Next Article in Journal
Functional Characterization of the N-Acetylmuramyl-l-Alanine Amidase, Ami1, from Mycobacterium abscessus
Next Article in Special Issue
Neurohormonal Modulation as a Therapeutic Target in Pulmonary Hypertension
Previous Article in Journal
Signal Transduction Pathways Activated by Innate Immunity in Mast Cells: Translating Sensing of Changes into Specific Responses
Previous Article in Special Issue
Novel Insights into Beta 2 Adrenergic Receptor Function in the rd10 Model of Retinitis Pigmentosa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiac and Vascular α1-Adrenoceptors in Congestive Heart Failure: A Systematic Review

1
Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey
2
Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
3
Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
4
Department of Pharmacology, Johannes Gutenberg University, 55131 Mainz, Germany
*
Author to whom correspondence should be addressed.
These authors have contributed equally.
Cells 2020, 9(11), 2412; https://doi.org/10.3390/cells9112412
Submission received: 29 September 2020 / Revised: 29 October 2020 / Accepted: 2 November 2020 / Published: 4 November 2020
(This article belongs to the Special Issue Advances in Adrenergic Receptor Biology)

Abstract

:
As heart failure (HF) is a devastating health problem worldwide, a better understanding and the development of more effective therapeutic approaches are required. HF is characterized by sympathetic system activation which stimulates α- and β-adrenoceptors (ARs). The exposure of the cardiovascular system to the increased locally released and circulating levels of catecholamines leads to a well-described downregulation and desensitization of β-ARs. However, information on the role of α-AR is limited. We have performed a systematic literature review examining the role of both cardiac and vascular α1-ARs in HF using 5 databases for our search. All three α1-AR subtypes (α1A, α1B and α1D) are expressed in human and animal hearts and blood vessels in a tissue-dependent manner. We summarize the changes observed in HF regarding the density, signaling and responses of α1-ARs. Conflicting findings arise from different studies concerning the influence that HF has on α1-AR expression and function; in contrast to β-ARs there is no consistent evidence for down-regulation or desensitization of cardiac or vascular α1-ARs. Whether α1-ARs are a therapeutic target in HF remains a matter of debate.

1. Introduction

The sympathetic nervous system primarily regulates cardiac function via β-adrenoceptors (β-ARs) [1,2], whereas α1-adrenoceptors (α1-ARs) play an important role in the regulation of vascular tone [3]. Yet, α1-ARs are also expressed in various cell types of the heart and can contribute to the regulation of cardiac contraction [4,5], rhythm [6,7] and hypertrophy [8]. However, the role of cardiac α1-ARs appears to be species-dependent with a considerable contribution in rodents but a much smaller (if any) contribution in the healthy human heart [1]. On the other hand, cardiac expression of other subtypes such as β3-ARs also appears to be lower in the human heart than in that of other species, but such receptors become up-regulated in congestive heart failure (CHF) [9].
The sympatho-adrenal system is activated in acute and chronic heart failure (HF). Accordingly, plasma levels of noradrenaline and its co-transmitters are increased in CHF [10,11]. This appears to correlate with a higher risk of mortality [12]. In line with this, patients with reduced left ventricular function appear to be more prone to post-operative vasoplegia that additionally increases the mortality and morbidity risk [13,14]. The regulation of β-ARs in CHF has been reviewed extensively and appears distinct for the three β-AR subtypes [1].
α1-ARs are also a subfamily with three members: α1A, α1B and α1D [15]. Data outside the cardiovascular system suggest that they can undergo differential regulation upon extended agonist exposure [16]. Nevertheless, until now, the α1-AR regulation in HF has not been fully elucidated and results might vary a lot between different studies. Against this background we discuss the expression and function of α1-ARs and their subtypes in the heart and vasculature of patients with CHF and animal models thereof based upon a systematic review of the literature.

2. Materials and Methods

A broad literature search was performed in 5 databases (PubMed/MEDLINE, Embase, Web of Science, Cochrane Library, Emcare). The search was performed using the MeSH terms “Heart failure” and “Receptors, Adrenergic alpha 1” and by combining each term with different keyword variations and free text words. No language limits were applied. The final search was performed on the 15th of April 2020. Details of the search strategy are provided in the Supplementary Materials.
Animal and human studies were included that investigated changes of cardiac and/or vascular α1-AR expression or function in CHF. There were no limitations regarding the study design or the etiology/model of CHF. Two independent reviewers (GKM and OP) assessed all articles for eligibility that the search strategy had yielded. The first screening was performed by assessing only the titles and abstracts of the articles, while the final decision for inclusion was based upon reading the full text of the selected articles. The reference lists of all relevant articles and reviews were manually checked for inclusion of additional eligible studies that were not identified by the primary search strategy. Any disagreements that emerged during the study selection process were resolved by discussion. A Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA, www.prisma-statement.org) flowchart is shown as Figure 1.
The data from the eligible studies were retrieved and recorded in predefined Google Spreadsheet forms that offer the possibility of auditing. Data collection included study characteristics and study design information and changes that were identified regarding the expression and function of α1-ARs in cells, tissue and in vivo.

3. Considerations Related to Data Interpretation

Most investigators have reported mRNA levels after normalization to a reference gene, often glyceraldehyde-3-phosphate dehydrogenase (GADPH) [17]. However, they typically failed to document whether the expression of the reference gene was stable. A stable expression of reference genes cannot necessarily be expected [18]. Specifically, studies in multiple cell types and tissues [19] including the heart [2] have found that exposure to β-AR agonists can down-regulate GAPDH expression; based on the elevated plasma catecholamine levels in CHF [10,11] this is likely to be of relevance for the mRNA data discussed below.
Cardiac α1-AR expression at the protein level has primarily been studied by radioligand binding and antibody-based techniques. While radioligands such as [3H]-prazosin or [125I]-BE 2254 (also known as [125I]-HEAT) have high specificity for α1-ARs, they do not discriminate between subtypes and, particularly in the case of [3H]-prazosin, are not very sensitive. In contrast, antibody-based approaches such as immunoblotting or immunohistochemistry potentially can be subtype-selective and be used to examine distribution within a tissue. However, multiple studies have demonstrated that the vast majority of commercially available α1-AR subtype antibodies lack target specificity when tested under stringent conditions [20,21,22]. These limitations need to be considered in the interpretation of the subsequently discussed studies.
Another important factor that might influence the results of different studies is the exact location of α1-ARs in the peripheral vasculature. It has been demonstrated that α1-AR receptors are located not only on smooth muscle cells, but, also on endothelial, adventitial and nerve cells [23,24]. This is a rather important finding that should be taken into account when interpreting results from expression and functional studies, since the total α1-AR mRNA and the actual role of specific α1-subtypes mediating vascular responses might differ depending on the specific cell on which they are expressed.
CHF can be caused by different pathologies, for instance by chronic myocardial ischemia, valvular heart disease or viruses, or can be idiopathic. As CHF in humans can be caused by a variety of pathophysiologic mechanisms, it is important to realize that subjects within an animal model typically are more homogeneous than patient cohorts; on the other hand, each model reflects a different cause and, potentially, a different pathophysiology. Among the many reported animal models of CHF, post-myocardial infarction [25,26,27], cardiac pacing causing tachycardiomyopathy [28], drug-induced [29,30] and hereditary models [31] have been applied to studies of cardiovascular α1-ARs. Finally, pathophysiology and regulation of α1-ARs may not only differ between patients and experimental models but also between species. While most animal studies related to cardiovascular α1-ARs in CHF were done with rats [2,26,32], guinea pigs [33,34], mice [27,29], hamster [35], cats [36] and dogs [37,38] have also been used.

4. Results

4.1. Study Selection

The literature search resulted in 1461 unique, original articles. G.K.-M. and O.P. independently screened the titles and abstracts of all the articles and excluded 1312 articles because of lack of relevance. After assessing the full text of the remaining 149 potentially relevant articles, and resolving any disagreements, the two reviewers decided to include 65 articles (Figure 1). Additional relevant studies that were retrieved from other articles were also included. This study selection process led to an inclusion of 33 unique reports on cardiac and 31 unique reports on vascular α1-AR. One study was reviewed for both cardiac and vascular α1-ARs leading to a total number of included studies of 65.

4.2. Cardiac α1-AR

4.2.1. mRNA Expression

There are three α1-AR subtypes: α1A-, α1B- and α1D-ARs which have been identified in both animal and human hearts at the mRNA level [1,39,40]. All studies agree that α1A-AR is the most abundant subtype in the heart but the presence of α1B- and α1D-AR mRNA were also confirmed [1]. Although animal hearts express more α1-AR in the heart, human and rodent hearts are reported to locate α1-AR subtypes similarly, as α1A- and α1B-subtypes are predominantly found in myocardium whereas the α1D-subtype is expressed in coronary arteries [40,41]. Myagmar et al. claimed that α1A-ARs are present in a myocyte subset while α1B-ARs are in all cells [42]. Several studies reported changes in α1-AR subtype mRNA levels in CHF, but the results are heterogeneous (Table 1).
In human failing hearts, Fischer et al. demonstrated that mRNA of α1A- and α1B-ARs in the left ventricle was increased while no change in mRNA levels was observed in other cardiac regions [43]. Similarly, Jensen et al. reported that α1A mRNA was increased in failing left ventricle LV and tended to increase in failing RV, but there was no change in α1B and α1D mRNA in any region [41]. On the other hand, Monto et al. did not confirm an altered expression of the three subtypes in human failing hearts; however, the relative abundance increased when considering the reduced β-AR expression [17]. They suggested that expression of α1A-ARs in the LV correlates with the left ventricular ejection fraction (LVEF) suggesting that this subtype may contribute to maintain cardiac inotropy in the failing heart. Moreover, a study in a rat coronary ligation model of CHF reported α1D-AR mRNA to be decreased; a numerical reduction of α1A- and α1B-AR mRNA did not reach statistical significance [2].

