting that some AR signaling, particularly α1-AR signaling, might be beneficial in human heart failure. Table 1 summarizes the trials.

TABLE 1.

Summary of clinical trials involving α1-blockers in cardiovascular disease

Trial Treatment Groups and Patient Numbers
Inclusion Criteria Result Interpretation
Control Groups α1-Blocker Groups
ALLHAT (2000) (ALLHAT, 2000, 2003; SoRelle, 2000; Davis et al., 2002; Piller et al., 2002) Chlorthalidone (diuretic) [12.5–25 mg/day (N = 15,268)] Doxazosin (α1-blocker) [2–8 mg/day (N = 9067)] Men and women age 55 or older with hypertension (systolic 140 mm Hg and/or diastolic 90 mm Hg, or on medication for hypertension) and at least 1 other risk factor for coronary heart disease Trial stopped early. Doxazosin increased cardiovascular events by 25% and doubled the risk of heart failure. α1-Blockade worsens outcomes in hypertension with cardiac risk factors.
V-HeFT I and II (Cohn et al., 1986; Cohn, 1993) V-HeFT I
Placebo (N = 273); Hydralazine/isosorbide dinitrate (direct vasodilators) [300/160 mg/day (N = 186)]V-HeFT II
Hydralazine/isosorbide dinitrate
Enalapril (ACE inhibitor) (N = 403) Prazosin (α1-blocker) [20 mg/day (N = 183) Male veterans mean age 58 with symptomatic heart failure due to dilated cardiomyopathy (ischemic or nonischemic), on digoxin and diuretics Prazosin did not change left ventricular ejection fraction or mortality versus placebo; both were improved with hydralazine/isosorbide. α1-Blockade shows no benefit in heart failure and might worsen mortality.
Trend toward increased mortality at 5 years in prazosin group versus placebo, survival improved by other vasodilators.
α1-Blockers in BPH effect on HF (Dhaliwal et al., 2009) Tamsulosin (58%), terazosin (40%), or doxazosin (2%) (N = 98) Male veterans with dilated cardiomyopathy admitted with heart failure (N = 388 overall) Among the 25% of patients taking α1-blockers, most for BPH, subsequent hospitalizations for heart failure were increased in patients receiving α1-blockers without β-blockers. α1-Blockade worsens outcomes in BPH in the absence of β-blockade.
COMET (Carvedilol or Metoprolol European Trial) (Poole-Wilson et al., 2002, 2003) Metoprolol tartrate (β1-selective antagonist) (50 mg bid; N = 1518) Carvedilol (β1/2-,α1-AR antagonist) [25 mg bid (N = 1511)] Men (80%) and women mean age 62 with NYHA class II–IV heart failure due to dilated cardiomyopathy (ischemic or nonischemic) on stable therapy Mortality reduced in carvedilol group versus metoprolol. Carvedilol reduces mortality, possibly related in part to potentiation of α1-AR signaling.
Subsequent analyses indicates that benefit might not be due to α1-blockade (Kubo et al., 2001; Hryniewicz et al., 2003; Van Tassell et al., 2008)
MOXSE (Swedberg et al., 2002) Placebo (N = 38) Moxonidine (sympatholytic) [0.3-1.5 mg bid (N = 227 total)] Patients NYHA class II–IV heart failure due to dilated cardiomyopathy (ischemic or nonischemic) Moxonidine reduced plasma NE and heart rate but increased adverse events Some degree of AR signaling is cardioprotective.
MOXCON (Coats, 1999; Cohn et al., 2003; Pocock et al., 2004) Placebo (N = 944) Moxonidine (1.5 mg bid [N = 990)] As in MOXSE Trial stopped early Some degree of AR signaling is cardioprotective.
Moxonidine reduced NE and increased morbidity and mortality
BEST (Bristow et al., 2004) Placebo [3 month (N = 845); 12 month (N = 654)]; Total for BEST (N = 2708) Bucindolol (β1/2-AR antagonist, sympatholytic) [3 month (N = 841); 12 month (N = 642)] Patients NYHA class III–IV heart failure due to dilated cardiomyopathy (ischemic or nonischemic) on stable therapy, subgroup with NE measured at 3 and 12 months Bucindolol reduced NE and increased mortality. Some degree of AR signaling is cardioprotective.

A. Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial: An α1-Adrenergic Receptor Antagonist in Hypertension Increases the Risk of Heart Failure

The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) was a large, randomized, double-blind, active controlled trial initiated in 1994 and funded by the National Heart, Lung, Blood Institute (ALLHAT, 2000, 2003). ALLHAT was designed to compare new treatments for hypertension versus older, standard treatments. Primary outcomes were fatal coronary heart disease and nonfatal myocardial infarction, and secondary outcomes included all-cause mortality, stroke, and combined cardiovascular disease. In one arm, 24,335 patients with hypertension and at least one other risk factor for coronary heart disease were randomized to the diuretic chlorthalidone (15,268) or the nonselective α1-AR antagonist (α1-blocker) doxazosin (9067) and were to be followed for 4–8 years.

In 2000, the ALLHAT data safety and monitoring board stopped this arm of the trial early, citing that patients on doxazosin had 25% more cardiovascular events and a significant doubling in the risk of heart failure versus patients on chlorthalidone (SoRelle, 2000). Although systolic blood pressure was approximately 3 mm Hg higher in the doxazosin group, the ALLHAT investigators concluded that this difference was unlikely to account for the doubling in the risk of heart failure (Davis et al., 2002; ALLHAT, 2003), and heart failure events were validated (Piller et al., 2002). Furthermore, approximately 60% of patients in both groups were on additional treatments to reduce blood pressure, but a follow up analysis revealed that the risk of heart failure was not reduced by this additional antihypertensive treatment, thereby confirming the initial findings (Davis et al., 2002). Subsequently, the ALLHAT results led to the recommendation against α1-blockers as a primary treatment of high blood pressure (Messerli, 2001); compliance with this recommendation has been modest (Stafford et al., 2004).

When it was initiated, ALLHAT was the largest trial yet to compare the efficacy of different methods for treating hypertension on cardiovascular outcomes. At the time, it was assumed that lowering blood pressure, regardless of mechanism, would by itself reduce morbidity and mortality (Messerli, 2000). However, ALLHAT shows that clinical trials involving new antihypertensive treatments must examine multiple cardiovascular outcomes.

More importantly, ALLHAT demonstrates that blocking α1-ARs has a negative impact on the heart, further challenging the established dogma that elevated AR signaling in heart disease is always pathologic. Although ALLHAT does not prove that the negative effect of α1-blockade is a direct effect on the heart, a direct effect is supported by studies on the α1ABKO mice and other animal and human data discussed above.

B. Vasodilator-Heart Failure Trial: An α1-Adrenergic Receptor Antagonist Does Not Improve Survival in Heart Failure

The Vasodilator-Heart Failure Trial (V-HeFT), phase I, was a small trial initiated in 1980 involving 642 men diagnosed with chronic congestive heart failure. V-HeFT was designed to evaluate the effects of vasodilators on mortality as the primary outcome (Cohn et al., 1986). Prior to V-HeFT, studies indicated that reducing systemic vascular resistance improved hemodynamics in patients with heart failure (Cohn and Franciosa, 1977a,b), and V-HeFT was designed to test whether this principal would translate into clinical benefit. Patients with heart failure and taking digoxin and a diuretic, a common therapeutic regimen for heart failure at the time, were randomized to receive placebo, the α1-blocker prazosin, or the combination of hydralazine and isosorbide dinitrate. At a mean follow-up of 2.3 years, the combination of hydralazine/isosorbide increased cardiac function (ejection fraction) and reduced mortality, but function and mortality were the same as placebo for prazosin (Cohn et al., 1986).

In phase II, the angiotensin converting enzyme inhibitor enalapril was included, and mortality was tracked in all groups from phase I and II for 5 years. As in phase I, prazosin showed no benefit in phase II, and at 5 years, a trend toward increased mortality was observed (Cohn, 1993). Interestingly, prazosin was the only vasodilator that failed to show any positive outcome. Although not as clear-cut as the results from ALLHAT, the negative results with prazosin in V-HeFT again suggest that α1-AR blockade is harmful to the heart and that α1-AR activation is indeed beneficial.

A caveat to consider with both prazosin (V-HeFT) and doxazosin (ALLHAT) is that both antagonists can cause myocyte apoptosis, independent of their α1-blocking activity (Gonzalez-Juanatey et al., 2003). However, the dose required to cause apoptosis in culture (10 μM) (Gonzalez-Juanatey et al., 2003) is far higher than the peak plasma levels attained in clinical use, e.g., ∼200 nM for doxazosin (Fawzy et al., 1999). Furthermore, the concern that the α1-blockers prazosin and doxazosin might be maladaptive for the human heart via an off-target effect is largely obviated by the observation of maladaptive cardiac effects in the α1ABKO, as reviewed above.

C. α1-Adrenergic Receptor Antagonist Therapy in Benign Prostatic Hyperplasia Might Exacerbate Heart Failure

In the prostate, α1-ARs mediate smooth muscle contraction, which is the basis for α1-blocker use in benign prostatic hyperplasia (BPH). Currently, estimates indicate that 9.5 million men over 65 are diagnosed with benign prostatic hyperplasia (Vaughan, 2003), with common comorbidities for hypertension (87%) and a previous admission for heart failure (40%) (Dhaliwal et al., 2009). The U.S. Food and Drug Administration has approved five α1-blockers for BPH: silodosin (Rapaflo), an α1A-subtype-selective antagonist, and the nonselective α1-antagonists terazosin (Hytrin), doxazosin (Cardura), tamsulosin (Flomax), and alfuzosin (Uroxatral).

A recent meta-analysis of 388 men with heart failure revealed that cotreatment with an α1-blocker increased the risk of heart failure hospitalizations, unless patients were concurrently treated with a β-blocker for heart failure (Dhaliwal et al., 2009). Given that only two-thirds of patients in this trial were being treated with a β-blocker (Dhaliwal et al., 2009) and had the common comorbidities of BPH, hypertension, and heart failure, there is reason for concern regarding the safety of α1-blockers in BPH.

D. Carvedilol: A Nonselective β1/2-Adrenergic Receptor/α1-Adrenergic Receptor Antagonist for Heart Failure

Carvedilol is a nonselective β1/2-AR/α1-AR antagonist indicated for the treatment of heart failure based on its efficacy as established in several trials, for example, Carvedilol Prospective Randomized Cumulative Survival Trial (Packer et al., 2001, 2002) and US Carvedilol (Packer et al., 1996), as reviewed previously (Teerlink and Massie, 1999; Foody et al., 2002; Wollert and Drexler, 2002; Chatterjee et al., 2013). Interestingly, the Carvedilol or Metoprolol European Trial suggested that carvedilol extended survival relative to metoprolol, a β1-AR selective antagonist, in patients with NYHA Class II–IV heart failure (Poole-Wilson et al., 2002, 2003). Among other explanations for the benefit provided by carvedilol (Bristow et al., 2003), one hypothesis is that by blocking both β2- and α1-ARs, as well as β1-ARs, carvedilol confers additional benefit over selective β1-AR antagonism alone (Poole-Wilson et al., 2002).

As a mixed acting β1/β2/α1-blocker, carvedilol is proposed to block α1-mediated vasoconstriction to account for the added benefit. Indeed, in HEK cells expressing the human α1-subtypes, carvedilol has higher binding affinity for the α1B and α1D than for β-ARs or the α1A and selectively inhibits α1B- and α1D-subtype-specific calcium transients (Koshimizu et al., 2004). However, the idea that benefit with carvedilol relies on its α1-blocking properties might seem counterintuitive, given the failure of the α1-blocker prazosin to improve mortality in V-HeFT (Cohn et al., 1986; Cohn, 1993). Indeed, α1-mediated vasopressor responses are not reduced during chronic treatment with carvedilol (Kubo et al., 2001; Hryniewicz et al., 2003). These studies show that chronic carvedilol treatment does not inhibit α1-AR-mediated vascular contraction and further suggest that the long-term benefits of carvedilol are not likely due to α1-AR antagonism.

In another smaller study of patients with heart failure, the blood pressure responses to α1-AR agonist infusion (phenylephrine) were actually increased in patients receiving carvedilol (Van Tassell et al., 2008). These data were interpreted to mean that chronic carvedilol treatment actually potentiates α1-mediated vasoconstriction (Van Tassell et al., 2008). Although this was a small study, an effect on the heart is also possible, and the implications are that carvedilol might provide an added benefit in heart failure through augmented α1-AR signaling.

E. Sympatholytics: Reducing Norepinephrine Levels Does Not Improve Heart Failure

If the central tenet of heart failure therapy for the last several decades is correct, namely that increased catecholamine signaling exacerbates heart failure, then by extension, reducing catecholamine levels should improve heart failure outcomes. Following that logic, both the Moxonidine Safety and Efficacy Trial (MOXSE) (Swedberg et al., 2002) and the Moxonidine Congestive Heart Failure Trial (MOXCON) (Cohn et al., 2003) examined the effects of the sympatholytic imidazoline receptor agonist moxonidine on mortality in heart failure. MOXSE examined 268 patients with New York Heart Association class II–IV systolic heart failure and found that moxonidine reduced heart rate and modestly improved ejection fraction but increased adverse events.

MOXCON, the larger of the two trials, also targeted patients with New York Heart Association class II-IV systolic heart failure. After enrolling roughly 1900 patients, the trial was stopped early due to increased mortality in the moxonidine group (Coats, 1999; Cohn et al., 2003; Pocock et al., 2004).

Another clinical trial showed that the β1/β2-blocker/sympatholytic agent bucindolol increased mortality associated with pronounced NE reduction in the β-Blocker Evaluation of Survival Trial (BEST) (Bristow et al., 2004). In total, the failure of these trials suggests that some catecholamine signaling is beneficial in heart failure. Although the mechanism whereby sympatholysis leads to increased mortality is uncertain, it is possible that decreasing catecholamine levels abrogates the cardioprotective effects of α1-AR signaling.