4.2.2. Protein Expression

Cardiac α1-ARs were first identified at the protein level in rats by Williams and Lefkowitz [44] and thereafter confirmed in many species. Their quantitative expression at the protein level ranges from a relatively high expression in rodents to a low expression in humans, where their role appears considerably lower than that of β-ARs [1,39,40]. Despite numerous studies, data on α1-AR binding properties in human HF have remained inconsistent (Table 2). The density of α1-ARs was reported as increased [45,46,47,48], unaltered [41,45,49,50,51,52], or decreased [43,53].
However, even a lack of change in α1-AR protein expression may be biologically relevant for two reasons: Firstly, it is interesting to note that expression does not consistently decrease despite exposure to elevated plasma catecholamines [10,11]. However, studies outside the cardiovascular system have found that prolonged agonist exposure does not always lead to down-regulation of α1-AR subtypes and in some cases can even be associated with an up-regulation [16]. Secondly, an unaltered expression of α1-ARs in face of a consistently found down-regulation of β-ARs [1,39] implies a change in the relative role of the two adrenoceptor sub-families, which in its own right may have implications for the regulation of cardiac function [50,55].
A possible explanation for the controversial results on the regulation of cardiac α1-AR mRNA and protein is heterogeneity based on various etiologies of CHF. For instance, it was reported that α1-AR protein was increased in ischemic human cardiomyopathy but remained constant in dilated cardiomyopathy within the same study [45]. Similarly, the degree of HF may contribute to heterogeneous findings. While α1-AR density was not changed in a mild-to-moderate heart failure group, it was decreased in severely failing human hearts [52]. Finally, the change in α1-AR density may depend on the cardiac region under investigation: α1A- and α1B-AR protein were not changed in failing left ventricle, but α1A-AR binding was increased in right ventricle [41].
Studies in rats mainly found that α1-AR density remained constant after myocardial infarction [25,26,32,55,56], which is in line with the lack of a consistent change in human CHF (Table 2). However, an increase was also reported in a rat model of myocardial infarction [54]. Noradrenaline infusion, on the other hand, resulted in a decrease in α1-AR density in rats [30], demonstrating that cardiac α1-ARs are in principle susceptible to agonist-induced down-regulation.
There is also a substantial number of studies investigating the changes in cardiac α1-ARs using different animal models. Cardiomyopathic hamster [35] and dog [37] hearts were shown to have decreased α1-AR concentrations. In contrast, an increase in cardiac α1-AR density was demonstrated in a guinea pig model of HF caused by aortic constriction [33]. In addition, increase α1-AR densities were reported in cat [36] and guinea pig [34] ischemia models following coronary ligation.

4.2.3. α1-AR Subtypes

For the interpretation of subtype expression data, there is a technical issue between α1-AR mRNA and protein levels. While mRNA of all α1-AR subtypes were detected in animal and human hearts, binding studies demonstrated the presence of only α1A- and α1B-subtypes, but not α1D-AR [39,41,57]. Lack of α1D-AR expression at the protein level has been shown in many tissues expressing corresponding mRNA [58].
Animal species do not only differ from humans by expressing more α1-AR in the heart but also by exhibiting a different subtype expression profile. For instance, rat [59] and mouse [58] hearts were reported to express α1A1B-AR in an approximate 20/80 ratio. Moreover, previous studies suggest that the three subtypes mediate different effects in the heart. α1A-AR has been reported to mediate positive inotropy in right ventricles of human hearts and this response has been found to be reduced only slightly in failing hearts. Thus, α1A-subtype-mediated inotropy is suggested to be significant source of inotropic support [60]. However, in nonfailing murine hearts, stimulation by the α1A-AR selective agonist, A61603 or the non-subtype-selective α1-AR agonist, phenylephrine mediated negative inotropic effects in right ventricle which was switched to positive inotropy in failing hearts [29]. On the other hand, the inotropic response mediated by α1B-subtype remained negative and unaltered. In the light of these findings, the authors suggested that α1A-subtype may play a role in improving contraction in RV failure and might be a therapeutic target. Another study using transgenic mice with heart-specific overexpression of α1A-AR displayed enhanced inotropy after myocardial infarction [61]. Moreover, α1D-AR mRNA decreased in rats after coronary ligation; however, this study was inconclusive because there was a non-significant trend for reduced mRNA of α1A- and α1B-ARs [2]. Thus, the authors proposed that a decrease in α1D-AR might be a protective response of the failing heart. However, the relevance of these data remains unclear because studies in several mouse tissues have demonstrated that many tissues express α1D-AR at the mRNA but not at the protein level [58].
Patients who receive left ventricular assist device to unload the failing heart and improve cardiac function were shown to have a redistribution of α1A-AR from a peri-myocytic to an intra-myocytic location along with a rise in the overall density of receptors [62]. On the other hand, α1A-AR subtype was decreased in human dilated cardiomyopathy and this result was concluded to be consistent with the theory that α1A-AR agonism may be cardioprotective [53]. Fischer et al. demonstrated that α1A- and α1B-AR mRNA levels in left ventricle of human failing hearts were increased, although α1A-, α1B- and α1D-AR binding sites were decreased [43]. They suggested that a decrease of α1B-AR can have cardioprotective effects while a decrease of α1A-AR is more likely the result of impaired heart function.

4.2.4. Signaling Pathways Involved

A schematic representation of α1-AR signal transduction is shown in Figure 2. The prototypical signaling pathway of α1-ARs involves coupling to Gq with subsequent activation of phospholipase C (PLC), which releases Ca2+ from intracellular stores and activates protein kinase C (PKC) [63]. However, cardiac α1-ARs can also couple to Gi [5,7], G12/13 [64], even Gs proteins [65] and Gh which is a GTP-binding protein with Transglutaminase activity [66] and to pathways such as inhibition of adenylyl cyclase [67], modulation of Ca2+, K+ and Cl- channel activity [7,68,69], ras activity [70] or activation of various mitogen activated protein kinases [71]. Most of these pathways have also been studied in human CHF or CHF animal models [72].
Activation of Gq-signaling pathways mediates the development of hypertrophy and the transition to HF [73]. Overexpression of Gq was demonstrated to result in marked hypertrophy [74] and HF [75]. Moreover, inhibition of Gq-mediated signaling was shown to inhibit cardiac dysfunction [76,77]. On the other hand, multiple studies in humans and several animal species have shown that HF is accompanied by an increased expression of Gi at the mRNA and protein level, whereas a reduced expression of Gs has been found less consistently [78]. Reduced GTP binding with Transglutaminase activity of Gαh were found in human HF [45]. α1-AR coupling to a GTP binding protein was detected only in pathological, but not in healthy human hearts [47].
α1-AR-mediated phosphoinositol turnover is not changed in both animal and human studies when accompanied by an unaltered receptor density in human [50] and rat [56] HF. On the other hand, in a rat model of myocardial infarction, an increase in phosphoinositol turnover was reported along with an increase in G-protein content and an upregulation of α-skeletal actin which suggests an involvement of α1-ARs in the reactive hypertrophic response [26].
α1-AR stimulation increases both the intracellular Ca2+ concentration, which activates muscle contraction, but also the Ca2+ sensitivity of cardiomyocytes. α1-AR-mediated Ca2+-transients and phosphorylation of cardiac myosin regulatory light chain were increased in a mouse right ventricular HF model [29]. The increase in Ca2+ sensitivity expected from these observations was reported in a canine HF model, apparently occurring via the Gq-RhoA-ROCK signaling pathway [38]. Similarly, the switch from negative to positive inotropy observed in mouse failing right ventricles (see below) was claimed to be mediated through a pathway involving increased myofilament Ca2+ sensitivity [27].

4.2.5. Cell and Tissue Responses

α1-AR stimulation induces a positive inotropic effect in isolated cardiac preparations such as atria, left and right ventricles (Table 3) [25,55,56,60]. The positive inotropic effect of α1-AR stimulation involves multiple subtypes, but mostly occurs via α1B-AR in rodents; when these become inactivated in vitro, the α1A-component becomes stronger and compensates for the loss of α1B response resulting in overall maintained contractile responses [79]. However, depending on experimental conditions α1-AR stimulation can also have negative inotropic effects (Table 3) [29]. Moreover, stimulation of cardiac α1-AR can increase heart rate [56], but systemic stimulation can reduce heart rate secondary to an increase in total peripheral resistance and activation of the baroreflex [80], which can mask direct chronotropic effects of α1-AR stimulation.
The chronotropic and inotropic responses to the α1-AR agonist phenylephrine were decreased in isolated right and left atria of rats with myocardial infarction [56]. As this was not accompanied by alterations of receptor density or phosphoinositol turnover, this may reflect changes of post-receptor signaling (see above). In contrast, several studies in isolated papillary muscle from humans [49,51] or rats [25,55] reported unaltered inotropic responses in CHF, which was in line with unaltered receptor density reported in these studies. The positive inotropic response to α1A-AR stimulation in human right ventricle also exhibited little change in HF in two studies [60,81]. In contrast, the phenylephrine-induced positive inotropic response in human CHF was abolished in another study [46]. In mouse models, negative inotropic effect observed in control animals was switched to positive inotropy in the failing right ventricles [29]. However PE-mediated positive inotropy in LV of nonfailing hearts remained unchanged in failing mouse hearts after myocardial infarction [27]. Cardiomyopathic hearts of Syrian hamsters were shown to have increased inotropy accompanied by increased Ca2+ levels and pertussis toxin-sensitive G-proteins [82]. Thus, with few exceptions, most studies reported little change in inotropic responses in the failing heart across species and models. This may nonetheless be biologically relevant because it would indicate an α1/β-AR balance in light of the consistently found reduction in β-adrenergic function in CHF [50,55].
Moreover, there are opposing views on the role of α1-AR subtypes on hypertrophy. While some suggest that α1A-AR were found to mediate hypertrophy development in neonatal rat cardiomyocytes in vitro [8], transgenic overexpression of α1B-AR caused a hypertrophic response resulting in HF, while α1A-AR overexpression did not affect hypertrophy and did not hasten development of HF [29,83].

4.3. Vascular α1-AR

4.3.1. mRNA Expression

Multiple studies have explored the expression of α1-AR subtype mRNA in arteries and veins of various species including rat [84,85,86,87,88], rabbit [89,90,91] and human [89,92,93,94]. It appears from these and many other studies, that all three α1-AR subtypes are expressed in the vasculature of these species in a species- and vascular bed-dependent manner. This conclusion is in line with studies on the role of α1-AR subtypes in functional studies [3]. Our search did not identify any animal studies reporting on the regulation of vascular α1-AR subtype mRNA in CHF. However, it is likely that such regulation occurs because CHF is characterized by increased circulating catecholamine levels and the endogenous agonist noradrenaline has been shown to transiently down-regulate α1-AR mRNA in rabbit aortic smooth muscle cells whereas the down-regulation of α1-AR protein was longer lasting (at least 96 h) [95]. Moreover, CHF can be associated with hypoperfusion and hypoxia of tissues and vascular α1-AR mRNA expression can be regulated by hypoxia [96]. Vascular α1-AR mRNA expression is also regulated by age [97]. HF did not appear to affect the total levels of α1-AR mRNA in human coronary arteries [98]. On the contrary, the use of β-blockers was associated with a significant decrease in both α1D and total α1-AR mRNA levels. α1D-AR appeared to be the principal receptor in human epicardial coronary arteries, which resembles the mouse coronary arteries [99,100].

4.3.2. Protein Expression

Quantitative, ligand-binding studies have been performed in order to assess the α1-AR distribution in the various vascular beds of different species. Existing discrepancies might be due to interspecies differences, variant α1-AR subtype distribution in the peripheral vasculature, differences in models of HF, for example pacing-induced HF versus myocardial infarction, or even due to poorly reproducible results.
Such studies have been performed in animals and humans without coexisting diseases in order to assess the physiological α1-ARs at the protein level. In humans a considerable difference in α1-AR protein expression has been demonstrated between somatic (mammary artery) and splanchnic arteries [97]. However, this was not true for veins, in which the α1-AR density seemed to be independent from differences in vessel diameter. This might be explained by the importance of splanchnic circulation, especially during a situation of hypovolemic shock. Sympathetic stimulation of α1-ARs in those vascular territories leads to maintenance of blood flow and O2 delivery to vital organs. On the other hand, constriction of conduit arteries and veins contribute to the preservation of blood pressure and cardiac afterload. In human epicardial coronary arteries 8.7 fmol/mg protein of total α1-ARs were found, of which 75% constituted of the α1D subtype, almost twice as much as in the left ventricle [41,98].
Considering that progression of HF is associated with a decrease in β-AR protein in the heart, one could argue that similar findings would be expected for α1-ARs. On the contrary, α1-AR density has been reported to be increased, yet not statistically significant, in studies with myocardial infarction-induced CHF in rats [101,102]. However, the lack of statistical significance of this change leads to rather inconclusive results.

4.3.3. α1-AR Subtypes

All three subtypes have been identified in the peripheral vasculature of various species, however the subtype distribution varies between animals and humans, between different vascular beds and with age. For instance, α1B and α1D subtype mRNAs have been detected in human aorta [93] and are considered to mediate vascular constriction. A controversy exists in the rat aorta since some studies have identified only the subtype α1D [103,104,105], while others report both α1B and α1D-ARs [101]. The α1B subtype has been recognized to mediate vasoconstriction after sympathetic stimulation in the mouse, rabbit and dog aorta [106,107,108]. Similarly, mesenteric responses in humans have been found to be mediated by all three α1-AR subtypes [109], whereas in rats mesenteric vascular constriction is mediated by α1D and α1B subtypes in superior mesenteric artery and the mesenteric resistance arteries respectively [101,104]. In the small mesenteric arteries the subtype α1A has also been identified [101,110]. The same receptor was found to mediate contractile responses in rat small renal arteries [111].
Similarities in vascular tone regulation between different species also exist. As it was mentioned before, α1D is recognized as the main subtype that mediate coronary contraction in humans [98] and mice [99,100] and the same has also been reported in rats [105]. In this latter study, it was additionally shown that CHF did not modify the function of α1D-AR, neither in the carotid artery nor in the aorta, whereas the α1D-AR-mediated vasoconstriction was enhanced in older animals.
Furthermore, blood pressure regulation in mice is considered to be achieved via the stimulation of all three subtypes. This is confirmed in knockout mice studies in which deletion of any subtype results in a decrease in resting blood pressure [106,112,113].
The heterogeneous α1-AR population in the various vascular beds might be modified depending on the disease state. In support of this, antagonist studies have explicitly displayed an alteration of the antagonism against α1-ARs before and after the onset of CHF [114,115,116,117] and showed changes from competitive to non-competitive antagonism, or the other way around, which, however, was also related to the specific agonist and vessel studied. Distinct patterns of antagonism have been identified in different disease states and in different vessels. The specific subtype populations, however, and their involvement in the vascular responses in different disease settings are still to be elucidated.

4.3.4. Signaling Pathways Involved

Changes in α1-AR responsiveness due to chronic exposure of the peripheral vasculature to the endogenously released catecholamines might also derive from alterations in signal transduction. These changes might be due to modification of Ca2+ sensitivity and/or other changes in the signaling pathways involved. For example, the continuous exposure of smooth muscle cells from rabbit aorta to α1-AR agonists led to a major loss in sensitivity, which did not seem to be due to down-regulation of α1-ARs, but rather to blunting of phosphatidylinositol turnover [118].
Disease progression might also modify the dependency of α1-AR-mediated vascular contraction on extracellular Ca2+ which seems to increase in end-stage HF [119,120]. More precisely, the existence of a high affinity component of the α1-AR-mediated responses was identified which is inhibitable by a PKC inhibitor, polymyxin B, and which becomes apparent with the progression of HF. Interestingly, in this study, the dorsal pedal artery presented lower dependency on PKC activation than the saphenous vein, which underlines the existing differences between separate vascular beds [120]. Likewise, after investigating the α1-AR in mesenteric resistance arteries from CHF rats, it was confirmed that Ca2+ plays a more crucial role in the vascular contractility as CHF develops [121]. Furthermore, it has been demonstrated that the diminished contractile response of rat mesenteric resistance arteries to phenylephrine and Ca2+ is not due to changes in Ca2+ sensitivity, nor in intracellular Ca2+ release, but rather to changes in the α1-AR-triggered Ca2+ influx [102].
Other studies, however, have identified a modification in the sensitivity of α1-AR vasoconstriction to Ca2+. For instance, in CHF rats an increased responsiveness of CHF femoral resistance arteries was identified which appeared to be the result of first, a RhoA/Rho kinase-depended increase in sensitivity to Ca2+ and second, of the IP3-induced increase in intracellular Ca2+ [122]. On the contrary, this arterial sensitivity to Ca2+ was reported to be decreased in femoral resistance and muscle resistance arteries of CHF rats, which presented as a dissociation between the intracellular Ca2+ and the generated wall tension [123].