F. Conclusions and Implications: Are Myocardial α1-Adrenergic Receptors Cardioprotective in Humans?

In summary, one large clinical trial and several smaller studies show that α1-blockers or a reduction in NE levels worsens outcomes in patients with hypertension or heart failure, as summarized in Table 1. One implication of these results is to force a reconsideration of the notion that all AR signaling in heart failure is pathologic. Although β-AR blockade is clearly beneficial in heart failure, the negative results of trials involving sympatholytics indicate that reducing NE levels excessively can be harmful. Perhaps more importantly, another implication of these results would be to suggest that myocardial α1-ARs are protective, based on the negative results in trials with α1-blockers. None of the trials involving α1-blockers were designed to address mechanisms whereby α1-AR inhibition worsens heart failure. However, as discussed in section IV, α1-ARs are clearly cardioprotective in cell and animal models.

VI. Final Summary

The functional significance of cardiac α1-ARs has dramatically advanced over the last 30 years since α1-ARs were first demonstrated to have a direct trophic effect on cardiac myocytes (Simpson, 1983). Clinical data now show that α1-blockers cause heart failure in hypertensive patients and possibly in patients taking α1-blockers for BPH. These clinical data imply a protective function for cardiac α1-ARs, and data indicate that unlike β1-ARs, α1-ARs are not downregulated in human heart failure but are proportionally increased, available to mediate protective signaling in heart failure. In fact, studies in cultured myocytes and animal models show that α1-ARs are cardioprotective and prevent pathologic remodeling in heart failure. The mechanisms are multifactorial, including activation of adaptive or physiologic hypertrophy, prevention of cardiac myocyte death, augmentation of positive inotropic responses, and induction of ischemic preconditioning. Importantly, these studies provide a mechanistic basis to explain the failure of α1-blockers in humans. Perhaps most surprising is the finding that α1-ARs localize to and signal at the nuclear membrane in adult cardiac myocytes and engage “inside-out” signaling to regulate α1-AR cardioprotective signaling. Overall, these findings raise several new questions regarding the functional role of cardiac α1-ARs. Perhaps the most important are whether activation of cardiac α1-ARs, especially the α1A subtype, might be a viable therapeutic strategy in heart failure and how nuclear α1-AR signaling might differ functionally from α1-AR signaling at the plasma membrane.

Abbreviations

α1-AR

α1-adrenergic receptor

β-AR

β-adrenergic receptor

α1A-subtype

α1A-adrenergic receptor

α1B-subtype

α1B-adrenergic receptor

α1-blocker

α1-adrenergic receptor antagonist

α1AKO

α1A-adrenergic receptor knockout

α1BKO

α1B-adrenergic receptor knockout

α1ABKO

α1AB-adrenergic receptor double knockout

αSkAct

α-skeletal actin

AR

adrenergic receptor

ATR

angiotensin receptor

ALLHAT

Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial

BPH

benign prostatic hyperplasia

BEST

β-Blocker Evaluation of Survival Trial

cav-3

caveolin-3

CAM

constitutively active mutant

DAG

diacylglycerol

ETR

endothelin receptor

EMT

extraneuronal monoamine transporter

ERK

extracellular signal–regulated kinase

GFP

green fluorescent protein

GPCR

G-protein-coupled receptor

HEK

human embryonic kidney

HW

heart weight

IP

inositol phosphate

IP3

inositol 1,4,5-trisphosphate

KO

knockout

MEK

mitogen-activated protein kinase kinase

MOXCON

Moxonidine Congestive Heart Failure Trial

MOXSE

Moxonidine Safety and Efficacy Trial

MyHC

myosin heavy chain

NE

norepinephrine

NFAT

nuclear factor of activated T cells

NRVM

neonatal rat ventricular myocyte

NYHA

New York Heart Association

OCT3

organic cation transporter 3

PLCβ1

phospholipase Cβ1

PKC

protein kinase C

PKD

protein kinase D

V-HeFT

Vasodilator-Heart Failure Trial

WT

wild type

Authorship Contributions

Wrote or contributed to the writing of the manuscript: O’Connell, Jensen, Baker, Simpson.

Footnotes

This work was supported by the National Institutes of Health National Institute of General Medical Sciences [Grants P20-RR017662 (to T.D.O.)]; National Institutes of Health National Heart, Lung, and Blood Institute [Grants K08-HL096836 (to B.C.J.) and R01-HL31113 (to P.C.S.)]; the Department of Veteran’s Affairs [Grants IO1-BX 001078 and 001970 (to P.C.S.), I01 BX000740 (to A.J.B.)]; the American Heart Association, Western States Affiliate (to P.C.S., A.J.B.); the American Heart Association, Greater Midwest Affiliate (to T.D.O.); the GlaxoSmithKline Research and Education Foundation for Cardiovascular Disease (to B.C.J.); and the University of California, San Francisco, Foundation for Cardiac Research (to B.C.J.).

References

  1. Adams JW, Brown JH. (2001) G-proteins in growth and apoptosis: lessons from the heart. Oncogene 20:1626–1634 [DOI] [PubMed] [Google Scholar]
  2. Adams JW, Sakata Y, Davis MG, Sah VP, Wang Y, Liggett SB, Chien KR, Brown JH, Dorn GW., 2nd (1998) Enhanced Gαq signaling: a common pathway mediates cardiac hypertrophy and apoptotic heart failure. Proc Natl Acad Sci USA 95:10140–10145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Agüero J, Almenar L, Montó F, Oliver E, Sánchez-Lázaro I, Vicente D, Martínez-Dolz L, D’Ocon P, Rueda J, Salvador A. (2012) Myocardial G protein receptor-coupled kinase expression correlates with functional parameters and clinical severity in advanced heart failure. J Card Fail 18:53–61 [DOI] [PubMed] [Google Scholar]
  4. Ainscough JF, Drinkhill MJ, Sedo A, Turner NA, Brooke DA, Balmforth AJ, Ball SG. (2009) Angiotensin II type-1 receptor activation in the adult heart causes blood pressure-independent hypertrophy and cardiac dysfunction. Cardiovasc Res 81:592–600 [DOI] [PubMed] [Google Scholar]
  5. Akhter SA, Milano CA, Shotwell KF, Cho MC, Rockman HA, Lefkowitz RJ, Koch WJ. (1997) Transgenic mice with cardiac overexpression of alpha1B-adrenergic receptors. In vivo alpha1-adrenergic receptor-mediated regulation of beta-adrenergic signaling. J Biol Chem 272:21253–21259 [DOI] [PubMed] [Google Scholar]
  6. ALLHAT Collaborative Research Group (2000) Major cardiovascular events in hypertensive patients randomized to doxazosin vs chlorthalidone: the antihypertensive and lipid-lowering treatment to prevent heart attack trial (ALLHAT) [published correction appears in JAMA (2002) 288:2976]. JAMA 283:1967–1975 [PubMed] [Google Scholar]
  7. Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial Collaborative Research Group (2003) Diuretic versus alpha-blocker as first-step antihypertensive therapy: final results from the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). Hypertension 42:239–246 [DOI] [PubMed] [Google Scholar]
  8. Amirahmadi F, Turnbull L, Du XJ, Graham RM, Woodcock EA. (2008) Heightened alpha1A-adrenergic receptor activity suppresses ischaemia/reperfusion-induced Ins(1,4,5)P3 generation in the mouse heart: a comparison with ischaemic preconditioning. Clin Sci (Lond) 114:157–164 [DOI] [PubMed] [Google Scholar]
  9. Anand IS, Florea VG. (2003) Alterations in ventricular structure: Role of left ventricular remodeling, in Heart Failure: A Companion to Braunwald's Heart Disease (Mann DL, ed) pp 229–245, WB Saunders Co. [Google Scholar]
  10. Andersen GO, Qvigstad E, Schiander I, Aass H, Osnes JB, Skomedal T. (2002) Alpha(1)-AR-induced positive inotropic response in heart is dependent on myosin light chain phosphorylation. Am J Physiol Heart Circ Physiol 283:H1471–H1480 [DOI] [PubMed] [Google Scholar]
  11. Apkon M, Nerbonne JM. (1988) Alpha 1-adrenergic agonists selectively suppress voltage-dependent K+ current in rat ventricular myocytes. Proc Natl Acad Sci USA 85:8756–8760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Aries A, Paradis P, Lefebvre C, Schwartz RJ, Nemer M. (2004) Essential role of GATA-4 in cell survival and drug-induced cardiotoxicity. Proc Natl Acad Sci USA 101:6975–6980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Autelitano DJ, Woodcock EA. (1998) Selective activation of alpha1A-adrenergic receptors in neonatal cardiac myocytes is sufficient to cause hypertrophy and differential regulation of alpha1-adrenergic receptor subtype mRNAs. J Mol Cell Cardiol 30:1515–1523 [DOI] [PubMed] [Google Scholar]
  14. Avkiran M, Rowland AJ, Cuello F, Haworth RS. (2008) Protein kinase d in the cardiovascular system: emerging roles in health and disease. Circ Res 102:157–163 [DOI] [PubMed] [Google Scholar]
  15. Backs J, Backs T, Bezprozvannaya S, McKinsey TA, Olson EN. (2008) Histone deacetylase 5 acquires calcium/calmodulin-dependent kinase II responsiveness by oligomerization with histone deacetylase 4. Mol Cell Biol 28:3437–3445 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Backs J, Song K, Bezprozvannaya S, Chang S, Olson EN. (2006) CaM kinase II selectively signals to histone deacetylase 4 during cardiomyocyte hypertrophy. J Clin Invest 116:1853–1864 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Baghelai K, Graham LJ, Wechsler AS, Jakoi ER. (1999a) Delayed myocardial preconditioning by alpha1-adrenoceptors involves inhibition of apoptosis. J Thorac Cardiovasc Surg 117:980–986 [DOI] [PubMed] [Google Scholar]
  18. Baghelai K, Graham LJ, Wechsler AS, Jakoi ER. (1999b) Phenylephrine induces delayed cardioprotection against necrosis without amelioration of stunning. Ann Thorac Surg 68:1219–1224 [DOI] [PubMed] [Google Scholar]
  19. Baines CP. (2010) Role of the mitochondrion in programmed necrosis. Front Physiol 1:156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska H, Hambleton MA, Brunskill EW, Sayen MR, Gottlieb RA, Dorn GW, et al. (2005) Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 434:658–662 [DOI] [PubMed] [Google Scholar]
  21. Banerjee A, Locke-Winter C, Rogers KB, Mitchell MB, Brew EC, Cairns CB, Bensard DD, Harken AH. (1993) Preconditioning against myocardial dysfunction after ischemia and reperfusion by an alpha 1-adrenergic mechanism. Circ Res 73:656–670 [DOI] [PubMed] [Google Scholar]
  22. Bankwala Z, Hale SL, Kloner RA. (1994) Alpha-adrenoceptor stimulation with exogenous norepinephrine or release of endogenous catecholamines mimics ischemic preconditioning. Circulation 90:1023–1028 [DOI] [PubMed] [Google Scholar]
  23. Barron AJ, Finn SG, Fuller SJ. (2003) Chronic activation of extracellular-signal-regulated protein kinases by phenylephrine is required to elicit a hypertrophic response in cardiac myocytes. Biochem J 371:71–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Belevych AE, Nulton-Persson A, Sims C, Harvey RD. (2001) Role of tyrosine kinase activity in alpha-adrenergic inhibition of the beta-adrenergically regulated L-type Ca(2+) current in guinea-pig ventricular myocytes. J Physiol 537:779–792 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Bishopric NH, Simpson PC, Ordahl CP. (1987) Induction of the skeletal alpha-actin gene in alpha 1-adrenoceptor-mediated hypertrophy of rat cardiac myocytes. J Clin Invest 80:1194–1199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bkaily G, Avedanian L, Jacques D. (2009) Nuclear membrane receptors and channels as targets for drug development in cardiovascular diseases. Can J Physiol Pharmacol 87:108–119 [DOI] [PubMed] [Google Scholar]
  27. Bleasdale RA, Frenneaux MP. (2002) Prognostic importance of right ventricular dysfunction. Heart 88:323–324 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Böhm M, Diet F, Feiler G, Kemkes B, Erdmann E. (1988) Alpha-adrenoceptors and alpha-adrenoceptor-mediated positive inotropic effects in failing human myocardium. J Cardiovasc Pharmacol 12:357–364 [DOI] [PubMed] [Google Scholar]
  29. Boivin B, Chevalier D, Villeneuve LR, Rousseau E, Allen BG. (2003) Functional endothelin receptors are present on nuclei in cardiac ventricular myocytes. J Biol Chem 278:29153–29163 [DOI] [PubMed] [Google Scholar]
  30. Boivin B, Lavoie C, Vaniotis G, Baragli A, Villeneuve LR, Ethier N, Trieu P, Allen BG, Hébert TE. (2006) Functional beta-adrenergic receptor signalling on nuclear membranes in adult rat and mouse ventricular cardiomyocytes. Cardiovasc Res 71:69–78 [DOI] [PubMed] [Google Scholar]
  31. Boivin B, Vaniotis G, Allen BG, Hébert TE. (2008) G protein-coupled receptors in and on the cell nucleus: a new signaling paradigm? J Recept Signal Transduct Res 28:15–28 [DOI] [PubMed] [Google Scholar]
  32. Bossuyt J, Chang CW, Helmstadter K, Kunkel MT, Newton AC, Campbell KS, Martin JL, Bossuyt S, Robia SL, Bers DM. (2011) Spatiotemporally distinct protein kinase D activation in adult cardiomyocytes in response to phenylephrine and endothelin. J Biol Chem 286:33390–33400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Bossuyt J, Helmstadter K, Wu X, Clements-Jewery H, Haworth RS, Avkiran M, Martin JL, Pogwizd SM, Bers DM. (2008) Ca2+/calmodulin-dependent protein kinase IIdelta and protein kinase D overexpression reinforce the histone deacetylase 5 redistribution in heart failure. Circ Res 102:695–702 [DOI] [PubMed] [Google Scholar]
  34. Brahmadevara N, Shaw AM, MacDonald A. (2003) Evidence against beta 3-adrenoceptors or low affinity state of beta 1-adrenoceptors mediating relaxation in rat isolated aorta. Br J Pharmacol 138:99–106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Brahmadevara N, Shaw AM, MacDonald A. (2004) Alpha1-adrenoceptor antagonist properties of CGP 12177A and other beta-adrenoceptor ligands: evidence against beta(3)- or atypical beta-adrenoceptors in rat aorta. Br J Pharmacol 142:781–787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Braun AP, Fedida D, Giles WR. (1992) Activation of alpha 1-adrenoceptors modulates the inwardly rectifying potassium currents of mammalian atrial myocytes. Pflugers Arch 421:431–439 [DOI] [PubMed] [Google Scholar]
  37. Braz JC, Bueno OF, De Windt LJ, Molkentin JD. (2002) PKC alpha regulates the hypertrophic growth of cardiomyocytes through extracellular signal-regulated kinase1/2 (ERK1/2). J Cell Biol 156:905–919 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Braz JC, Gregory K, Pathak A, Zhao W, Sahin B, Klevitsky R, Kimball TF, Lorenz JN, Nairn AC, Liggett SB, et al. (2004) PKC-alpha regulates cardiac contractility and propensity toward heart failure. Nat Med 10:248–254 [DOI] [PubMed] [Google Scholar]
  39. Bristow MR. (2000) beta-adrenergic receptor blockade in chronic heart failure. Circulation 101:558–569 [DOI] [PubMed] [Google Scholar]
  40. Bristow MR, Feldman AM, Adams KF, Jr, Goldstein S. (2003) Selective versus nonselective beta-blockade for heart failure therapy: are there lessons to be learned from the COMET trial? J Card Fail 9:444–453 [DOI] [PubMed] [Google Scholar]
  41. Bristow MR, Ginsburg R, Minobe W, Cubicciotti RS, Sageman WS, Lurie K, Billingham ME, Harrison DC, Stinson EB. (1982) Decreased catecholamine sensitivity and beta-adrenergic-receptor density in failing human hearts. N Engl J Med 307:205–211 [DOI] [PubMed] [Google Scholar]
  42. Bristow MR, Krause-Steinrauf H, Nuzzo R, Liang CS, Lindenfeld J, Lowes BD, Hattler B, Abraham WT, Olson L, Krueger S, et al. (2004) Effect of baseline or changes in adrenergic activity on clinical outcomes in the beta-blocker evaluation of survival trial. Circulation 110:1437–1442 [DOI] [PubMed] [Google Scholar]
  43. Bristow MR, Minobe W, Rasmussen R, Hershberger RE, Hoffman BB. (1988) Alpha-1 adrenergic receptors in the nonfailing and failing human heart. J Pharmacol Exp Ther 247:1039–1045 [PubMed] [Google Scholar]
  44. Bueno OF, De Windt LJ, Tymitz KM, Witt SA, Kimball TR, Klevitsky R, Hewett TE, Jones SP, Lefer DJ, Peng CF, et al. (2000) The MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in transgenic mice. EMBO J 19:6341–6350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Buu NT, Hui R, Falardeau P. (1993) Norepinephrine in neonatal rat ventricular myocytes: association with the cell nucleus and binding to nuclear alpha 1- and beta-adrenergic receptors. J Mol Cell Cardiol 25:1037–1046 [DOI] [PubMed] [Google Scholar]
  46. Cabanes L, Costes F, Weber S, Regnard J, Benvenuti C, Castaigne A, Guerin F, Lockhart A. (1992) Improvement in exercise performance by inhalation of methoxamine in patients with impaired left ventricular function. N Engl J Med 326:1661–1665 [DOI] [PubMed] [Google Scholar]
  47. Caine M, Perlberg S, Meretyk S. (1978) A placebo-controlled double-blind study of the effect of phenoxybenzamine in benign prostatic obstruction. Br J Urol 50:551–554 [DOI] [PubMed] [Google Scholar]
  48. Caine M, Pfau A, Perlberg S. (1976) The use of alpha-adrenergic blockers in benign prostatic obstruction. Br J Urol 48:255–263 [DOI] [PubMed] [Google Scholar]
  49. Carnegie GK, Soughayer J, Smith FD, Pedroja BS, Zhang F, Diviani D, Bristow MR, Kunkel MT, Newton AC, Langeberg LK, et al. (2008) AKAP-Lbc mobilizes a cardiac hypertrophy signaling pathway. Mol Cell 32:169–179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Cavalli A, Lattion AL, Hummler E, Nenniger M, Pedrazzini T, Aubert JF, Michel MC, Yang M, Lembo G, Vecchione C, et al. (1997) Decreased blood pressure response in mice deficient of the alpha1b-adrenergic receptor. Proc Natl Acad Sci USA 94:11589–11594 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Chalothorn D, McCune DF, Edelmann SE, García-Cazarín ML, Tsujimoto G, Piascik MT. (2002) Differences in the cellular localization and agonist-mediated internalization properties of the alpha(1)-adrenoceptor subtypes. Mol Pharmacol 61:1008–1016 [DOI] [PubMed] [Google Scholar]
  52. Chalothorn D, McCune DF, Edelmann SE, Tobita K, Keller BB, Lasley RD, Perez DM, Tanoue A, Tsujimoto G, Post GR, et al. (2003) Differential cardiovascular regulatory activities of the alpha 1B- and alpha 1D-adrenoceptor subtypes. J Pharmacol Exp Ther 305:1045–1053 [DOI] [PubMed] [Google Scholar]
  53. Chan T, Dash R, Simpson PC. (2008) An alpha-1A-adrenergic receptor subtype agonist prevents cardiomyopathy without increasing blood pressure. abstract Circulation 118:S533 [Google Scholar]
  54. Chatterjee S, Biondi-Zoccai G, Abbate A, D’Ascenzo F, Castagno D, Van Tassell B, Mukherjee D, Lichstein E. (2013) Benefits of β blockers in patients with heart failure and reduced ejection fraction: network meta-analysis. BMJ 346:f55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Chaulet H, Lin F, Guo J, Owens WA, Michalicek J, Kesteven SH, Guan Z, Prall OW, Mearns BM, Feneley MP, et al. (2006) Sustained augmentation of cardiac alpha1A-adrenergic drive results in pathological remodeling with contractile dysfunction, progressive fibrosis and reactivation of matricellular protein genes. J Mol Cell Cardiol 40:540–552 [DOI] [PubMed] [Google Scholar]