4.3.5. Cell and Tissue Responses

α1-ARs constitute an important homeostatic mechanism that contributes to the maintenance of peripheral vascular resistance at normal conditions and also in response to shock. Nevertheless, α1-AR regulation and function might alter due to disease. Even though HF is characterized by enhanced vascular resistance [124], there is little, and conflicting evidence to date concerning the vascular contractile responses to α1-AR stimulation in HF vessels.
The chronic sympathetic stimulation during HF might result in the desensitization of vascular α1-AR and consequently, in the failure of the contractile apparatus to function properly. Attenuation of α1-AR responsiveness after continuous exposure to catecholamines has been validated in various animal studies [125,126] but also in vitro [127,128]. However, despite the fact that several studies have tried to assess the α1-AR mediated vascular responses in both animals and humans with HF, the findings are not unanimous (Table 4). Multiple studies have shown an increase in vascular responsiveness to α1-AR stimulation in animals [114,115,116,117,119,120,122,129,130,131]. On the contrary, others have reported desensitization of α1-ARs in the peripheral vasculature in animals [102,121,132,133,134,135,136] and in humans [137,138,139,140], while numerous studies failed to demonstrate any alterations of the α1-AR responsiveness in vessels from animals [105,123,133,141,142,143] and humans [144].
Inconsistency in findings might originate from differences in the anatomy and function of distinct vascular beds. Additionally, differences in CHF model and disease state might also influence the results of different studies. For example, in some studies it was demonstrated that the saphenous vein of CHF dogs was at all times more sensitive than the dorsal pedal artery [114,115,116,120]. This might be associated to a heterogeneous smooth muscle function between the artery and the vein, but, also, to differences in receptor population in the canine vasculature [148]. In addition, differences in vascular responsiveness between different vascular beds were also identified in rat models of HF [123,130]. Moreover, differences have also been identified between the various arteries studied. For example, in similar models of pacing-induced CHF in dogs, some studies have demonstrated an increased responsiveness to α1-AR stimulation in the dorsal pedal artery and the contralateral saphenous vein [114,115,116,119], while others have identified a desensitization of α1-ARs in coronary arteries of CHF dogs [132].
Other important parameters that influence the peripheral vascular responses are the integrity and physiological function of endothelium. More specifically, the endothelium-induced vasodilation counteracts the contraction via α1-ARs. Therefore, the malfunction of the endothelium might lead to the absence of this inhibitory function in CHF vessels [143] and result in an enhancement of vascular contraction. Increased vascular responses to noradrenaline were reported in the intact thoracic aorta from HF rats compared to controls, however the actual vascular α1-AR responsiveness was diminished when the endothelium was removed [146]. On the other hand, an increase in endothelial function as was observed in other studies might oppose the α1-AR-mediated vascular contractions [131,132]. Others, however, did not demonstrate a difference in vascular responses between intact and denuded vessels [123].
Even though the vascular responsiveness in patients with HF has not been widely investigated, many studies have yielded data that might have important clinical implications. More precisely, Schwinn et al. [145] reported on the vascular responsiveness of HF patients that underwent coronary artery revascularization. The results from this study support that left ventricular dysfunction and more precisely, left ventricular ejection fraction equal to or lower than 40% is associated with a diminished vascular responsiveness during fentanyl anesthesia in this patient population. In contrast to more recent reports that identify cardiac surgery and cardiopulmonary bypass as predisposing factors for an attenuated vascular responsiveness [13,14,149], Schwinn et al. [145] reported that phenylephrine-induced changes in systemic vascular resistance were increased during cardiopulmonary bypass and aortic cross-clamp in both HF and control subjects. This vascular responsiveness is of interest in HF patients that undergo cardiac surgery, since this patient population exhibits an increased risk for post-operative vasoplegia [150]. This complication is characterized by low systemic vascular resistance, severe hypotension, normal or increased cardiac output and a blunted or absent response to the administration of vasopressors [151]. The diminished sensitivity of HF patients is thought to originate from the continuous, chronic exposure of these patients’ vascular α1-ARs to catecholamines which leads to desensitization and/or downregulation of those receptors. If vasoplegia is not promptly treated, the resulted tissue malperfusion might lead to end-organ dysfunction and multiple organ failure. Therefore, this post-operative complication significantly increases the risk of morbidity and mortality [152,153]. Moreover, another study identified a maladapted sympathetic response of the skeletal muscle vasculature to exercise that was characterized by sustained vascular constriction [140]. This can have serious implications on normal blood flow during physical movement and might hamper the patient’s daily life. Furthermore, the apparent up-regulation of α1-AR responsiveness after chronic carvedilol treatment in HF patients [147] and the lack of change after chronic angiotensin-converting enzyme inhibitors [142] might yield important consequences regarding HF therapy.

5. Discussion

Understanding the role of α1-ARs is particularly important in HF because this condition is associated with high adrenergic activation leading to down-regulation and desensitization of cardiac β-ARs. α1-ARs have been identified in both animal and human hearts and vasculature with a high expression in rodents but a much lower abundance in humans. Thus, caution should be applied while extrapolating data from animals to humans.
The α1A-AR is the most abundant subtype in the human heart and α1B-AR in the rodent heart, while α1D-ARs are mainly found in coronary arteries (Figure 3). However, studies on a change in the expression of the subtypes in failing hearts are inconsistent, which may indicate a relative α1/β-AR imbalance in light of the well-known reduction in β-adrenergic function in CHF.
Another important point is that, while in heart α1A- and α1B-ARs dominate, all three subtypes are expressed in the vasculature and their relative expression and physiological role appear to differ between vascular beds and species (Figure 3) [3]. This varied expression of α1-AR has been reported to correlate with the adrenergic innervation in peripheral vessels [154]. Moreover, studies with transfected receptors suggest that the three α1-AR subtypes undergo differential regulation upon exposure to agonists [16]. Certainly, those discrepancies also affect the way in which blood pressure is regulated depending on which α1-AR subtypes play the predominant role. For that reason, the differences in subtype distribution might also have influenced studies that have investigated the function of α1-ARs in vivo or in vitro, leading to conflicting results.
α1-ARs can couple to multiple intracellular signal transduction pathways in the heart. Under most conditions α1-AR signal transduction results in enhanced inotropy. Although data on their role in HF is limited, there is evidence indicating that they mediate enhanced positive inotropy, as well as physiological hypertrophy and decreased apoptosis [46]. α1-AR occupancy was estimated as 10% in failing human heart [155]. Thus, activation of unoccupied receptors with an exogenous ligand is suggested to be cardioprotective [46]. Clinical trials such as ALLHAT in hypertension and V-HeFT in CHF reported that treatment with α1-AR antagonists was associated with greater mortality and apoptosis than with other treatments [156]. Moreover, double knockout of α1A- and α1B-AR was shown to augment apoptosis, worsen cardiomyopathy and reduce survival [157]. Taken together, cardiac α1-ARs appear to mediate cardioprotective effects. Data on the regulation of vascular α1-ARs in HF are also conflicting. This is unfortunate because of their role in controlling total peripheral resistance and the role of afterload reductions in the treatment of patients with HF. However, clinical studies in hypertensive patients have shown that α1-AR antagonism provides less protection from HF than other classes of anti-hypertensive drugs [156]. More specifically, the non-selective α-blocker doxazosin was associated with significantly higher risk of HF compared with the other treatment arms of this trial. In the absence of a placebo arm, it remained unclear whether this reflects an adverse effect of doxazosin or less protection against hypertension-induced HF. The results from the V-HeFT trial [158] further support the detrimental effects of α-blocking therapy-, since prazosin use gave a signal for possibly increased mortality that failed to reach statistical significance. The reason for those results could possibly be explained by the findings from knock-out mice studies which confirm the importance of α1-AR signaling for the adaptational mechanisms of cardiomyocytes in disease settings [157,159].
In contrast to those studies that investigated the effect of selective α-blockade and discouraged their widespread use, the mixed β- and α1-AR antagonist carvedilol was demonstrated to be more beneficial than the β1-selective antagonist metoprolol in diminishing the risk of mortality [155]. It could, therefore, be postulated that the favorable effects of carvedilol are to some extent related to its α-blocking properties. Nevertheless, the combination of an α- with a β-blocker for long-term treatment in chronic CHF patients, did not yield better clinical outcomes than β-blocker therapy alone [160], which highlights the fact that such a combined therapy does not generate satisfactory outcomes similar to that of carvedilol.
The exact reason for those discrepancies in effectiveness still remains unclear. One explanation could be that carvedilol’s α-blocking properties are less potent compared to both its innate β-blocking feature and to the α-blockade produced by a specific α-AR antagonist like prazosin. This is in fact supported by the results of a small trial in which carvedilol did not antagonize phenylephrine induced responses [161]. Another hypothesis is that the existence of distinct pharmacologic properties and mechanisms of action between the various α- and β-blockers could also lead to the occurrence of different side effects. Based on this assumption, one could possibly explain the contradictory findings from studies with carvedilol versus studies investigating the combination of an α- with a β-blocker. Last but not least, carvedilol use leads to pharmacologic tolerance after long term use [162]. This implies that its beneficial effects at long-term use derive mostly from its β-blocking properties rather than antagonizing α1-ARs.
In brief, one could state that α1-AR agonism could potentially be a valid treatment option for HF patients that would stimulate innate adaptive signaling procedures and would eventually illustrate its cardioprotective features. However, caution is required, since α1 agonism could also have detrimental effects by increasing peripheral vascular resistance leading to an increased cardiac afterload and by inducing coronary vasoconstriction in those patients. Therefore, future research is necessary which should focus on compartment and/or subtype selective treatment in HF patients and which will investigate in more detail the treatment modalities of α1 agonism.
On the other hand, carvedilol seems promising for the treatment of HF patients and might be an option for the pretreatment of HF patients that will undergo cardiac surgery. Short term α1 antagonism with carvedilol treatment could shield α1-ARs against continuous catecholamine exposure in the preoperative period. This could perhaps increase the sensitivity of those receptors to the administration of catecholamines for the treatment of vasoplegia during the postoperative course [147].
In conclusion, more research is required to further understand the complex mechanisms of α1 adrenergic function and adaptation in HF. Taking into account the existing differences between animals and humans regarding the expression of α1-ARs, we should carefully consider our next steps and we should promote additional research which would focus more on human tissue (heart and vessels). Moreover, a cardioselective approach in the case of α1 agonism appears to be crucial for the exploitation of only its advantageous characteristics by avoiding potential vascular side effects. A subtype approach could also be beneficial but further studies are necessary in order to gain more insight in such treatment techniques. Finally, in view of the positive outcomes from clinical studies with carvedilol, more extensive investigation is to be expected so as to better comprehend the pharmacologic properties and mechanism of action of this agent. It is, therefore, essential that we expand our knowledge, and we apprehend current inconsistencies in order to pave the way to safer and more effective therapies.