  56. Chen HH, Mullett SJ, Stewart AF. (2004) Vgl-4, a novel member of the vestigial-like family of transcription cofactors, regulates alpha1-adrenergic activation of gene expression in cardiac myocytes. J Biol Chem 279:30800–30806 [DOI] [PubMed] [Google Scholar]
  57. Chen L, el-Sherif N, Boutjdir M. (1996) Alpha 1-adrenergic activation inhibits beta-adrenergic-stimulated unitary Ca2+ currents in cardiac ventricular myocytes. Circ Res 79:184–193 [DOI] [PubMed] [Google Scholar]
  58. Chen R, Mukhin YV, Garnovskaya MN, Thielen TE, Iijima Y, Huang C, Raymond JR, Ullian ME, Paul RV. (2000) A functional angiotensin II receptor-GFP fusion protein: evidence for agonist-dependent nuclear translocation. Am J Physiol Renal Physiol 279:F440–F448 [DOI] [PubMed] [Google Scholar]
  59. Chrysant SG. (2008) Angiotensin II receptor blockers in the treatment of the cardiovascular disease continuum. Clin Ther 30:2181–2190 [DOI] [PubMed] [Google Scholar]
  60. Chu C, Thai K, Park KW, Wang P, Makwana O, Lovett DH, Simpson PC, Baker AJ. (2013) Intraventricular and interventricular cellular heterogeneity of inotropic responses to α(1)-adrenergic stimulation. Am J Physiol Heart Circ Physiol 304:H946–H953 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. CIBIS-II (1999) The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet 353:9–13 [PubMed] [Google Scholar]
  62. Clark WA, Rudnick SJ, LaPres JJ, Andersen LC, LaPointe MC. (1993) Regulation of hypertrophy and atrophy in cultured adult heart cells. Circ Res 73:1163–1176 [DOI] [PubMed] [Google Scholar]
  63. Cleveland JC, Jr, Meldrum DR, Rowland RT, Cain BS, Meng X, Gamboni-Robertson F, Banerjee A, Harken AH. (1997) Ischemic preconditioning of human myocardium: protein kinase C mediates a permissive role for alpha 1-adrenoceptors. Am J Physiol 273:H902–H908 [DOI] [PubMed] [Google Scholar]
  64. Cleveland JC, Jr, Wollmering MM, Meldrum DR, Rowland RT, Rehring TF, Sheridan BC, Harken AH, Banerjee A. (1996) Ischemic preconditioning in human and rat ventricle. Am J Physiol 271:H1786–H1794 [DOI] [PubMed] [Google Scholar]
  65. Coats AJ. (1999) Heart Failure 99 — the MOXCON story. Int J Cardiol 71:109–111 [DOI] [PubMed] [Google Scholar]
  66. Cohn JN. (1993) The Vasodilator-Heart Failure Trials (V-HeFT). Mechanistic data from the VA Cooperative Studies. Introduction. Circulation 87(6, Suppl)VI1–VI4 [PubMed] [Google Scholar]
  67. Cohn JN, Archibald DG, Ziesche S, Franciosa JA, Harston WE, Tristani FE, Dunkman WB, Jacobs W, Francis GS, Flohr KH, et al. (1986) Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration Cooperative Study. N Engl J Med 314:1547–1552 [DOI] [PubMed] [Google Scholar]
  68. Cohn JN, Franciosa JA. (1977a) Vasodilator therapy of cardiac failure (second of two parts). N Engl J Med 297:254–258 [DOI] [PubMed] [Google Scholar]
  69. Cohn JN, Franciosa JA. (1977b) Vasodilator therapy of cardiac failure: (first of two parts). N Engl J Med 297:27–31 [DOI] [PubMed] [Google Scholar]
  70. Cohn JN, Levine TB, Olivari MT, Garberg V, Lura D, Francis GS, Simon AB, Rector T. (1984) Plasma norepinephrine as a guide to prognosis in patients with chronic congestive heart failure. N Engl J Med 311:819–823 [DOI] [PubMed] [Google Scholar]
  71. Cohn JN, Pfeffer MA, Rouleau J, Sharpe N, Swedberg K, Straub M, Wiltse C, Wright TJ, MOXCON Investigators (2003) Adverse mortality effect of central sympathetic inhibition with sustained-release moxonidine in patients with heart failure (MOXCON). Eur J Heart Fail 5:659–667 [DOI] [PubMed] [Google Scholar]
  72. Communal C, Singh K, Pimentel DR, Colucci WS. (1998) Norepinephrine stimulates apoptosis in adult rat ventricular myocytes by activation of the beta-adrenergic pathway. Circulation 98:1329–1334 [DOI] [PubMed] [Google Scholar]
  73. Communal C, Singh M, Menon B, Xie Z, Colucci WS, Singh K. (2003) beta1 integrins expression in adult rat ventricular myocytes and its role in the regulation of beta-adrenergic receptor-stimulated apoptosis. J Cell Biochem 89:381–388 [DOI] [PubMed] [Google Scholar]
  74. Cook A, Bono F, Jinek M, Conti E. (2007) Structural biology of nucleocytoplasmic transport. Annu Rev Biochem 76:647–671 [DOI] [PubMed] [Google Scholar]
  75. Cope JT, Mauney MC, Banks D, Binns OA, Moore CL, Rentz JJ, Shockey KS, King RC, Kron IL, Tribble CG. (1997) Intravenous phenylephrine preconditioning of cardiac grafts from non-heart-beating donors. Ann Thorac Surg 63:1664–1668 [DOI] [PubMed] [Google Scholar]
  76. Cotecchia S. (2010) The α1-adrenergic receptors: diversity of signaling networks and regulation. J Recept Signal Transduct Res 30:410–419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Curiel R, Pérez-González J, Brito N, Zerpa R, Téllez D, Cabrera J, Curiel C, Cubeddu L. (1989) Positive inotropic effects mediated by alpha 1 adrenoceptors in intact human subjects. J Cardiovasc Pharmacol 14:603–615 [DOI] [PubMed] [Google Scholar]
  78. Daly CJ, Milligan CM, Milligan G, Mackenzie JF, Mcgrath JC. (1998) Cellular localization and pharmacological characterization of functioning alpha-1 adrenoceptors by fluorescent ligand binding and image analysis reveals identical binding properties of clustered and diffuse populations of receptors. J Pharmacol Exp Ther 286:984–990 [PubMed] [Google Scholar]
  79. Dash R, Chung J, Chan T, Yamada M, Barral J, Nishimura D, Yang PC, Simpson PC. (2011) A molecular MRI probe to detect treatment of cardiac apoptosis in vivo. Magn Reson Med 66:1152–1162 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Davis BR, Cutler JA, Furberg CD, Wright JT, Farber MA, Felicetta JV, Stokes JD, ALLHAT Collaborative Research Group (2002) Relationship of antihypertensive treatment regimens and change in blood pressure to risk for heart failure in hypertensive patients randomly assigned to doxazosin or chlorthalidone: further analyses from the Antihypertensive and Lipid-Lowering treatment to prevent Heart Attack Trial. Ann Intern Med 137:313–320 [DOI] [PubMed] [Google Scholar]
  81. Debrus S, Rahbani L, Marttila M, Delorme B, Paradis P, Nemer M. (2005) The zinc finger-only protein Zfp260 is a novel cardiac regulator and a nuclear effector of alpha1-adrenergic signaling. Mol Cell Biol 25:8669–8682 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Dhaliwal AS, Habib G, Deswal A, Verduzco M, Souchek J, Ramasubbu K, Aguilar D, Ma TS, Jneid HM, Bolos M, et al. (2009) Impact of alpha 1-adrenergic antagonist use for benign prostatic hypertrophy on outcomes in patients with heart failure. Am J Cardiol 104:270–275 [DOI] [PubMed] [Google Scholar]
  83. Dono R, James D, Zeller R. (1998) A GR-motif functions in nuclear accumulation of the large FGF-2 isoforms and interferes with mitogenic signalling. Oncogene 16:2151–2158 [DOI] [PubMed] [Google Scholar]
  84. Dorn GW., 2nd (2005) Physiologic growth and pathologic genes in cardiac development and cardiomyopathy. Trends Cardiovasc Med 15:185–189 [DOI] [PubMed] [Google Scholar]
  85. Dorn GW, 2nd, Brown JH. (1999) Gq signaling in cardiac adaptation and maladaptation. Trends Cardiovasc Med 9:26–34 [DOI] [PubMed] [Google Scholar]
  86. Drake MT, Shenoy SK, Lefkowitz RJ. (2006) Trafficking of G protein-coupled receptors. Circ Res 99:570–582 [DOI] [PubMed] [Google Scholar]
  87. Du XJ, Fang L, Gao XM, Kiriazis H, Feng X, Hotchkin E, Finch AM, Chaulet H, Graham RM. (2004) Genetic enhancement of ventricular contractility protects against pressure-overload-induced cardiac dysfunction. J Mol Cell Cardiol 37:979–987 [DOI] [PubMed] [Google Scholar]
  88. Du XJ, Gao XM, Kiriazis H, Moore XL, Ming Z, Su Y, Finch AM, Hannan RA, Dart AM, Graham RM. (2006) Transgenic alpha1A-adrenergic activation limits post-infarct ventricular remodeling and dysfunction and improves survival. Cardiovasc Res 71:735–743 [DOI] [PubMed] [Google Scholar]
  89. Duan H, Wang J. (2010) Selective transport of monoamine neurotransmitters by human plasma membrane monoamine transporter and organic cation transporter 3. J Pharmacol Exp Ther 335:743–753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Eckhart AD, Duncan SJ, Penn RB, Benovic JL, Lefkowitz RJ, Koch WJ. (2000) Hybrid transgenic mice reveal in vivo specificity of G protein-coupled receptor kinases in the heart. Circ Res 86:43–50 [DOI] [PubMed] [Google Scholar]
  91. Endoh M, Blinks JR. (1988) Actions of sympathomimetic amines on the Ca2+ transients and contractions of rabbit myocardium: reciprocal changes in myofibrillar responsiveness to Ca2+ mediated through alpha- and beta-adrenoceptors. Circ Res 62:247–265 [DOI] [PubMed] [Google Scholar]
  92. Fawzy A, Vashi V, Chung M, Dias N, Gaffney M, Multicenter Study Group (1999) Clinical correlation of maximal urinary flow rate and plasma doxazosin concentrations in the treatment of benign prostatic hyperplasia. Urology 53:329–335 [DOI] [PubMed] [Google Scholar]
  93. Fedida D, Braun AP, Giles WR. (1991) Alpha 1-adrenoceptors reduce background K+ current in rabbit ventricular myocytes. J Physiol 441:673–684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Fedida D, Shimoni Y, Giles WR. (1989) A novel effect of norepinephrine on cardiac cells is mediated by alpha 1-adrenoceptors. Am J Physiol 256:H1500–H1504 [DOI] [PubMed] [Google Scholar]
  95. Fedida D, Shimoni Y, Giles WR. (1990) Alpha-adrenergic modulation of the transient outward current in rabbit atrial myocytes. J Physiol 423:257–277 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Filipeanu CM, Zhou F, Fugetta EK, Wu G. (2006) Differential regulation of the cell-surface targeting and function of beta- and alpha1-adrenergic receptors by Rab1 GTPase in cardiac myocytes. Mol Pharmacol 69:1571–1578 [DOI] [PubMed] [Google Scholar]
  97. Filtz TM, Grubb DR, McLeod-Dryden TJ, Luo J, Woodcock EA. (2009) Gq-initiated cardiomyocyte hypertrophy is mediated by phospholipase Cbeta1b. FASEB J 23:3564–3570 [DOI] [PubMed] [Google Scholar]
  98. Foo RS, Mani K, Kitsis RN. (2005) Death begets failure in the heart. J Clin Invest 115:565–571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Foody JM, Farrell MH, Krumholz HM. (2002) beta-Blocker therapy in heart failure: scientific review. JAMA 287:883–889 [DOI] [PubMed] [Google Scholar]
  100. Frey N, Olson EN. (2003) Cardiac hypertrophy: the good, the bad, and the ugly. Annu Rev Physiol 65:45–79 [DOI] [PubMed] [Google Scholar]
  101. Frishman WH, Kotob F. (1999) Alpha-adrenergic blocking drugs in clinical medicine. J Clin Pharmacol 39:7–16 [DOI] [PubMed] [Google Scholar]
  102. Fujita T, Toya Y, Iwatsubo K, Onda T, Kimura K, Umemura S, Ishikawa Y. (2001) Accumulation of molecules involved in alpha1-adrenergic signal within caveolae: caveolin expression and the development of cardiac hypertrophy. Cardiovasc Res 51:709–716 [DOI] [PubMed] [Google Scholar]
  103. Fuller SJ, Gaitanaki CJ, Sugden PH. (1990) Effects of catecholamines on protein synthesis in cardiac myocytes and perfused hearts isolated from adult rats. Stimulation of translation is mediated through the alpha 1-adrenoceptor. Biochem J 266:727–736 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Gambassi G, Spurgeon HA, Lakatta EG, Blank PS, Capogrossi MC. (1992) Different effects of alpha- and beta-adrenergic stimulation on cytosolic pH and myofilament responsiveness to Ca2+ in cardiac myocytes. Circ Res 71:870–882 [DOI] [PubMed] [Google Scholar]
  105. Gambassi G, Spurgeon HA, Ziman BD, Lakatta EG, Capogrossi MC. (1998) Opposing effects of alpha 1-adrenergic receptor subtypes on Ca2+ and pH homeostasis in rat cardiac myocytes. Am J Physiol 274:H1152–H1162 [DOI] [PubMed] [Google Scholar]
  106. Gao XM, Wang BH, Woodcock E, Du XJ. (2000) Expression of active alpha(1B)-adrenergic receptors in the heart does not alleviate ischemic reperfusion injury. J Mol Cell Cardiol 32:1679–1686 [DOI] [PubMed] [Google Scholar]
  107. Gaughan JP, Hefner CA, Houser SR. (1998) Electrophysiological properties of neonatal rat ventricular myocytes with alpha1-adrenergic-induced hypertrophy. Am J Physiol 275:H577–H590 [DOI] [PubMed] [Google Scholar]
  108. Glembotski CC. (2013) Classic studies of cultured cardiac myocyte hypertrophy: interview with a transformer. Circ Res 113:1112–1116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Gobeil F, Jr, Dumont I, Marrache AM, Vazquez-Tello A, Bernier SG, Abran D, Hou X, Beauchamp MH, Quiniou C, Bouayad A, et al. (2002) Regulation of eNOS expression in brain endothelial cells by perinuclear EP(3) receptors. Circ Res 90:682–689 [DOI] [PubMed] [Google Scholar]
  110. Gobeil F, Fortier A, Zhu T, Bossolasco M, Leduc M, Grandbois M, Heveker N, Bkaily G, Chemtob S, Barbaz D. (2006) G-protein-coupled receptors signalling at the cell nucleus: an emerging paradigm. Can J Physiol Pharmacol 84:287–297 [DOI] [PubMed] [Google Scholar]
  111. González-Juanatey JR, Iglesias MJ, Alcaide C, Piñeiro R, Lago F. (2003) Doxazosin induces apoptosis in cardiomyocytes cultured in vitro by a mechanism that is independent of alpha1-adrenergic blockade. Circulation 107:127–131 [DOI] [PubMed] [Google Scholar]
  112. Graham RM, Perez DM, Hwa J, Piascik MT. (1996) Alpha 1-adrenergic receptor subtypes. Molecular structure, function, and signaling. Circ Res 78:737–749 [DOI] [PubMed] [Google Scholar]
  113. Gray MO, Long CS, Kalinyak JE, Li HT, Karliner JS. (1998) Angiotensin II stimulates cardiac myocyte hypertrophy via paracrine release of TGF-beta 1 and endothelin-1 from fibroblasts. Cardiovasc Res 40:352–363 [DOI] [PubMed] [Google Scholar]
  114. Grubb DR, Vasilevski O, Huynh H, Woodcock EA. (2008) The extreme C-terminal region of phospholipase Cbeta1 determines subcellular localization and function; the “b” splice variant mediates alpha1-adrenergic receptor responses in cardiomyocytes. FASEB J 22:2768–2774 [DOI] [PubMed] [Google Scholar]
  115. Grupp IL, Lorenz JN, Walsh RA, Boivin GP, Rindt H. (1998) Overexpression of alpha1B-adrenergic receptor induces left ventricular dysfunction in the absence of hypertrophy. Am J Physiol 275:H1338–H1350 [DOI] [PubMed] [Google Scholar]
  116. Guerra S, Leri A, Wang X, Finato N, Di Loreto C, Beltrami CA, Kajstura J, Anversa P. (1999) Myocyte death in the failing human heart is gender dependent. Circ Res 85:856–866 [DOI] [PubMed] [Google Scholar]
  117. Guo J, Gertsberg Z, Ozgen N, Steinberg SF. (2009) p66Shc links alpha1-adrenergic receptors to a reactive oxygen species-dependent AKT-FOXO3A phosphorylation pathway in cardiomyocytes. Circ Res 104:660–669 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Harada K, Sugaya T, Murakami K, Yazaki Y, Komuro I. (1999) Angiotensin II type 1A receptor knockout mice display less left ventricular remodeling and improved survival after myocardial infarction. see comments Circulation 100:2093–2099 [DOI] [PubMed] [Google Scholar]
  119. Harrison BC, Kim MS, van Rooij E, Plato CF, Papst PJ, Vega RB, McAnally JA, Richardson JA, Bassel-Duby R, Olson EN, et al. (2006) Regulation of cardiac stress signaling by protein kinase d1. Mol Cell Biol 26:3875–3888 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Hartmann M, Stumpe T, Schrader J. (1995) alpha 1-Adrenoceptor stimulation inhibits the isoproterenol-induced effects on myocardial contractility and protein phosphorylation. Eur J Pharmacol 287:57–64 [DOI] [PubMed] [Google Scholar]
  121. Haworth RS, Goss MW, Rozengurt E, Avkiran M. (2000) Expression and activity of protein kinase D/protein kinase C mu in myocardium: evidence for alpha1-adrenergic receptor- and protein kinase C-mediated regulation. J Mol Cell Cardiol 32:1013–1023 [DOI] [PubMed] [Google Scholar]
  122. Hein P, Michel MC. (2007) Signal transduction and regulation: are all alpha1-adrenergic receptor subtypes created equal? Biochem Pharmacol 73:1097–1106 [DOI] [PubMed] [Google Scholar]
  123. Hennenberg M, Schlenker B, Roosen A, Strittmatter F, Walther S, Stief C, Gratzke C. (2011) β-arrestin-2 is expressed in human prostate smooth muscle and a binding partner of α1A-adrenoceptors. World J Urol 29:157–163 [DOI] [PubMed] [Google Scholar]
  124. Henrich CJ, Simpson PC. (1988) Differential acute and chronic response of protein kinase C in cultured neonatal rat heart myocytes to alpha 1-adrenergic and phorbol ester stimulation. J Mol Cell Cardiol 20:1081–1085 [DOI] [PubMed] [Google Scholar]
  125. Hirano S, Kusakari Y, O-Uchi J, Morimoto S, Kawai M, Hongo K, Kurihara S. (2006) Intracellular mechanism of the negative inotropic effect induced by alpha1-adrenoceptor stimulation in mouse myocardium. J Physiol Sci 56:297–304 [DOI] [PubMed] [Google Scholar]
  126. Hock R, Scheer U, Bustin M. (1998) Chromosomal proteins HMG-14 and HMG-17 are released from mitotic chromosomes and imported into the nucleus by active transport. J Cell Biol 143:1427–1436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Hosoda C, Koshimizu TA, Tanoue A, Nasa Y, Oikawa R, Tomabechi T, Fukuda S, Shinoura H, Oshikawa S, Takeo S, et al. (2005) Two alpha1-adrenergic receptor subtypes regulating the vasopressor response have differential roles in blood pressure regulation. Mol Pharmacol 67:912–922 [DOI] [PubMed] [Google Scholar]
  128. Hryniewicz K, Androne AS, Hudaihed A, Katz SD. (2003) Comparative effects of carvedilol and metoprolol on regional vascular responses to adrenergic stimuli in normal subjects and patients with chronic heart failure. Circulation 108:971–976 [DOI] [PubMed] [Google Scholar]
  129. Hu K, Nattel S. (1995) Mechanisms of ischemic preconditioning in rat hearts. Involvement of alpha 1B-adrenoceptors, pertussis toxin-sensitive G proteins, and protein kinase C. Circulation 92:2259–2265 [DOI] [PubMed] [Google Scholar]
  130. Huang Y, Wright CD, Kobayashi S, Healy CL, Elgethun M, Cypher A, Liang Q, O'Connell TD. (2008) GATA4 is a survival factor in adult cardiac myocytes but is not required for {alpha}1A-adrenergic receptor survival signaling. Am J Physiol 295:H699–H707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Huang Y, Wright CD, Merkwan CL, Baye NL, Liang Q, Simpson PC, O’Connell TD. (2007) An alpha1A-adrenergic-extracellular signal-regulated kinase survival signaling pathway in cardiac myocytes. Circulation 115:763–772 [DOI] [PubMed] [Google Scholar]
  132. Hunt SA, Abraham WT, Chin MH, Feldman AM, Francis GS, Ganiats TG, Jessup M, Konstam MA, Mancini DM, Michl K, et al. American College of Cardiology. American Heart Association Task Force on Practice Guidelines. American College of Chest Physicians. International Society for Heart and Lung Transplantation. Heart Rhythm Society (2005) ACC/AHA 2005 Guideline Update for the Diagnosis and Management of Chronic Heart Failure in the Adult: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure): developed in collaboration with the American College of Chest Physicians and the International Society for Heart and Lung Transplantation: endorsed by the Heart Rhythm Society. Circulation 112:e154–e235 [DOI] [PubMed] [Google Scholar]
  133. Hwang KC, Gray CD, Sweet WE, Moravec CS, Im MJ. (1996) Alpha 1-adrenergic receptor coupling with Gh in the failing human heart. Circulation 94:718–726 [DOI] [PubMed] [Google Scholar]
  134. Iaccarino G, Keys JR, Rapacciuolo A, Shotwell KF, Lefkowitz RJ, Rockman HA, Koch WJ. (2001) Regulation of myocardial betaARK1 expression in catecholamine-induced cardiac hypertrophy in transgenic mice overexpressing alpha1B-adrenergic receptors. J Am Coll Cardiol 38:534–540 [DOI] [PubMed] [Google Scholar]
  135. Ichishima K, Yamamoto S, Iwamoto T, Ehara T. (2010) alpha-Adrenoceptor-mediated depletion of phosphatidylinositol 4, 5-bisphosphate inhibits activation of volume-regulated anion channels in mouse ventricular myocytes. Br J Pharmacol 161:193–206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Ikeda U, Tsuruya Y, Yaginuma T. (1991) Alpha 1-adrenergic stimulation is coupled to cardiac myocyte hypertrophy. Am J Physiol 260:H953–H956 [DOI] [PubMed] [Google Scholar]
  137. Imani A, Faghihi M, Sadr SS, Keshavarz M, Niaraki SS. (2008) Noradrenaline reduces ischemia-induced arrhythmia in anesthetized rats: involvement of alpha1-adrenoceptors and mitochondrial K ATP channels. J Cardiovasc Electrophysiol 19:309–315 [DOI] [PubMed] [Google Scholar]
  138. Iwai-Kanai E, Hasegawa K, Araki M, Kakita T, Morimoto T, Sasayama S. (1999) alpha- and beta-adrenergic pathways differentially regulate cell type-specific apoptosis in rat cardiac myocytes. Circulation 100:305–311 [DOI] [PubMed] [Google Scholar]
  139. Jensen BC, O’Connell TD, Simpson PC. (2011) Alpha-1-adrenergic receptors: targets for agonist drugs to treat heart failure. J Mol Cell Cardiol 51:518–528 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Jensen BC, Swigart PM, De Marco T, Hoopes C, Simpson PC. (2009a) alpha1-Adrenergic receptor subtypes in nonfailing and failing human myocardium. Circ Heart Fail 2:654–663 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Jensen BC, Swigart PM, Laden ME, DeMarco T, Hoopes C, Simpson PC. (2009b) The alpha-1D Is the predominant alpha-1-adrenergic receptor subtype in human epicardial coronary arteries. J Am Coll Cardiol 54:1137–1145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Jensen BC, Swigart PM, Montgomery MD, Simpson PC. (2010) Functional alpha-1B adrenergic receptors on human epicardial coronary artery endothelial cells. Naunyn Schmiedebergs Arch Pharmacol 382:475–482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Jensen BC, Swigart PM, Simpson PC. (2009c) Ten commercial antibodies for alpha-1-adrenergic receptor subtypes are nonspecific. Naunyn Schmiedebergs Arch Pharmacol 379:409–412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Jin Y, Sheikh F, Detillieux KA, Cattini PA. (2000) Role for early growth response-1 protein in alpha(1)-adrenergic stimulation of fibroblast growth factor-2 promoter activity in cardiac myocytes. Mol Pharmacol 57:984–990 [PubMed] [Google Scholar]
  145. Kariya K, Farrance IK, Simpson PC. (1993) Transcriptional enhancer factor-1 in cardiac myocytes interacts with an alpha 1-adrenergic- and beta-protein kinase C-inducible element in the rat beta-myosin heavy chain promoter. J Biol Chem 268:26658–26662 [PubMed] [Google Scholar]
  146. Kariya K, Karns LR, Simpson PC. (1991) Expression of a constitutively activated mutant of the beta-isozyme of protein kinase C in cardiac myocytes stimulates the promoter of the beta-myosin heavy chain isogene. J Biol Chem 266:10023–10026 [PubMed] [Google Scholar]
  147. Kariya K, Karns LR, Simpson PC. (1994) An enhancer core element mediates stimulation of the rat beta-myosin heavy chain promoter by an alpha 1-adrenergic agonist and activated beta-protein kinase C in hypertrophy of cardiac myocytes. J Biol Chem 269:3775–3782 [PubMed] [Google Scholar]
  148. Karns LR, Kariya K, Simpson PC. (1995) M-CAT, CArG, and Sp1 elements are required for alpha 1-adrenergic induction of the skeletal alpha-actin promoter during cardiac myocyte hypertrophy. Transcriptional enhancer factor-1 and protein kinase C as conserved transducers of the fetal program in cardiac growth. J Biol Chem 270:410–417 [DOI] [PubMed] [Google Scholar]
  149. Kim NN, Villarreal FJ, Printz MP, Lee AA, Dillmann WH. (1995) Trophic effects of angiotensin II on neonatal rat cardiac myocytes are mediated by cardiac fibroblasts. Am J Physiol 269:E426–E437 [DOI] [PubMed] [Google Scholar]
  150. King BD, Sack D, Kichuk MR, Hintze TH. (1987) Absence of hypertension despite chronic marked elevations in plasma norepinephrine in conscious dogs. Hypertension 9:582–590 [DOI] [PubMed] [Google Scholar]
  151. King MC, Lusk CP, Blobel G. (2006) Karyopherin-mediated import of integral inner nuclear membrane proteins. Nature 442:1003–1007 [DOI] [PubMed] [Google Scholar]
  152. Kitakaze M, Hori M, Morioka T, Minamino T, Takashima S, Sato H, Shinozaki Y, Chujo M, Mori H, Inoue M, et al. (1994) Alpha 1-adrenoceptor activation mediates the infarct size-limiting effect of ischemic preconditioning through augmentation of 5′-nucleotidase activity. J Clin Invest 93:2197–2205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Kitakaze M, Hori M, Sato H, Iwakura K, Gotoh K, Inoue M, Kitabatake A, Kamada T. (1991) Beneficial effects of alpha 1-adrenoceptor activity on myocardial stunning in dogs. Circ Res 68:1322–1339 [DOI] [PubMed] [Google Scholar]
  154. Kitakaze M, Hori M, Tamai J, Iwakura K, Koretsune Y, Kagiya T, Iwai K, Kitabatake A, Inoue M, Kamada T. (1987) Alpha 1-adrenoceptor activity regulates release of adenosine from the ischemic myocardium in dogs. Circ Res 60:631–639 [DOI] [PubMed] [Google Scholar]
  155. Knowlton KU, Baracchini E, Ross RS, Harris AN, Henderson SA, Evans SM, Glembotski CC, Chien KR. (1991) Co-regulation of the atrial natriuretic factor and cardiac myosin light chain-2 genes during alpha-adrenergic stimulation of neonatal rat ventricular cells. Identification of cis sequences within an embryonic and a constitutive contractile protein gene which mediate inducible expression. J Biol Chem 266:7759–7768 [PubMed] [Google Scholar]
  156. Knowlton KU, Michel MC, Itani M, Shubeita HE, Ishihara K, Brown JH, Chien KR. (1993) The alpha 1A-adrenergic receptor subtype mediates biochemical, molecular, and morphologic features of cultured myocardial cell hypertrophy. J Biol Chem 268:15374–15380 [PubMed] [Google Scholar]
  157. Koshimizu TA, Tsujimoto G, Hirasawa A, Kitagawa Y, Tanoue A. (2004) Carvedilol selectively inhibits oscillatory intracellular calcium changes evoked by human alpha1D- and alpha1B-adrenergic receptors. Cardiovasc Res 63:662–672 [DOI] [PubMed] [Google Scholar]
  158. Kostin S, Pool L, Elsässer A, Hein S, Drexler HC, Arnon E, Hayakawa Y, Zimmermann R, Bauer E, Klövekorn WP, et al. (2003) Myocytes die by multiple mechanisms in failing human hearts. Circ Res 92:715–724 [DOI] [PubMed] [Google Scholar]
  159. Kubo T, Azevedo ER, Newton GE, Parker JD, Floras JS. (2001) Lack of evidence for peripheral alpha(1)- adrenoceptor blockade during long-term treatment of heart failure with carvedilol. J Am Coll Cardiol 38:1463–1469 [DOI] [PubMed] [Google Scholar]
  160. Laks MM, Morady F, Swan HJ. (1973) Myocardial hypertrophy produced by chronic infusion of subhypertensive doses of norepinephrine in the dog. Chest 64:75–78 [DOI] [PubMed] [Google Scholar]
  161. Landzberg JS, Parker JD, Gauthier DF, Colucci WS. (1991) Effects of myocardial alpha 1-adrenergic receptor stimulation and blockade on contractility in humans. Circulation 84:1608–1614 [DOI] [PubMed] [Google Scholar]
  162. Lanzafame AA, Turnbull L, Amiramahdi F, Arthur JF, Huynh H, Woodcock EA. (2006) Inositol phospholipids localized to caveolae in rat heart are regulated by alpha1-adrenergic receptors and by ischemia-reperfusion. Am J Physiol Heart Circ Physiol 290:H2059–H2065 [DOI] [PubMed] [Google Scholar]
  163. Lee DK, Lança AJ, Cheng R, Nguyen T, Ji XD, Gobeil F, Jr, Chemtob S, George SR, O’Dowd BF. (2004) Agonist-independent nuclear localization of the Apelin, angiotensin AT1, and bradykinin B2 receptors. J Biol Chem 279:7901–7908 [DOI] [PubMed] [Google Scholar]
  164. Lemire I, Ducharme A, Tardif JC, Poulin F, Jones LR, Allen BG, Hébert TE, Rindt H. (2001) Cardiac-directed overexpression of wild-type alpha1B-adrenergic receptor induces dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 281:H931–H938 [DOI] [PubMed] [Google Scholar]
  165. Levin MC, Marullo S, Muntaner O, Andersson B, Magnusson Y. (2002) The myocardium-protective Gly-49 variant of the beta 1-adrenergic receptor exhibits constitutive activity and increased desensitization and down-regulation. J Biol Chem 277:30429–30435 [DOI] [PubMed] [Google Scholar]