Supplementary Materials

The full search strategy is available online at https://www.mdpi.com/2073-4409/9/11/2412/s1.

Author Contributions

Conceptualization, M.C.M. and E.A.-I.; methodology, G.K.-M. and O.P.; validation, M.C.M., E.A.-I., A.H.J.D. and M.P.; investigation, G.K.-M. and O.P.; writing—original draft preparation, G.K.-M. and O.P.; writing—review and editing, M.C.M., E.A.-I., M.P. and A.H.J.D.; visualization, G.K.-M., O.P. and M.C.M.; supervision, M.C.M., M.P., A.H.J.D., E.A.-I.; project administration, M.C.M.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We would like to thank Jan W. Schoones for assisting with the drafting of the full search strategy for this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brodde, O.E.; Michel, M.C. Adrenergic and muscarinic receptors in the human heart. Pharmacol. Rev. 1999, 51, 651–689. [Google Scholar]
  2. Brattelid, T.; Tveit, K.; Birkeland, J.A.; Sjaastad, I.; Qvigstad, E.; Krobert, K.A.; Hussain, R.I.; Skomedal, T.; Osnes, J.B.; Levy, F.O. Expression of mRNA encoding G protein-coupled receptors involved in congestive heart failure; A quantitative RT-PCR study and the question of normalisation. Basic Res. Cardiol. 2007, 102, 198–208. [Google Scholar]
  3. Guimaraes, S.; Moura, D. Vascular adrenoceptors: An update. Pharmacol. Rev. 2001, 53, 319–356. [Google Scholar]
  4. Puceat, M.; Terzic, A.; Clement, O.; Scamps, F.; Vogel, S.M.; Vassort, G. Cardiac α1-adrenoceptors mediate positive inotropy via myofibrillar sensitization. Trends Pharmacol. Sci. 1992, 13, 263–265. [Google Scholar] [PubMed]
  5. Steinberg, S.F.; Drugge, E.D.; Bilezikian, J.P.; Robinson, R.B. Acquisition by innervated cardiac myocytes of a pertussis toxin-specific regulatory protein linked to the alpha 1-receptor. Science 1985, 230, 186–188. [Google Scholar]
  6. Li, K.; He, H.; Li, C.; Sirois, P.; Rouleau, J.L. Myocardial α1-adrenoceptor: Inotropic effect and physiologic and pathologic implications. Life Sci. 1997, 60, 1305–1318. [Google Scholar] [PubMed]
  7. Anyukhovsky, E.P.; Rybin, V.O.; Nikashin, A.V.; Budanova, O.P.; Rosen, M.R. Positive chronotropic responses induced by alpha 1-adrenergic stimulation of normal and “ischemic” Purkinje fibers have different receptor-effector coupling mechanisms. Circ. Res. 1992, 71, 526–534. [Google Scholar]
  8. Knowlton, K.U.; Michel, M.C.; Itani, M.; Shubeita, H.E.; Ishihara, K.; Brown, J.H.; Chien, K.R. The α1A-adrenergic receptor subtype mediates biochemical, molecular, and morphologic features of cultured myocardial cell hypertrophy. J. Biol. Chem. 1993, 268, 15374–15380. [Google Scholar] [PubMed]
  9. Dal Monte, M.; Evans, B.A.; Arioglu-Inan, E.; Michel, M.C. Upregulation of β3-adrenoceptors—A general marker of and protective mechanism against hypoxia? Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 141–146. [Google Scholar]
  10. Cohn, J.N. Abnormalities of peripheral sympathetic nervous system control in congestive heart failure. Circulation 1990, 82, 59–67. [Google Scholar]
  11. Maisel, A.S.; Scott, N.A.; Motulsky, H.J.; Michel, M.C.; Boublik, J.H.; Rivier, J.E.; Ziegler, M.; Allen, R.S.; Brown, M.R. Elevation of plasma neuropeptide Y levels in congestive heart failure. Am. J. Med. 1989, 86, 43–48. [Google Scholar] [CrossRef]
  12. Cohn, J.N.; Levine, T.B.; Olivari, M.T.; Garberg, V.; Lura, D.; Francis, G.S.; Simon, A.B.; Rector, T. Plasma Norepinephrine as a Guide to Prognosis in Patients with Chronic Congestive Heart Failure. N. Engl. J. Med. 1984, 311, 819–823. [Google Scholar] [CrossRef] [Green Version]
  13. Carrel, T.; Englberger, L.; Mohacsi, P.; Neidhart, P.; Schmidli, J. Low systemic vascular resistance after cardiopulmonary bypass: Incidence, etiology, and clinical importance. J. Card. Surg. 2000, 15, 347–353. [Google Scholar] [CrossRef]
  14. Sun, X.; Zhang, L.; Hill, P.C.; Lowery, R.; Lee, A.T.; Molyneaux, R.E.; Corso, P.J.; Boyce, S.W. Is incidence of postoperative vasoplegic syndrome different between off-pump and on-pump coronary artery bypass grafting surgery? Eur. J. Cardiothorac. Surg. 2008, 34, 820–825. [Google Scholar] [CrossRef]
  15. Michel, M.C.; Kenny, B.; Schwinn, D.A. Classification of alpha 1-adrenoceptor subtypes. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1995, 352, 1–10. [Google Scholar] [CrossRef] [PubMed]
  16. Yang, M.; Ruan, J.; Voller, M.; Schalken, J.; Michel, M.C. Differential regulation of human alpha1-adrenoceptor subtypes. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1999, 359, 439–446. [Google Scholar] [CrossRef]
  17. Monto, F.; Oliver, E.; Vicente, D.; Rueda, J.; Aguero, J.; Almenar, L.; Ivorra, M.D.; Barettino, D.; D’Ocon, P. Different expression of adrenoceptors and GRKs in the human myocardium depends on heart failure ethiology and correlates to clinical variables. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, 368–376. [Google Scholar] [CrossRef] [Green Version]
  18. Hendriks-Balk, M.C.; Michel, M.C.; Alewijnse, A.E. Pitfalls in the normalization of real-time polymerase chain reaction data. Basic Res. Cardiol. 2007, 102, 195–197. [Google Scholar] [CrossRef] [Green Version]
  19. Michel-Reher, M.B.; Michel, M.C. Regulation of GAPDH expression by treatment with the ß-adrenoceptor agonist isoprenaline—Is GAPDH a suitable loading control in immunoblot experiments? Naunyn-Schmiedeberg’s Arch. Pharmacol. 2015, 388, 1119–1120. [Google Scholar] [CrossRef]
  20. Pradidarcheep, W.; Stallen, J.; Labruyere, W.T.; Dabhoiwala, N.F.; Michel, M.C.; Lamers, W.H. Lack of specificity of commercially available antisera against muscarinic and adrenergic receptors. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2009, 379, 397–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Jensen, B.C.; Swigart, P.M.; Simpson, P.C. Ten commercial antibodies for alpha-1-adrenergic receptor subtypes are nonspecific. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2009, 379, 409–412. [Google Scholar] [CrossRef] [Green Version]
  22. Böhmer, T.; Pfeiffer, N.; Gericke, A. Three commerical antibodies against α1-adrenergic receptor subtypes lack specificity in paraffin-embedded sections of murine tissues. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2014, 387, 703–706. [Google Scholar] [CrossRef]
  23. McGrath, J.C. Localization of α-adrenoceptors: JR Vane Medal Lecture. Br. J. Pharmacol. 2015, 172, 1179–1194. [Google Scholar] [CrossRef] [Green Version]
  24. Faber, J.E.; Yang, N.; Xin, X. Expression of α-Adrenoceptor Subtypes by Smooth Muscle Cells and Adventitial Fibroblasts in Rat Aorta and in Cell Culture. J. Pharmacol. Exp. Ther. 2001, 298, 441. [Google Scholar]
  25. Litwin, S.E.; Vatner, D.E.; Morgan, J.P. Inotropic effects of alpha 1-adrenergic agonists in myocardium from rats with postinfarction heart failure. Am. J. Physiol. 1995, 269, H1553–H1563. [Google Scholar] [CrossRef]
  26. Meggs, L.G.; Tillotson, J.; Huang, H.; Sonnenblick, E.H.; Capasso, J.M.; Anversa, P. Noncoordinate regulation of alpha-1 adrenoceptor coupling and reexpression of alpha skeletal actin in myocardial infarction-induced left ventricular failure in rats. J. Clin. Investig. 1990, 86, 1451–1458. [Google Scholar] [CrossRef] [Green Version]
  27. Wang, G.Y.; Yeh, C.C.; Jensen, B.C.; Mann, M.J.; Simpson, P.C.; Baker, A.J. Heart failure switches the RV alpha1-adrenergic inotropic response from negative to positive. Am. J. Physiol. Heart Circ. Physiol. 2010, 298, H913–H920. [Google Scholar] [CrossRef] [Green Version]
  28. Calderone, A.; Bouvier, M.; Li, K.; Juneau, C.; de Champlain, J.; Rouleau, J.L. Dysfunction of the beta- and alpha-adrenergic systems in a model of congestive heart failure. The pacing-overdrive dog. Circ. Res. 1991, 69, 332–343. [Google Scholar] [CrossRef] [Green Version]
  29. Cowley, P.M.; Wang, G.; Chang, A.N.; Makwana, O.; Swigart, P.M.; Lovett, D.H.; Stull, J.T.; Simpson, P.C.; Baker, A.J. The α1A-adrenergic receptor subtype mediates increased contraction of failing right ventricular myocardium. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H888–H896. [Google Scholar] [CrossRef] [Green Version]
  30. Zhao, M.; Hagler, H.K.; Muntz, K.H. Regulation of alpha 1-, beta 1-, and beta 2-adrenergic receptors in rat heart by norepinephrine. Am. J. Physiol. 1996, 271, H1762–H1768. [Google Scholar] [CrossRef] [PubMed]
  31. Eschenhagen, T.; Diederich, M.; Kluge, S.H.; Magnussen, O.; Mene, U.; Muller, F.; Schmitz, W.; Scholz, H.; Weil, J.; Sent, U.; et al. Bovine hereditary cardiomyopathy: An animal model of human dilated cardiomyopathy. J. Mol. Cell Cardiol. 1995, 27, 357–370. [Google Scholar] [CrossRef]
  32. Gopalakrishnan, M.; Triggle, D.J.; Rutledge, A.; Kwon, Y.W.; Bauer, J.A.; Fung, H.L. Regulation of K+ and Ca2+ channels in experimental cardiac failure. Am. J. Physiol. 1991, 261, H1979–H1987. [Google Scholar] [CrossRef]
  33. Karliner, J.S.; Barnes, P.; Brown, M.; Dollery, C. Chronic heart failure in the guinea pig increases cardiac alpha 1- and beta-adrenoceptors. Eur. J. Pharmacol. 1980, 67, 115–118. [Google Scholar] [CrossRef]
  34. Maisel, A.S.; Motulsky, H.J.; Ziegler, M.G.; Insel, P.A. Ischemia- and agonist-induced changes in alpha- and beta-adrenergic receptor traffic in guinea pig hearts. Am. J. Physiol. Heart Circ. Physiol. 1987, 253, H1159–H1166. [Google Scholar] [CrossRef]
  35. Watanabe, K.; Shibata, A.; Wakabayashi, H.; Shimada, K.; Sasaki, K.; Nagatomo, T. Cardiomyopathic hamster hearts: Long-term effects of drugs on catecholamine contents and binding characteristics of alpha 1- and beta 1-adrenergic receptors. Biol. Pharm. Bull. 1993, 16, 660–663. [Google Scholar] [CrossRef] [Green Version]
  36. Corr, P.B.; Shayman, J.A.; Kramer, J.B.; Kipnis, R.J. Increased alpha-adrenergic receptors in ischemic cat myocardium. A potential mediator of electrophysiological derangements. J. Clin. Investig. 1981, 67, 1232–1236. [Google Scholar] [CrossRef] [PubMed]
  37. Re, G.; Bergamasco, L.; Badino, P.; Borgarelli, M.; Odore, R.; Tarducci, A.; Zanatta, R.; Girardi, C. Canine dilated cardiomyopathy: Lymphocyte and cardiac α1- and β-adrenoceptor concentrations in normal and affected great danes. Vet. J. 1999, 158, 120–127. [Google Scholar] [CrossRef]
  38. Suematsu, N.; Satoh, S.; Kinugawa, S.; Tsutsui, H.; Hayashidani, S.; Nakamura, R.; Egashira, K.; Makino, N.; Takeshita, A. Alpha1-adrenoceptor-Gq-RhoA signaling is upregulated to increase myofibrillar Ca2+ sensitivity in failing hearts. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, H637–H646. [Google Scholar] [CrossRef]
  39. Brodde, O.E.; Leineweber, K. Autonomic receptor systems in the failing and aging human heart: Similarities and differences. Eur. J. Pharmacol. 2004, 500, 167–176. [Google Scholar] [CrossRef]
  40. Jensen, B.C.; O’Connell, T.D.; Simpson, P.C. Alpha-1-adrenergic receptors in heart failure: The adaptive arm of the cardiac response to chronic catecholamine stimulation. J. Cardiovasc. Pharmacol. 2014, 63, 291–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Jensen, B.C.; Swigart, P.M.; de Marco, T.; Hoopes, C.; Simpson, P.C. α1-Adrenergic receptor subtypes in nonfailing and failing human myocardium. Circ. Heart Fail. 2009, 2, 654–663. [Google Scholar] [CrossRef] [Green Version]
  42. Myagmar, B.E.; Flynn, J.M.; Cowley, P.M.; Swigart, P.M.; Montgomery, M.D.; Thai, K.; Nair, D.; Gupta, R.; Deng, D.X.; Hosoda, C.; et al. Adrenergic Receptors in Individual Ventricular Myocytes: The Beta-1 and Alpha-1B are in All Cells, the Alpha-1A is in a Subpopulation, and the Beta-2 and Beta-3 are Mostly Absent. Circ. Res. 2017, 120, 1103–1115. [Google Scholar] [CrossRef] [Green Version]
  43. Fischer, V.; Gabauer, I.; Tillinger, A.; Novakova, M.; Pechan, I.; Krizanova, O.; Kvetnansky, R.; Myslivecek, J. Heart adrenoceptor gene expression and binding sites in the human failing heart. Ann. N. Y. Acad. Sci. 2008, 1148, 400–408. [Google Scholar] [CrossRef]
  44. Williams, R.S.; Lefkowitz, R.J. Alpha-adrenergic receptors in rat myocardium. Identification by binding of [3H]dihydroergocryptine. Circ. Res. 1978, 43, 721–727. [Google Scholar] [CrossRef] [Green Version]
  45. Hwang, K.C.; Gray, C.D.; Sweet, W.E.; Moravec, C.S.; Im, M.J. Alpha 1-adrenergic receptor coupling with Gh in the failing human heart. Circulation 1996, 94, 718–726. [Google Scholar] [CrossRef]
  46. Steinfath, M.; Danielsen, W.; von der Leyen, H.; Mende, U.; Meyer, W.; Neumann, J.; Nose, M.; Reich, T.; Schmitz, W.; Scholz, H.; et al. Reduced alpha 1- and beta 2-adrenoceptor-mediated positive inotropic effects in human end-stage heart failure. Br. J. Pharmacol. 1992, 105, 463–469. [Google Scholar] [CrossRef]
  47. Vago, T.; Bevilacqua, M.; Norbiato, G.; Baldi, G.; Chebat, E.; Bertora, P.; Baroldi, G.; Accinni, R. 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. 1989, 64, 474–481. [Google Scholar] [CrossRef] [Green Version]
  48. Grigore, A.; Poindexter, B.; Vaughn, W.K.; Nussmeier, N.; Frazier, O.H.; Cooper, J.R.; Gregoric, I.D.; Buja, L.M.; Bick, R.J. Alterations in alpha adrenoreceptor density and localization after mechanical left ventricular unloading with the Jarvik flowmaker left ventricular assist device. J. Heart Lung Transplant. 2005, 24, 609–613. [Google Scholar] [CrossRef]
  49. Bohm, M.; Diet, F.; Feiler, G.; Kemkes, B.; Erdmann, E. Alpha-adrenoceptors and alpha-adrenoceptor-mediated positive inotropic effects in failing human myocardium. J. Cardiovasc. Pharmacol. 1988, 12, 357–364. [Google Scholar] [CrossRef]
  50. Bristow, M.R.; Minobe, W.; Rasmussen, R.; Hershberger, R.E.; Hoffman, B.B. Alpha-1 adrenergic receptors in the nonfailing and failing human heart. J. Pharmacol. Exp. Ther. 1988, 247, 1039–1045. [Google Scholar]
  51. Erdmann, E.; Bohm, M. Positive inotropic stimulation in the normal and insufficient human myocardium. Basic Res. Cardiol. 1989, 84, 125–133. [Google Scholar] [CrossRef]
  52. Limas, C.J.; Limas, C.; Goldenberg, I.F. Intracellular distribution of adrenoceptors in the failing human myocardium. Am. Heart J. 1989, 117, 1310–1316. [Google Scholar] [CrossRef]
  53. Shi, T.; Moravec, C.S.; Perez, D.M. Novel proteins associated with human dilated cardiomyopathy: Selective reduction in α1A-adrenergic receptors and increased desensitization proteins. J. Recept. Signal Transduct. 2013, 33, 96–106. [Google Scholar] [CrossRef] [Green Version]
  54. Dixon, I.M.C.; Dhalla, N.S. Alterations in cardiac adrenoceptors in congestive heart failure secondary to myocardial infarction. Coron. Artery Dis. 1991, 2, 805–814. [Google Scholar]