  166. Levy D, Garrison RJ, Savage DD, Kannel WB, Castelli WP. (1990) Prognostic implications of echocardiographically determined left ventricular mass in the Framingham Heart Study. N Engl J Med 322:1561–1566 [DOI] [PubMed] [Google Scholar]
  167. Li Y, Zhang H, Liao W, Song Y, Ma X, Chen C, Lu Z, Li Z, Zhang Y. (2011) Transactivated EGFR mediates α₁-AR-induced STAT3 activation and cardiac hypertrophy. Am J Physiol Heart Circ Physiol 301:H1941–H1951 [DOI] [PubMed] [Google Scholar]
  168. Liang Q, De Windt LJ, Witt SA, Kimball TR, Markham BE, Molkentin JD. (2001a) The transcription factors GATA4 and GATA6 regulate cardiomyocyte hypertrophy in vitro and in vivo. J Biol Chem 276:30245–30253 [DOI] [PubMed] [Google Scholar]
  169. Liang Q, Wiese RJ, Bueno OF, Dai YS, Markham BE, Molkentin JD. (2001b) The transcription factor GATA4 is activated by extracellular signal-regulated kinase 1- and 2-mediated phosphorylation of serine 105 in cardiomyocytes. Mol Cell Biol 21:7460–7469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Lin F, Owens WA, Chen S, Stevens ME, Kesteven S, Arthur JF, Woodcock EA, Feneley MP, Graham RM. (2001) Targeted alpha(1A)-adrenergic receptor overexpression induces enhanced cardiac contractility but not hypertrophy. Circ Res 89:343–350 [DOI] [PubMed] [Google Scholar]
  171. Lips DJ, Bueno OF, Wilkins BJ, Purcell NH, Kaiser RA, Lorenz JN, Voisin L, Saba-El-Leil MK, Meloche S, Pouysségur J, et al. (2004) MEK1-ERK2 signaling pathway protects myocardium from ischemic injury in vivo. Circulation 109:1938–1941 [DOI] [PubMed] [Google Scholar]
  172. Litwin SE, Vatner DE, Morgan JP. (1995) Inotropic effects of alpha 1-adrenergic agonists in myocardium from rats with postinfarction heart failure. Am J Physiol 269:H1553–H1563 [DOI] [PubMed] [Google Scholar]
  173. Liu Y, Contreras M, Shen T, Randall WR, Schneider MF. (2009) Alpha-adrenergic signalling activates protein kinase D and causes nuclear efflux of the transcriptional repressor HDAC5 in cultured adult mouse soleus skeletal muscle fibres. J Physiol 587:1101–1115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Lohse MJ, Engelhardt S, Eschenhagen T. (2003) What is the role of beta-adrenergic signaling in heart failure? Circ Res 93:896–906 [DOI] [PubMed] [Google Scholar]
  175. Long CS, Ordahl CP, Simpson PC. (1989) Alpha 1-adrenergic receptor stimulation of sarcomeric actin isogene transcription in hypertrophy of cultured rat heart muscle cells. J Clin Invest 83:1078–1082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Lopez I, Mak EC, Ding J, Hamm HE, Lomasney JW. (2001) A novel bifunctional phospholipase c that is regulated by Galpha 12 and stimulates the Ras/mitogen-activated protein kinase pathway. J Biol Chem 276:2758–2765 [DOI] [PubMed] [Google Scholar]
  177. López JE, Myagmar BE, Swigart PM, Montgomery MD, Haynam S, Bigos M, Rodrigo MC, Simpson PC. (2011) β-myosin heavy chain is induced by pressure overload in a minor subpopulation of smaller mouse cardiac myocytes. Circ Res 109:629–638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Loubani M, Galiñanes M. (2001) alpha1-Adrenoceptors during simulated ischemia and reoxygenation of the human myocardium: effect of the dose and time of administration. J Thorac Cardiovasc Surg 122:103–112 [DOI] [PubMed] [Google Scholar]
  179. Loubani M, Galiñanes M. (2002) Pharmacological and ischemic preconditioning of the human myocardium: mitoK(ATP) channels are upstream and p38MAPK is downstream of PKC. BMC Physiol 2:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Lu D, Yang H, Shaw G, Raizada MK. (1998) Angiotensin II-induced nuclear targeting of the angiotensin type 1 (AT1) receptor in brain neurons. Endocrinology 139:365–375 [DOI] [PubMed] [Google Scholar]
  181. Lund-Johansen P, Omvik P. (1991) Acute and chronic hemodynamic effects of drugs with different actions on adrenergic receptors: a comparison between alpha blockers and different types of beta blockers with and without vasodilating effect. Cardiovasc Drugs Ther 5:605–615 [DOI] [PubMed] [Google Scholar]
  182. Luo DL, Gao J, Fan LL, Tang Y, Zhang YY, Han QD. (2007) Receptor subtype involved in alpha 1-adrenergic receptor-mediated Ca2+ signaling in cardiomyocytes. Acta Pharmacol Sin 28:968–974 [DOI] [PubMed] [Google Scholar]
  183. Lusk CP, Blobel G, King MC. (2007) Highway to the inner nuclear membrane: rules for the road. Nat Rev Mol Cell Biol 8:414–420 [DOI] [PubMed] [Google Scholar]
  184. MacGowan GA, Rager J, Shroff SG, Mathier MA. (2005) In vivo alpha-adrenergic responses and troponin I phosphorylation: anesthesia interactions. J Appl Physiol (1985) 98:1163–1170 [DOI] [PubMed] [Google Scholar]
  185. Mackenzie JF, Daly CJ, Pediani JD, McGrath JC. (2000) Quantitative imaging in live human cells reveals intracellular alpha(1)-adrenoceptor ligand-binding sites. J Pharmacol Exp Ther 294:434–443 [PubMed] [Google Scholar]
  186. Mann DL, Kent RL, Parsons B, Cooper G., 4th (1992) Adrenergic effects on the biology of the adult mammalian cardiocyte. Circulation 85:790–804 [DOI] [PubMed] [Google Scholar]
  187. Marino TA, Cassidy M, Marino DR, Carson NL, Houser S. (1991) Norepinephrine-induced cardiac hypertrophy of the cat heart. Anat Rec 229:505–510 [DOI] [PubMed] [Google Scholar]
  188. Markou T, Hadzopoulou-Cladaras M, Lazou A. (2004) Phenylephrine induces activation of CREB in adult rat cardiac myocytes through MSK1 and PKA signaling pathways. J Mol Cell Cardiol 37:1001–1011 [DOI] [PubMed] [Google Scholar]
  189. Marrache AM, Gobeil F, Jr, Bernier SG, Stankova J, Rola-Pleszczynski M, Choufani S, Bkaily G, Bourdeau A, Sirois MG, Vazquez-Tello A, et al. (2002) Proinflammatory gene induction by platelet-activating factor mediated via its cognate nuclear receptor. J Immunol 169:6474–6481 [DOI] [PubMed] [Google Scholar]
  190. McCloskey DT, Turnbull L, Swigart P, O’Connell TD, Simpson PC, Baker AJ. (2003) Abnormal myocardial contraction in alpha(1A)- and alpha(1B)-adrenoceptor double-knockout mice. J Mol Cell Cardiol 35:1207–1216 [DOI] [PubMed] [Google Scholar]
  191. McLean BG, Lee KS, Simpson PC, Farrance IK. (2003) Basal and alpha1-adrenergic-induced activity of minimal rat betaMHC promoters in cardiac myocytes requires multiple TEF-1 but not NFAT binding sites. J Mol Cell Cardiol 35:461–471 [DOI] [PubMed] [Google Scholar]
  192. McMurray JJ, Teerlink JR, Cotter G, Bourge RC, Cleland JG, Jondeau G, Krum H, Metra M, O’Connor CM, Parker JD, et al. VERITAS Investigators (2007) Effects of tezosentan on symptoms and clinical outcomes in patients with acute heart failure: the VERITAS randomized controlled trials. JAMA 298:2009–2019 [DOI] [PubMed] [Google Scholar]
  193. McWhinney CD, Hansen C, Robishaw JD. (2000) Alpha-1 adrenergic signaling in a cardiac murine atrial myocyte (HL-1) cell line. Mol Cell Biochem 214:111–119 [DOI] [PubMed] [Google Scholar]
  194. Meldrum DR, Cleveland JC, Jr, Rowland RT, Banerjee A, Harken AH, Meng X. (1997) Early and delayed preconditioning: differential mechanisms and additive protection. Am J Physiol 273:H725–H733 [DOI] [PubMed] [Google Scholar]
  195. Melien O, Sandnes D, Johansen EJ, Christoffersen T. (2000) Effects of pertussis toxin on extracellular signal-regulated kinase activation in hepatocytes by hormones and receptor-independent agents: evidence suggesting a stimulatory role of G(i) proteins at a level distal to receptor coupling. J Cell Physiol 184:27–36 [DOI] [PubMed] [Google Scholar]
  196. Meng X, Brown JM, Ao L, Banerjee A, Harken AH. (1996a) Norepinephrine induces cardiac heat shock protein 70 and delayed cardioprotection in the rat through alpha 1 adrenoceptors. Cardiovasc Res 32:374–383 [DOI] [PubMed] [Google Scholar]
  197. Meng X, Cleveland JC, Jr, Rowland RT, Mitchell MB, Brown JM, Banerjee A, Harken AH. (1996b) Norepinephrine-induced sustained myocardial adaptation to ischemia is dependent on alpha 1-adrenoceptors and protein synthesis. J Mol Cell Cardiol 28:2017–2025 [DOI] [PubMed] [Google Scholar]
  198. Meng X, Shames BD, Pulido EJ, Meldrum DR, Ao L, Joo KS, Harken AH, Banerjee A. (1999) Adrenergic induction of bimodal myocardial protection: signal transduction and cardiac gene reprogramming. Am J Physiol 276:R1525–R1533 [DOI] [PubMed] [Google Scholar]
  199. MERIT (1999) Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet 353:2001–2007 [PubMed] [Google Scholar]
  200. Messerli FH. (2000) Implications of discontinuation of doxazosin arm of ALLHAT. Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial. Lancet 355:863–864 [DOI] [PubMed] [Google Scholar]
  201. Messerli FH. (2001) Doxazosin and congestive heart failure. J Am Coll Cardiol 38:1295–1296 [DOI] [PubMed] [Google Scholar]
  202. Methven L, Simpson PC, McGrath JC. (2009) Alpha1A/B-knockout mice explain the native alpha1D-adrenoceptor’s role in vasoconstriction and show that its location is independent of the other alpha1-subtypes. Br J Pharmacol 158:1663–1675 [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Michel MC. (2010) The forefront for novel therapeutic agents based on the pathophysiology of lower urinary tract dysfunction: alpha-blockers in the treatment of male voiding dysfunction - how do they work and why do they differ in tolerability? J Pharmacol Sci 112:151–157 [DOI] [PubMed] [Google Scholar]
  204. Michel MC, Hanft G, Gross G. (1994) Radioligand binding studies of alpha 1-adrenoceptor subtypes in rat heart. Br J Pharmacol 111:533–538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Michel MC, Wieland T, Tsujimoto G. (2009) How reliable are G-protein-coupled receptor antibodies? Naunyn Schmiedebergs Arch Pharmacol 379:385–388 [DOI] [PubMed] [Google Scholar]
  206. Michelotti GA, Price DT, Schwinn DA. (2000) Alpha 1-adrenergic receptor regulation: basic science and clinical implications. Pharmacol Ther 88:281–309 [DOI] [PubMed] [Google Scholar]
  207. Milano CA, Dolber PC, Rockman HA, Bond RA, Venable ME, Allen LF, Lefkowitz RJ. (1994) Myocardial expression of a constitutively active alpha 1B-adrenergic receptor in transgenic mice induces cardiac hypertrophy. Proc Natl Acad Sci USA 91:10109–10113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Mitchell MB, Meng X, Ao L, Brown JM, Harken AH, Banerjee A. (1995) Preconditioning of isolated rat heart is mediated by protein kinase C. Circ Res 76:73–81 [DOI] [PubMed] [Google Scholar]
  209. Modesti PA, Vanni S, Paniccia R, Bandinelli B, Bertolozzi I, Polidori G, Sani G, Neri Serneri GG. (1999) Characterization of endothelin-1 receptor subtypes in isolated human cardiomyocytes. J Cardiovasc Pharmacol 34:333–339 [DOI] [PubMed] [Google Scholar]
  210. Mohl MC, Iismaa SE, Xiao XH, Friedrich O, Wagner S, Nikolova-Krstevski V, Wu J, Yu ZY, Feneley M, Fatkin D, et al. (2011) Regulation of murine cardiac contractility by activation of α(1A)-adrenergic receptor-operated Ca(2+) entry. Cardiovasc Res 91:310–319 [DOI] [PubMed] [Google Scholar]
  211. Morimoto T, Hasegawa K, Kaburagi S, Kakita T, Wada H, Yanazume T, Sasayama S. (2000) Phosphorylation of GATA-4 is involved in alpha 1-adrenergic agonist-responsive transcription of the endothelin-1 gene in cardiac myocytes. J Biol Chem 275:13721–13726 [DOI] [PubMed] [Google Scholar]
  212. Morris JB, Huynh H, Vasilevski O, Woodcock EA. (2006) Alpha1-adrenergic receptor signaling is localized to caveolae in neonatal rat cardiomyocytes. J Mol Cell Cardiol 41:17–25 [DOI] [PubMed] [Google Scholar]
  213. Morris JB, Pham TM, Kenney B, Sheppard KE, Woodcock EA. (2004) UTP transactivates epidermal growth factor receptors and promotes cardiomyocyte hypertrophy despite inhibiting transcription of the hypertrophic marker gene, atrial natriuretic peptide. J Biol Chem 279:8740–8746 [DOI] [PubMed] [Google Scholar]
  214. Mylona P, Cleland JG. (1999) Update of REACH-1 and MERIT-HF clinical trials in heart failure. Cardio.net Editorial Team. Eur J Heart Fail 1:197–200 [DOI] [PubMed] [Google Scholar]
  215. Naga Prasad SV, Nienaber J, Rockman HA. (2001) Beta-adrenergic axis and heart disease. Trends Genet 17:S44–S49 [DOI] [PubMed] [Google Scholar]
  216. Nakayama H, Chen X, Baines CP, Klevitsky R, Zhang X, Zhang H, Jaleel N, Chua BH, Hewett TE, Robbins J, et al. (2007) Ca2+- and mitochondrial-dependent cardiomyocyte necrosis as a primary mediator of heart failure. J Clin Invest 117:2431–2444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Nemchenko A, Chiong M, Turer A, Lavandero S, Hill JA. (2011) Autophagy as a therapeutic target in cardiovascular disease. J Mol Cell Cardiol 51:584–593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Nishida K, Otsu K. (2008) Cell death in heart failure. Circ J 72 Suppl A:A17–21 [DOI] [PubMed] [Google Scholar]
  219. Node K, Kitakaze M, Sato H, Minamino T, Komamura K, Shinozaki Y, Mori H, Hori M. (1997) Role of intracellular Ca2+ in activation of protein kinase C during ischemic preconditioning. Circulation 96:1257–1265 [DOI] [PubMed] [Google Scholar]
  220. Noguchi H, Muraoka R, Kigoshi S, Muramatsu I. (1995) Pharmacological characterization of alpha 1-adrenoceptor subtypes in rat heart: a binding study. Br J Pharmacol 114:1026–1030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. O’Connell TD, Ishizaka S, Nakamura A, Swigart PM, Rodrigo MC, Simpson GL, Cotecchia S, Rokosh DG, Grossman W, Foster E, et al. (2003) The alpha(1A/C)- and alpha(1B)-adrenergic receptors are required for physiological cardiac hypertrophy in the double-knockout mouse. J Clin Invest 111:1783–1791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. O’Connell TD, Rokosh DG, Simpson PC. (2001) Cloning and characterization of the mouse alpha1C/A-adrenergic receptor gene and analysis of an alpha1C promoter in cardiac myocytes: role of an MCAT element that binds transcriptional enhancer factor-1 (TEF-1). Mol Pharmacol 59:1225–1234 [DOI] [PubMed] [Google Scholar]
  223. O’Connell TD, Swigart PM, Rodrigo MC, Ishizaka S, Joho S, Turnbull L, Tecott LH, Baker AJ, Foster E, Grossman W, et al. (2006) Alpha1-adrenergic receptors prevent a maladaptive cardiac response to pressure overload. J Clin Invest 116:1005–1015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. O’Malley KL, Jong YJ, Gonchar Y, Burkhalter A, Romano C. (2003) Activation of metabotropic glutamate receptor mGlu5 on nuclear membranes mediates intranuclear Ca2+ changes in heterologous cell types and neurons. J Biol Chem 278:28210–28219 [DOI] [PubMed] [Google Scholar]
  225. Obst OO, Rose H, Kammermeier H. (1996) Characterization of catecholamine uptake2 in isolated cardiac myocytes. Mol Cell Biochem 163-164:181–183 [DOI] [PubMed] [Google Scholar]
  226. O-Uchi J, Komukai K, Kusakari Y, Obata T, Hongo K, Sasaki H, and Kurihara S (2005) alpha1-Adrenoceptor stimulation potentiates L-type Ca2+ current through Ca2+/calmodulin-dependent PK II (CaMKII) activation in rat ventricular myocytes. Proc Natl Acad Sci U S A 102:9400–9405. [DOI] [PMC free article] [PubMed]
  227. O-Uchi J, Sasaki H, Morimoto S, Kusakari Y, Shinji H, Obata T, Hongo K, Komukai K, Kurihara S. (2008) Interaction of alpha1-adrenoceptor subtypes with different G proteins induces opposite effects on cardiac L-type Ca2+ channel. Circ Res 102:1378–1388 [DOI] [PubMed] [Google Scholar]
  228. Packer M, Bristow MR, Cohn JN, Colucci WS, Fowler MB, Gilbert EM, Shusterman NH, U.S. Carvedilol Heart Failure Study Group (1996) The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. N Engl J Med 334:1349–1355 [DOI] [PubMed] [Google Scholar]
  229. Packer M, Coats AJ, Fowler MB, Katus HA, Krum H, Mohacsi P, Rouleau JL, Tendera M, Castaigne A, Roecker EB, et al. Carvedilol Prospective Randomized Cumulative Survival Study Group (2001) Effect of carvedilol on survival in severe chronic heart failure. N Engl J Med 344:1651–1658 [DOI] [PubMed] [Google Scholar]
  230. Packer M, Fowler MB, Roecker EB, Coats AJ, Katus HA, Krum H, Mohacsi P, Rouleau JL, Tendera M, Staiger C, et al. Carvedilol Prospective Randomized Cumulative Survival (COPERNICUS) Study Group (2002) Effect of carvedilol on the morbidity of patients with severe chronic heart failure: results of the carvedilol prospective randomized cumulative survival (COPERNICUS) study. Circulation 106:2194–2199 [DOI] [PubMed] [Google Scholar]
  231. Papay RS, Shi T, Piascik MT, Naga Prasad SV, Perez DM. (2013) α₁A-adrenergic receptors regulate cardiac hypertrophy in vivo through interleukin-6 secretion. Mol Pharmacol 83:939–948 [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Paradis P, Dali-Youcef N, Paradis FW, Thibault G, Nemer M. (2000) Overexpression of angiotensin II type I receptor in cardiomyocytes induces cardiac hypertrophy and remodeling. Proc Natl Acad Sci USA 97:931–936 [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Patel MB, Stewart JM, Loud AV, Anversa P, Wang J, Fiegel L, Hintze TH. (1991) Altered function and structure of the heart in dogs with chronic elevation in plasma norepinephrine. Circulation 84:2091–2100 [DOI] [PubMed] [Google Scholar]
  234. Pediani JD, Colston JF, Caldwell D, Milligan G, Daly CJ, McGrath JC. (2005) Beta-arrestin-dependent spontaneous alpha1a-adrenoceptor endocytosis causes intracellular transportation of alpha-blockers via recycling compartments. Mol Pharmacol 67:992–1004 [DOI] [PubMed] [Google Scholar]
  235. Piascik MT, Perez DM. (2001) Alpha1-adrenergic receptors: new insights and directions. J Pharmacol Exp Ther 298:403–410 [PubMed] [Google Scholar]
  236. Pickard BW, Hodsman AB, Fraher LJ, Watson PH. (2006) Type 1 parathyroid hormone receptor (PTH1R) nuclear trafficking: association of PTH1R with importin alpha1 and beta. Endocrinology 147:3326–3332 [DOI] [PubMed] [Google Scholar]
  237. Pickard BW, Hodsman AB, Fraher LJ, Watson PH. (2007) Type 1 parathyroid hormone receptor (PTH1R) nuclear trafficking: regulation of PTH1R nuclear-cytoplasmic shuttling by importin-alpha/beta and chromosomal region maintenance 1/exportin 1. Endocrinology 148:2282–2289 [DOI] [PubMed] [Google Scholar]
  238. Piller LB, Davis BR, Cutler JA, Cushman WC, Wright JT, Jr, Williamson JD, Leenen FH, Einhorn PT, Randall OS, Golden JS, et al. The ALLHAT Collaborative Research Group (2002) Validation of Heart Failure Events in the Antihypertensive and Lipid Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) Participants Assigned to Doxazosin and Chlorthalidone. Curr Control Trials Cardiovasc Med 3:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Pocock S, Wilhelmsen L, Dickstein K, Francis G, Wittes J. (2004) The data monitoring experience in the MOXCON trial. Eur Heart J 25:1974–1978 [DOI] [PubMed] [Google Scholar]
  240. Pönicke K, Vogelsang M, Heinroth M, Becker K, Zolk O, Böhm M, Zerkowski HR, Brodde OE. (1998) Endothelin receptors in the failing and nonfailing human heart. Circulation 97:744–751 [DOI] [PubMed] [Google Scholar]
  241. Poole-Wilson PA, Cleland JG, Di Lenarda A, Hanrath P, Komajda M, Metra M, J Remme W, Swedberg K, Torp-Pedersen C. (2002) Rationale and design of the carvedilol or metoprolol European trial in patients with chronic heart failure: COMET. Eur J Heart Fail 4:321–329 [DOI] [PubMed] [Google Scholar]
  242. Poole-Wilson PA, Swedberg K, Cleland JG, Di Lenarda A, Hanrath P, Komajda M, Lubsen J, Lutiger B, Metra M, Remme WJ, et al. Carvedilol Or Metoprolol European Trial Investigators (2003) Comparison of carvedilol and metoprolol on clinical outcomes in patients with chronic heart failure in the Carvedilol Or Metoprolol European Trial (COMET): randomised controlled trial. Lancet 362:7–13 [DOI] [PubMed] [Google Scholar]
  243. Post GR, Swiderski C, Waldrop BA, Salty L, Glembotski CC, Wolthuis RM, Mochizuki N. (2002) Guanine nucleotide exchange factor-like factor (Rlf) induces gene expression and potentiates α 1-adrenergic receptor-induced transcriptional responses in neonatal rat ventricular myocytes. J Biol Chem 277:15286–15292 [DOI] [PubMed] [Google Scholar]
  244. Pu WT, Ma Q, Izumo S. (2003) NFAT transcription factors are critical survival factors that inhibit cardiomyocyte apoptosis during phenylephrine stimulation in vitro. Circ Res 92:725–731 [DOI] [PubMed] [Google Scholar]
  245. Ravens U, Wang XL, Wettwer E. (1989) Alpha adrenoceptor stimulation reduces outward currents in rat ventricular myocytes. J Pharmacol Exp Ther 250:364–370 [PubMed] [Google Scholar]
  246. Re M, Pampillo M, Savard M, Dubuc C, McArdle CA, Millar RP, Conn PM, Gobeil F, Jr, Bhattacharya M, Babwah AV. (2010) The human gonadotropin releasing hormone type I receptor is a functional intracellular GPCR expressed on the nuclear membrane. PLoS ONE 5:e11489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Rockman HA, Choi DJ, Rahman NU, Akhter SA, Lefkowitz RJ, Koch WJ. (1996) Receptor-specific in vivo desensitization by the G protein-coupled receptor kinase-5 in transgenic mice. Proc Natl Acad Sci USA 93:9954–9959 [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Roger VL, Go AS, Lloyd-Jones DM, Adams RJ, Berry JD, Brown TM, Carnethon MR, Dai S, de Simone G, Ford ES, et al. American Heart Association Statistics Committee and Stroke Statistics Subcommittee (2011) Heart disease and stroke statistics—2011 update: a report from the American Heart Association. Circulation 123:e18–e209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Rohde S, Sabri A, Kamasamudran R, Steinberg SF. (2000) The alpha(1)-adrenoceptor subtype- and protein kinase C isoform-dependence of Norepinephrine’s actions in cardiomyocytes. J Mol Cell Cardiol 32:1193–1209 [DOI] [PubMed] [Google Scholar]
  250. Rokosh DG, Bailey BA, Stewart AF, Karns LR, Long CS, Simpson PC. (1994) Distribution of alpha 1C-adrenergic receptor mRNA in adult rat tissues by RNase protection assay and comparison with alpha 1B and alpha 1D. Biochem Biophys Res Commun 200:1177–1184 [DOI] [PubMed] [Google Scholar]
  251. Rokosh DG, Simpson PC. (2002) Knockout of the alpha 1A/C-adrenergic receptor subtype: the alpha 1A/C is expressed in resistance arteries and is required to maintain arterial blood pressure. Proc Natl Acad Sci USA 99:9474–9479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Rokosh DG, Stewart AF, Chang KC, Bailey BA, Karliner JS, Camacho SA, Long CS, Simpson PC. (1996) Alpha1-adrenergic receptor subtype mRNAs are differentially regulated by alpha1-adrenergic and other hypertrophic stimuli in cardiac myocytes in culture and in vivo. Repression of alpha1B and alpha1D but induction of alpha1C. J Biol Chem 271:5839–5843 [DOI] [PubMed] [Google Scholar]
  253. Rorabaugh BR, Ross SA, Gaivin RJ, Papay RS, McCune DF, Simpson PC, Perez DM. (2005) alpha1A- but not alpha1B-adrenergic receptors precondition the ischemic heart by a staurosporine-sensitive, chelerythrine-insensitive mechanism. Cardiovasc Res 65:436–445 [DOI] [PubMed] [Google Scholar]
  254. Ross SA, Rorabaugh BR, Chalothorn D, Yun J, Gonzalez-Cabrera PJ, McCune DF, Piascik MT, Perez DM. (2003) The alpha(1B)-adrenergic receptor decreases the inotropic response in the mouse Langendorff heart model. Cardiovasc Res 60:598–607 [DOI] [PubMed] [Google Scholar]
  255. Sakata Y, Hoit BD, Liggett SB, Walsh RA, Dorn GW., 2nd (1998) Decompensation of pressure-overload hypertrophy in G alpha q-overexpressing mice. Circulation 97:1488–1495 [DOI] [PubMed] [Google Scholar]
  256. Salazar NC, Chen J, Rockman HA. (2007) Cardiac GPCRs: GPCR signaling in healthy and failing hearts. Biochim Biophys Acta 1768:1006–1018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Sato R, Koumi S. (1995) Modulation of the inwardly rectifying K+ channel in isolated human atrial myocytes by alpha 1-adrenergic stimulation. J Membr Biol 148:185–191 [DOI] [PubMed] [Google Scholar]
  258. Schomig E, Lazar A, Grundemann D. (2006) Extraneuronal monoamine transporter and organic cation transporters 1 and 2: A review of transport efficiency. Handb Exp Pharmacol 175:151–180 [DOI] [PubMed] [Google Scholar]
  259. Schwinn DA, Roehrborn CG. (2008) Alpha1-adrenoceptor subtypes and lower urinary tract symptoms. Int J Urol 15:193–199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Sica DA. (2005) Alpha1-adrenergic blockers: current usage considerations. J Clin Hypertens (Greenwich) 7:757–762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Simpson P. (1983) Norepinephrine-stimulated hypertrophy of cultured rat myocardial cells is an alpha 1 adrenergic response. J Clin Invest 72:732–738 [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Simpson P. (1985) Stimulation of hypertrophy of cultured neonatal rat heart cells through an alpha 1-adrenergic receptor and induction of beating through an alpha 1- and beta 1-adrenergic receptor interaction. Evidence for independent regulation of growth and beating. Circ Res 56:884–894 [DOI] [PubMed] [Google Scholar]
  263. Simpson P, McGrath A, Savion S. (1982) Myocyte hypertrophy in neonatal rat heart cultures and its regulation by serum and by catecholamines. Circ Res 51:787–801 [DOI] [PubMed] [Google Scholar]
  264. Simpson PC. (1988) Comments on “Load regulation of the properties of adult feline cardiocytes: the role of substrate adhesion.”Circ Res 1986;58:692-705. Circ Res 62:864–869 [DOI] [PubMed] [Google Scholar]
  265. Simpson PC. (2006) Lessons from Knockouts: the alpha-1-ARs, in The Adrenergic Receptors in the 21st Century (Perez DM, ed) pp 207–240, Humana Press, Totowa, NJ [Google Scholar]
  266. Simpson PC. (2011) Where are the new drugs to treat heart failure? Introduction to the special issue on “key signaling molecules in hypertrophy and heart failure”. J Mol Cell Cardiol 51:435–437 [DOI] [PubMed] [Google Scholar]
  267. Simpson PC, Long CS, Waspe LE, Henrich CJ, Ordahl CP. (1989) Transcription of early developmental isogenes in cardiac myocyte hypertrophy. J Mol Cell Cardiol 21 (Suppl 5):79–89 [DOI] [PubMed] [Google Scholar]
  268. Sjaastad I, Schiander I, Sjetnan A, Qvigstad E, Bøkenes J, Sandnes D, Osnes JB, Sejersted OM, Skomedal T. (2003) Increased contribution of alpha 1- vs. beta-adrenoceptor-mediated inotropic response in rats with congestive heart failure. Acta Physiol Scand 177:449–458 [DOI] [PubMed] [Google Scholar]
  269. Skomedal T, Borthne K, Aass H, Geiran O, Osnes JB. (1997) Comparison between alpha-1 adrenoceptor-mediated and beta adrenoceptor-mediated inotropic components elicited by norepinephrine in failing human ventricular muscle. J Pharmacol Exp Ther 280:721–729 [PubMed] [Google Scholar]
  270. Snabaitis AK, Muntendorf A, Wieland T, Avkiran M. (2005) Regulation of the extracellular signal-regulated kinase pathway in adult myocardium: differential roles of G(q/11), Gi and G(12/13) proteins in signalling by alpha1-adrenergic, endothelin-1 and thrombin-sensitive protease-activated receptors. Cell Signal 17:655–664 [DOI] [PubMed] [Google Scholar]
  271. SoRelle R. (2000) National Heart, Lung, and Blood Institute halts part of antihypertensive and lipid-lowering treatment to prevent heart attack trial (ALLHAT). Circulation 101:E9025. [DOI] [PubMed] [Google Scholar]
  272. Staehelin M, Simons P, Jaeggi K, Wigger N. (1983) CGP-12177. A hydrophilic beta-adrenergic receptor radioligand reveals high affinity binding of agonists to intact cells. J Biol Chem 258:3496–3502 [PubMed] [Google Scholar]
  273. Stafford RS, Furberg CD, Finkelstein SN, Cockburn IM, Alehegn T, Ma J. (2004) Impact of clinical trial results on national trends in alpha-blocker prescribing, 1996-2002. JAMA 291:54–62 [DOI] [PubMed] [Google Scholar]
  274. Stanasila L, Abuin L, Dey J, Cotecchia S. (2008) Different internalization properties of the alpha1a- and alpha1b-adrenergic receptor subtypes: the potential role of receptor interaction with beta-arrestins and AP50. Mol Pharmacol 74:562–573 [DOI] [PubMed] [Google Scholar]