  55. Sjaastad, I.; Schiander, I.; Sjetnan, A.; Qvigstad, E.; Bokenes, J.; Sandnes, D.; Osnes, J.B.; Sejersted, O.M.; Skomedal, T. Increased contribution of α1- vs. β-adrenoceptor-mediated inotropic response in rats with congestive heart failure. Acta Physiol. Scand. 2003, 177, 449–458. [Google Scholar] [CrossRef]
  56. Rowley, K.G.; Tung, L.H.; Hodsman, G.P.; Howes, L.G.; Jarrott, B.; Beart, P.M.; Louis, W.J. Altered α1-Adrenoceptor–Mediated Responses in Atria of Rats with Chronic Left Ventricular Infarction. J. Cardiovasc. Pharmacol. 1991, 17, 474–479. [Google Scholar] [CrossRef]
  57. O’Connell, T.D.; Jensen, B.C.; Baker, A.J.; Simpson, P.C. Cardiac alpha1-adrenergic receptors: Novel aspects of expression, signaling mechanisms, physiologic function, and clinical importance. Pharmacol. Rev. 2014, 66, 308–333. [Google Scholar] [CrossRef] [Green Version]
  58. Yang, M.; Reese, J.; Cotecchia, S.; Michel, M.C. Murine alpha1-adrenoceptor subtypes. I. Radioligand binding studies. J. Pharmacol. Exp. Ther. 1998, 286, 841–847. [Google Scholar]
  59. Michel, M.C.; Hanft, G.; Gross, G. Radioligand binding studies of alpha 1-adrenoceptor subtypes in rat heart. Br. J. Pharmacol. 1994, 111, 533–538. [Google Scholar] [CrossRef] [PubMed]
  60. Janssen, P.M.L.; Canan, B.D.; Kilic, A.; Whitson, B.A.; Baker, A.J. Human Myocardium Has a Robust alpha1A-Subtype Adrenergic Receptor Inotropic Response. J. Cardiovasc. Pharmacol. 2018, 72, 136–142. [Google Scholar] [CrossRef]
  61. Du, X.J.; Gao, X.M.; Kiriazis, H.; Moore, X.L.; Ming, Z.; Su, Y.; Finch, A.M.; Hannan, R.A.; Dart, A.M.; Graham, R.M. Transgenic α1A-adrenergic activation limits post-infarct ventricular remodeling and dysfunction and improves survival. Cardiovasc. Res. 2006, 71, 735–743. [Google Scholar] [CrossRef] [Green Version]
  62. Schnee, P.M.; Shah, N.; Bergheim, M.; Poindexter, B.J.; Buja, L.M.; Gemmato, C.; Radovancevic, B.; Letsou, G.V.; Frazier, O.H.; Bick, R.J. Location and density of of α- and β-adrenoreceptor sub-types in myocardium after mechanical left ventricular unloading. J. Heart Lung Transplant. 2008, 27, 710–717. [Google Scholar] [CrossRef]
  63. Hieble, J.P.; Bylund, D.B.; Clarke, D.E.; Eikenburg, D.C.; Langer, S.Z.; Lefkowitz, R.J.; Minneman, K.P.; Ruffolo, R.R., Jr. International Union of Pharmacology. X. Recommendation for nomenclature of alpha 1-adrenoceptors: Consensus update. Pharmacol. Rev. 1995, 47, 267–270. [Google Scholar]
  64. Maruyama, Y.; Nishida, M.; Sugimoto, Y.; Tanabe, S.; Turner, J.H.; Kozasa, T.; Wada, T.; Nagao, T.; Kurose, H. Gα12/13 Mediates α1-Adrenergic receptor-induced cardiac hypertrophy. Circ. Res. 2002, 91, 961–969. [Google Scholar] [CrossRef] [Green Version]
  65. Pönicke, K.; Heinroth-Hoffmann, I.; Brodde, O.E. Role of β1- and β2-adrenoceptors in hypertrophic and apoptotic effects of noradrenaline and adrenaline in adult rat ventricular cardiomyocytes. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2003, 367, 592–599. [Google Scholar] [CrossRef] [PubMed]
  66. Baek, K.J.; Das, T.; Gray, C.; Antar, S.; Murugesan, G.; Im, M.J. Evidence that the Gh protein is a signal mediator from alpha 1-adrenoceptor to a phospholipase C. I. Identification of alpha 1-adrenoceptor-coupled Gh family and purification of Gh7 from bovine heart. J. Biol. Chem. 1993, 268, 27390–27397. [Google Scholar] [PubMed]
  67. Barrett, S.; Honbo, N.; Karliner, J.S. Alpha 1-adrenoceptor-mediated inhibition of cellular cAMP accumulation in neonatal rat ventricular myocytes. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1993, 347, 384–393. [Google Scholar] [CrossRef]
  68. Hool, L.C.; Oleksa, L.M.; Harvey, R.D. Role of G proteins in alpha1-adrenergic inhibition of the beta-adrenergically activated chloride current in cardiac myocytes. Mol. Pharmacol. 1997, 51, 853–860. [Google Scholar] [CrossRef] [PubMed]
  69. Liu, Q.Y.; Karpinski, E.; Pang, P.K. L-channel modulation by alpha-1 adrenoceptor activation in neonatal rat ventricular cells: Intracellular mechanisms. J. Pharmacol. Exp. Ther. 1994, 271, 944–951. [Google Scholar]
  70. LaMorte, V.J.; Thorburn, J.; Absher, D.; Spiegel, A.; Brown, J.H.; Chien, K.R.; Feramisco, J.R.; Knowlton, K.U. Gq- and ras-dependent pathways mediate hypertrophy of neonatal rat ventricular myocytes following alpha 1-adrenergic stimulation. J. Biol. Chem. 1994, 269, 13490–13496. [Google Scholar]
  71. Bogoyevitch, M.A.; Clerk, A.; Sugden, P.H. Activation of the mitogen-activated protein kinase cascade by pertussis toxin-sensitive and -insensitive pathways in cultured ventricular cardiomyocytes. Biochem. J. 1995, 309, 437–443. [Google Scholar] [CrossRef] [Green Version]
  72. Jensen, B.C.; O’Connell, T.D.; Simpson, P.C. Alpha-1-adrenergic receptors: Targets for agonist drugs to treat heart failure. J. Mol. Cell Cardiol. 2011, 51, 518–528. [Google Scholar] [CrossRef] [Green Version]
  73. Salazar, N.C.; Chen, J.; Rockman, H.A. Cardiac GPCRs: GPCR signaling in healthy and failing hearts. Biochim. Biophys. Acta 2007, 1768, 1006–1018. [Google Scholar] [CrossRef] [Green Version]
  74. D’Angelo, D.D.; Sakata, Y.; Lorenz, J.N.; Boivin, G.P.; Walsh, R.A.; Liggett, S.B.; Dorn, G.W., II. Transgenic Galphaq overexpression induces cardiac contractile failure in mice. Proc. Natl. Acad. Sci. USA 1997, 94, 8121–8126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Adams, J.W.; Sakata, Y.; Davis, M.G.; Sah, V.P.; Wang, Y.; Liggett, S.B.; Chien, K.R.; Brown, J.H.; Dorn, G.W., II. Enhanced Galphaq signaling: A common pathway mediates cardiac hypertrophy and apoptotic heart failure. Proc. Natl. Acad. Sci. USA 1998, 95, 10140–10145. [Google Scholar] [CrossRef] [Green Version]
  76. Akhter, S.A.; Luttrell, L.M.; Rockman, H.A.; Iaccarino, G.; Lefkowitz, R.J.; Koch, W.J. Targeting the receptor-Gq interface to inhibit in vivo pressure overload myocardial hypertrophy. Science 1998, 280, 574–577. [Google Scholar] [CrossRef]
  77. Esposito, G.; Rapacciuolo, A.; Naga Prasad, S.V.; Takaoka, H.; Thomas, S.A.; Koch, W.J.; Rockman, H.A. Genetic alterations that inhibit in vivo pressure-overload hypertrophy prevent cardiac dysfunction despite increased wall stress. Circulation 2002, 105, 85–92. [Google Scholar] [CrossRef] [Green Version]
  78. Lohse, M.J.; Engelhardt, S.; Eschenhagen, T. What is the role of beta-adrenergic signaling in heart failure? Circ. Res. 2003, 93, 896–906. [Google Scholar] [CrossRef] [PubMed]
  79. Michel, M.C.; Hanft, G.; Gross, G. Functional studies on α1-adrenoceptor subtypes mediating inotropic effects in rat right ventricle. Br. J. Pharmacol. 1994, 111, 539–546. [Google Scholar] [CrossRef]
  80. Barcroft, H.; Konzett, H. On the actions of noradrenaline, adrenaline and isopropyl noradrenaline on the arterial blood pressure, heart rate and muscle blood flow in man. J. Physiol. 1949, 110, 194–204. [Google Scholar] [CrossRef] [Green Version]
  81. Skomedal, T.; Borthne, K.; Aass, H.; Geiran, O.; Osnes, J.B. Comparison between alpha-1 adrenoceptor-mediated and beta adrenoceptor-mediated inotropic components elicited by norepinephrine in failing human ventricular muscle. J. Pharmacol. Exp. Ther. 1997, 280, 721–729. [Google Scholar]
  82. Sen, L.; Liang, B.T.; Colucci, W.S.; Smith, T.W. Enhanced alpha 1-adrenergic responsiveness in cardiomyopathic hamster cardiac myocytes. Relation to the expression of pertussis toxin-sensitive G protein and alpha 1-adrenergic receptors. Circ. Res. 1990, 67, 1182–1192. [Google Scholar] [CrossRef] [Green Version]