  275. Starksen NF, Simpson PC, Bishopric N, Coughlin SR, Lee WM, Escobedo JA, Williams LT. (1986) Cardiac myocyte hypertrophy is associated with c-myc protooncogene expression. Proc Natl Acad Sci USA 83:8348–8350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Steinberg SF, Drugge ED, Bilezikian JP, Robinson RB. (1985) Acquisition by innervated cardiac myocytes of a pertussis toxin-specific regulatory protein linked to the alpha 1-receptor. Science 230:186–188 [DOI] [PubMed] [Google Scholar]
  277. Steinfath M, Chen YY, Lavický J, Magnussen O, Nose M, Rosswag S, Schmitz W, Scholz H. (1992a) Cardiac alpha 1-adrenoceptor densities in different mammalian species. Br J Pharmacol 107:185–188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Steinfath M, Danielsen W, von der Leyen H, Mende U, Meyer W, Neumann J, Nose M, Reich T, Schmitz W, Scholz H, et al. (1992b) Reduced alpha 1- and beta 2-adrenoceptor-mediated positive inotropic effects in human end-stage heart failure. Br J Pharmacol 105:463–469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Stewart AF, Rokosh DG, Bailey BA, Karns LR, Chang KC, Long CS, Kariya K, Simpson PC. (1994) Cloning of the rat alpha 1C-adrenergic receptor from cardiac myocytes. alpha 1C, alpha 1B, and alpha 1D mRNAs are present in cardiac myocytes but not in cardiac fibroblasts. Circ Res 75:796–802 [DOI] [PubMed] [Google Scholar]
  280. Stewart JM, Patel MB, Wang J, Ochoa M, Gewitz M, Loud AV, Anversa P, Hintze TH. (1992) Chronic elevation of norepinephrine in conscious dogs produces hypertrophy with no loss of LV reserve. Am J Physiol 262:H331–H339 [DOI] [PubMed] [Google Scholar]
  281. Swedberg K, Bristow MR, Cohn JN, Dargie H, Straub M, Wiltse C, Wright TJ, Moxonidine Safety and Efficacy (MOXSE) Investigators (2002) Effects of sustained-release moxonidine, an imidazoline agonist, on plasma norepinephrine in patients with chronic heart failure. Circulation 105:1797–1803 [DOI] [PubMed] [Google Scholar]
  282. Tadevosyan A, Maguy A, Villeneuve LR, Babin J, Bonnefoy A, Allen BG, Nattel S. (2010) Nuclear-delimited angiotensin receptor-mediated signaling regulates cardiomyocyte gene expression. J Biol Chem 285:22338–22349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Tadevosyan A, Vaniotis G, Allen BG, Hébert TE, Nattel S. (2012) G protein-coupled receptor signalling in the cardiac nuclear membrane: evidence and possible roles in physiological and pathophysiological function. J Physiol 590:1313–1330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Tanoue A, Nasa Y, Koshimizu T, Shinoura H, Oshikawa S, Kawai T, Sunada S, Takeo S, Tsujimoto G. (2002) The alpha(1D)-adrenergic receptor directly regulates arterial blood pressure via vasoconstriction. J Clin Invest 109:765–775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Teerlink JR, Massie BM. (1999) Beta-adrenergic blocker mortality trials in congestive heart failure. Am J Cardiol 84 (9A):94R–102R [DOI] [PubMed] [Google Scholar]
  286. Tejero-Taldo MI, Gursoy E, Zhao TC, Kukreja RC. (2002) Alpha-adrenergic receptor stimulation produces late preconditioning through inducible nitric oxide synthase in mouse heart. J Mol Cell Cardiol 34:185–195 [DOI] [PubMed] [Google Scholar]
  287. Terzic A, Pucéat M, Clément O, Scamps F, Vassort G. (1992) Alpha 1-adrenergic effects on intracellular pH and calcium and on myofilaments in single rat cardiac cells. J Physiol 447:275–292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Tohse N, Nakaya H, Hattori Y, Endou M, Kanno M. (1990) Inhibitory effect mediated by alpha 1-adrenoceptors on transient outward current in isolated rat ventricular cells. Pflugers Arch 415:575–581 [DOI] [PubMed] [Google Scholar]
  289. Tohse N, Nakaya H, Kanno M. (1992) Alpha 1-adrenoceptor stimulation enhances the delayed rectifier K+ current of guinea pig ventricular cells through the activation of protein kinase C. Circ Res 71:1441–1446 [DOI] [PubMed] [Google Scholar]
  290. Tosaki A, Behjet NS, Engelman DT, Engelman RM, Das DK. (1995) Alpha-1 adrenergic receptor agonist-induced preconditioning in isolated working rat hearts. J Pharmacol Exp Ther 273:689–694 [PubMed] [Google Scholar]
  291. Tsuchida A, Liu Y, Liu GS, Cohen MV, Downey JM. (1994) alpha 1-adrenergic agonists precondition rabbit ischemic myocardium independent of adenosine by direct activation of protein kinase C. Circ Res 75:576–585 [DOI] [PubMed] [Google Scholar]
  292. Turnbull L, McCloskey DT, O’Connell TD, Simpson PC, Baker AJ. (2003) Alpha 1-adrenergic receptor responses in alpha 1AB-AR knockout mouse hearts suggest the presence of alpha 1D-AR. Am J Physiol Heart Circ Physiol 284:H1104–H1109 [DOI] [PubMed] [Google Scholar]
  293. Vago T, Bevilacqua M, Norbiato G, Baldi G, Chebat E, Bertora P, Baroldi G, Accinni R. (1989) Identification of alpha 1-adrenergic receptors on sarcolemma from normal subjects and patients with idiopathic dilated cardiomyopathy: characteristics and linkage to GTP-binding protein. Circ Res 64:474–481 [DOI] [PubMed] [Google Scholar]
  294. Valks DM, Cook SA, Pham FH, Morrison PR, Clerk A, Sugden PH. (2002) Phenylephrine promotes phosphorylation of Bad in cardiac myocytes through the extracellular signal-regulated kinases 1/2 and protein kinase A. J Mol Cell Cardiol 34:749–763 [DOI] [PubMed] [Google Scholar]
  295. Van Tassell BW, Rondina MT, Huggins F, Gilbert EM, Munger MA. (2008) Carvedilol increases blood pressure response to phenylephrine infusion in heart failure subjects with systolic dysfunction: evidence of improved vascular alpha1-adrenoreceptor signal transduction. Am Heart J 156:315–321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Vaniotis G, Del Duca D, Trieu P, Rohlicek CV, Hébert TE, Allen BG. (2011) Nuclear β-adrenergic receptors modulate gene expression in adult rat heart. Cell Signal 23:89–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Vaughan ED., Jr (2003) Medical management of benign prostatic hyperplasia—are two drugs better than one? N Engl J Med 349:2449–2451 [DOI] [PubMed] [Google Scholar]
  298. Vecchione C, Fratta L, Rizzoni D, Notte A, Poulet R, Porteri E, Frati G, Guelfi D, Trimarco V, Mulvany MJ, et al. (2002) Cardiovascular influences of alpha1b-adrenergic receptor defect in mice. Circulation 105:1700–1707 [DOI] [PubMed] [Google Scholar]
  299. Vega RB, Harrison BC, Meadows E, Roberts CR, Papst PJ, Olson EN, McKinsey TA. (2004) Protein kinases C and D mediate agonist-dependent cardiac hypertrophy through nuclear export of histone deacetylase 5. Mol Cell Biol 24:8374–8385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Vettel C, Wittig K, Vogt A, Wuertz CM, El-Armouche A, Lutz S, Wieland T. (2012) A novel player in cellular hypertrophy: Giβγ/PI3K-dependent activation of the RacGEF TIAM-1 is required for α₁-adrenoceptor induced hypertrophy in neonatal rat cardiomyocytes. J Mol Cell Cardiol 53:165–175 [DOI] [PubMed] [Google Scholar]
  301. Vinge LE, Andressen KW, Attramadal T, Andersen GO, Ahmed MS, Peppel K, Koch WJ, Freedman NJ, Levy FO, Skomedal T, et al. (2007) Substrate specificities of g protein-coupled receptor kinase-2 and -3 at cardiac myocyte receptors provide basis for distinct roles in regulation of myocardial function. Mol Pharmacol 72:582–591 [DOI] [PubMed] [Google Scholar]
  302. Vinge LE, Øie E, Andersson Y, Grøgaard HK, Andersen G, Attramadal H. (2001) Myocardial distribution and regulation of GRK and beta-arrestin isoforms in congestive heart failure in rats. Am J Physiol Heart Circ Physiol 281:H2490–H2499 [DOI] [PubMed] [Google Scholar]
  303. Volz A, Piper HM, Siegmund B, Schwartz P. (1991) Longevity of adult ventricular rat heart muscle cells in serum-free primary culture. J Mol Cell Cardiol 23:161–173 [DOI] [PubMed] [Google Scholar]
  304. Wang BH, Du XJ, Autelitano DJ, Milano CA, Woodcock EA. (2000) Adverse effects of constitutively active alpha(1B)-adrenergic receptors after pressure overload in mouse hearts. Am J Physiol Heart Circ Physiol 279:H1079–H1086 [DOI] [PubMed] [Google Scholar]
  305. Wang GY, McCloskey DT, Turcato S, Swigart PM, Simpson PC, Baker AJ. (2006) Contrasting inotropic responses to alpha1-adrenergic receptor stimulation in left versus right ventricular myocardium. Am J Physiol Heart Circ Physiol 291:H2013–H2017 [DOI] [PubMed] [Google Scholar]
  306. Wang GY, Yeh CC, Jensen BC, Mann MJ, Simpson PC, Baker AJ. (2010) Heart failure switches the RV alpha1-adrenergic inotropic response from negative to positive. Am J Physiol Heart Circ Physiol 298:H913–H920 [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Wang H, Yang B, Zhang Y, Han H, Wang J, Shi H, Wang Z. (2001) Different subtypes of alpha1-adrenoceptor modulate different K+ currents via different signaling pathways in canine ventricular myocytes. J Biol Chem 276:40811–40816 [DOI] [PubMed] [Google Scholar]
  308. Wang XL, Wettwer E, Gross G, Ravens U. (1991) Reduction of cardiac outward currents by alpha-1 adrenoceptor stimulation: a subtype-specific effect? J Pharmacol Exp Ther 259:783–788 [PubMed] [Google Scholar]
  309. Waspe LE, Ordahl CP, Simpson PC. (1990) The cardiac beta-myosin heavy chain isogene is induced selectively in alpha 1-adrenergic receptor-stimulated hypertrophy of cultured rat heart myocytes. J Clin Invest 85:1206–1214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  310. Wencker D, Chandra M, Nguyen K, Miao W, Garantziotis S, Factor SM, Shirani J, Armstrong RC, Kitsis RN. (2003) A mechanistic role for cardiac myocyte apoptosis in heart failure. J Clin Invest 111:1497–1504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Whelan RS, Kaplinskiy V, Kitsis RN. (2010) Cell death in the pathogenesis of heart disease: mechanisms and significance. Annu Rev Physiol 72:19–44 [DOI] [PubMed] [Google Scholar]
  312. Wollert KC, Drexler H. (2002) Carvedilol prospective randomized cumulative survival (COPERNICUS) trial: carvedilol as the sun and center of the beta-blocker world? Circulation 106:2164–2166 [DOI] [PubMed] [Google Scholar]
  313. Woo SH, Lee CO. (1999) Role of PKC in the effects of alpha1-adrenergic stimulation on Ca2+ transients, contraction and Ca2+ current in guinea-pig ventricular myocytes. Pflugers Arch 437:335–344 [DOI] [PubMed] [Google Scholar]
  314. Wright CD, Chen Q, Baye NL, Huang Y, Healy CL, Kasinathan S, O’Connell TD. (2008) Nuclear alpha1-adrenergic receptors signal activated ERK localization to caveolae in adult cardiac myocytes. Circ Res 103:992–1000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Wright CD, Wu SC, Dahl EF, Sazama AJ, O’Connell TD. (2012) Nuclear localization drives α1-adrenergic receptor oligomerization and signaling in cardiac myocytes. Cell Signal 24:794–802 [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Xiao L, Pimental DR, Amin JK, Singh K, Sawyer DB, Colucci WS. (2001) MEK1/2-ERK1/2 mediates alpha1-adrenergic receptor-stimulated hypertrophy in adult rat ventricular myocytes. J Mol Cell Cardiol 33:779–787 [DOI] [PubMed] [Google Scholar]
  317. Xiao RP, Zhu W, Zheng M, Chakir K, Bond R, Lakatta EG, Cheng H. (2004) Subtype-specific beta-adrenoceptor signaling pathways in the heart and their potential clinical implications. Trends Pharmacol Sci 25:358–365 [DOI] [PubMed] [Google Scholar]
  318. Yang LL, Gros R, Kabir MG, Sadi A, Gotlieb AI, Husain M, Stewart DJ. (2004) Conditional cardiac overexpression of endothelin-1 induces inflammation and dilated cardiomyopathy in mice. Circulation 109:255–261 [DOI] [PubMed] [Google Scholar]
  319. Yang M, Reese J, Cotecchia S, Michel MC. (1998) Murine alpha1-adrenoceptor subtypes. I. Radioligand binding studies. J Pharmacol Exp Ther 286:841–847 [PubMed] [Google Scholar]
  320. Zakir RM, Folefack A, Saric M, Berkowitz RL. (2009) The use of midodrine in patients with advanced heart failure. Congest Heart Fail 15:108–111 [DOI] [PubMed] [Google Scholar]
  321. Zhang L, Malik S, Kelley GG, Kapiloff MS, Smrcka AV. (2011) Phospholipase C epsilon scaffolds to muscle-specific A kinase anchoring protein (mAKAPbeta) and integrates multiple hypertrophic stimuli in cardiac myocytes. J Biol Chem 286:23012–23021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Zhang L, Malik S, Pang J, Wang H, Park KM, Yule DI, Blaxall BC, Smrcka AV. (2013) Phospholipase Cε hydrolyzes perinuclear phosphatidylinositol 4-phosphate to regulate cardiac hypertrophy. Cell 153:216–227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Zhang S, Hiraoka M, Hirano Y. (1998) Effects of alpha1-adrenergic stimulation on L-type Ca2+ current in rat ventricular myocytes. J Mol Cell Cardiol 30:1955–1965 [DOI] [PubMed] [Google Scholar]
  324. Zhao X, Park J, Ho D, Gao S, Yan L, Ge H, Iismaa S, Lin L, Tian B, Vatner DE, et al. (2012) Cardiomyocyte overexpression of the α1A-adrenergic receptor in the rat phenocopies second but not first window preconditioning. Am J Physiol Heart Circ Physiol 302:H1614–H1624 [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Zhu H, McElwee-Witmer S, Perrone M, Clark KL, Zilberstein A. (2000) Phenylephrine protects neonatal rat cardiomyocytes from hypoxia and serum deprivation-induced apoptosis. Cell Death Differ 7:773–784 [DOI] [PubMed] [Google Scholar]
  326. Zuscik MJ, Chalothorn D, Hellard D, Deighan C, McGee A, Daly CJ, Waugh DJ, Ross SA, Gaivin RJ, Morehead AJ, et al. (2001) Hypotension, autonomic failure, and cardiac hypertrophy in transgenic mice overexpressing the alpha 1B-adrenergic receptor. J Biol Chem 276:13738–13743 [DOI] [PubMed] [Google Scholar]
  327. Zwart R, Verhaagh S, Buitelaar M, Popp-Snijders C, Barlow DP. (2001) Impaired activity of the extraneuronal monoamine transporter system known as uptake-2 in Orct3/Slc22a3-deficient mice. Mol Cell Biol 21:4188–4196 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Pharmacological Reviews are provided here courtesy of American Society for Pharmacology and Experimental Therapeutics

RESOURCES