  83. Woodcock, E.A.; Du, X.J.; Reichelt, M.E.; Graham, R.M. Cardiac alpha 1-adrenergic drive in pathological remodelling. Cardiovasc. Res. 2008, 77, 452–462. [Google Scholar] [CrossRef] [Green Version]
  84. Perez, D.M.; Piascik, M.T.; Malik, N.; Gaivin, R.; Graham, R.M. Cloning, expression, and tissue distribution of the rat homolog of the bovine alpha 1C-adrenergic receptor provide evidence for its classification as the alpha 1A subtype. Mol. Pharmacol. 1994, 46, 823. [Google Scholar]
  85. Piascik, M.T.; Smith, M.S.; Soltis, E.E.; Perez, D.M. Identification of the mRNA for the novel alpha 1D-adrenoceptor and two other alpha 1-adrenoceptors in vascular smooth muscle. Mol. Pharmacol. 1994, 46, 30. [Google Scholar]
  86. Ping, P.; Faber, J.E. Characterization of alpha-adrenoceptor gene expression in arterial and venous smooth muscle. Am. J. Physiol. Heart Circ. Physiol. 1993, 265, H1501–H1509. [Google Scholar] [CrossRef]
  87. Price, D.T.; Chari, R.S.; Berkowitz, D.E.; Meyers, W.C.; Schwinn, D.A. Expression of alpha 1-adrenergic receptor subtype mRNA in rat tissues and human SK-N-MC neuronal cells: Implications for alpha 1-adrenergic receptor subtype classification. Mol. Pharmacol. 1994, 46, 221. [Google Scholar] [PubMed]
  88. Kaiming, X.; Chide, H. Quantification of mRNAS for three α1-adrenoceptor subtypes in rat aorta by solution hybridization. Life Sci. 1996, 59, PL343–PL347. [Google Scholar] [CrossRef]
  89. Diehl, N.L.; Martin Shreeve, S. Identification of the α1C-adrenoceptor in rabbit arteries and the human saphenous vein using the polymerase chain reaction. Eur. J. Pharmacol. 1994, 268, 393–398. [Google Scholar] [CrossRef]
  90. Miyamoto, S.; Taniguchi, T.; Suzuki, F.; Takita, M.; Kosaka, N.; Negoro, E.; Okuda, T.; Kosaka, H.; Murata, S.; Nakamura, S.; et al. Cloning, functional expression and tissue distribution of rabbit αla-adrenoceptor. Life Sci. 1997, 60, 2069–2074. [Google Scholar] [CrossRef]
  91. Suzuki, F.; Miyamoto, S.; Takita, M.; Oshita, M.; Watanabe, Y.; Kakizuka, A.; Narumiya, S.; Taniguchi, T.; Muramatsu, I. Cloning, functional expression and tissue distribution of rabbit α1d-adrenoceptor1The nucleotide sequence reported in this paper has been submitted to the GenBank under the accession number U64032.1. Biochim. Biophys. Acta Biomembr. 1997, 1323, 6–11. [Google Scholar] [CrossRef] [Green Version]
  92. Faure, C.; Gouhier, C.; Langer, S.Z.; Graham, D. Quantification of α1-Adrenoceptor Subtypes in Human Tissues by Competitive RT-PCR Analysis. Biochem. Biophys. Res. Commun. 1995, 213, 935–943. [Google Scholar] [CrossRef]
  93. Price, D.T.; Lefkowitz, R.J.; Caron, M.G.; Berkowitz, D.; Schwinn, D.A. Localization of mRNA for three distinct alpha 1-adrenergic receptor subtypes in human tissues: Implications for human alpha-adrenergic physiology. Mol. Pharmacol. 1994, 45, 171. [Google Scholar]
  94. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  95. Izzo, N.J., Jr.; Seidman, C.E.; Collins, S.; Colucci, W.S. Alpha 1-adrenergic receptor mRNA level is regulated by norepinephrine in rabbit aortic smooth muscle cells. Proc. Natl. Acad. Sci. USA 1990, 87, 6268–6271. [Google Scholar] [CrossRef] [Green Version]
  96. Eckhart, A.D.; Zhu, Z.; Arendshorst, W.J.; Faber, J.E. Oxygen modulates alpha 1B-adrenergic receptor gene expression by arterial but not venous vascular smooth muscle. Am. J. Physiol. Heart Circ. Physiol. 1996, 271, H1599–H1608. [Google Scholar] [CrossRef]
  97. Rudner, X.L.; Berkowitz, D.E.; Booth, J.V.; Funk, B.L.; Cozart, K.L.; D’Amico, E.B.; El-Moalem, H.; Page, S.O.; Richardson, C.D.; Winters, B.; et al. Subtype Specific Regulation of Human Vascular α1-Adrenergic Receptors by Vessel Bed and Age. Circulation 1999, 100, 2336–2343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Jensen, B.C.; Swigart, P.M.; Laden, M.E.; DeMarco, T.; Hoopes, C.; Simpson, P.C. The alpha-1D is the predominant alpha-1-adrenergic receptor subtype in human epicardial coronary arteries. J. Am. Coll. Cardiol. 2009, 54, 1137–1145. [Google Scholar] [CrossRef] [Green Version]
  99. Chalothorn, D.; McCune, D.F.; Edelmann, S.E.; Tobita, K.; Keller, B.B.; Lasley, R.D.; Perez, D.M.; Tanoue, A.; Tsujimoto, G.; Post, G.R.; et al. Differential Cardiovascular Regulatory Activities of the α1B- and α1D-Adrenoceptor Subtypes. J. Pharmacol. Exp. Ther. 2003, 305, 1045. [Google Scholar] [CrossRef] [PubMed]
  100. Turnbull, L.; McCloskey, D.T.; O’Connell, T.D.; Simpson, P.C.; Baker, A.J. α1-Adrenergic receptor responses in α1AB-AR knockout mouse hearts suggest the presence of α1D-AR. Am. J. Physiol. Heart Circ. Physiol. 2003, 284, H1104–H1109. [Google Scholar] [CrossRef]
  101. Stassen, F.R.; Willemsen, M.J.; Janssen, G.M.; DeMey, J.G. α1-adrenoceptor subtypes in rat aorta and mesenteric small arteries are preserved during left ventricular dysfunction post-myocardial infarction. Cardiovasc. Res. 1997, 33, 706–713. [Google Scholar] [CrossRef]
  102. Stassen, F.R.; Willemsen, M.J.; Janssen, G.M.; Fazzi, G.E.; Schiffers, P.M.; Smits, J.F.; de Mey, J.G. Reduced responsiveness of rat mesenteric resistance artery smooth muscle to phenylephrine and calcium following myocardial infarction. Br. J. Pharmacol. 1997, 120, 1505–1512. [Google Scholar] [CrossRef]
  103. Piascik, M.T.; Guarino, R.D.; Smith, M.S.; Soltis, E.E.; Saussy, D.L.; Perez, D.M. The specific contribution of the novel alpha-1D adrenoceptor to the contraction of vascular smooth muscle. J. Pharmacol. Exp. Ther. 1995, 275, 1583. [Google Scholar] [PubMed]
  104. Piascik, M.T.; Hrometz, S.L.; Edelmann, S.E.; Guarino, R.D.; Hadley, R.W.; Brown, R.D. Immunocytochemical Localization of the Alpha-1B Adrenergic Receptor and the Contribution of This and the Other Subtypes to Vascular Smooth Muscle Contraction: Analysis with Selective Ligands and Antisense Oligonucleotides. J. Pharmacol. Exp. Ther. 1997, 283, 854. [Google Scholar]
  105. Martinez, L.; Carmona, L.; Villalobos-Molina, R. Vascular alpha 1D-adrenoceptor function is maintained during congestive heart failure after myocardial infarction in the rat. Arch. Med. Res. 1999, 30, 290–297. [Google Scholar] [CrossRef]
  106. Cavalli, A.; Lattion, A.L.; Hummler, E.; Nenniger, M.; Pedrazzini, T.; Aubert, J.F.; Michel, M.C.; Yang, M.; Lembo, G.; Vecchione, C.; et al. Decreased blood pressure response in mice deficient of the alpha1b-adrenergic receptor. Proc. Natl. Acad. Sci. USA 1997, 94, 11589–11594. [Google Scholar] [CrossRef] [Green Version]
  107. Muramatsu, I.; Kigoshi, S.; Ohmura, T. Subtypes of α1-Adrenoceptors Involved in Noradrenaline-Induced Contractions of Rat Thoracic Aorta and Dog Carotid Artery. Jpn. J. Pharmacol. 1991, 57, 535–544. [Google Scholar] [CrossRef]
  108. Suzuki, E.; Tsujimoto, G.; Tamura, K.; Hashimoto, K. Two pharmacologically distinct alpha 1-adrenoceptor subtypes in the contraction of rabbit aorta: Each subtype couples with a different Ca2+ signalling mechanism and plays a different physiological role. Mol. Pharmacol. 1990, 38, 725. [Google Scholar]
  109. Shibata, K.; Hirasawa, A.; Foglar, R.; Ogawa, S.; Tsujimoto, G. Effects of Quinidine and Verapamil on Human Cardiovascular α1-Adrenoceptors. Circulation 1998, 97, 1227–1230. [Google Scholar] [CrossRef] [Green Version]
  110. Fetscher, C.; Schäfers, R.F.; Philipp, T.; Michel, M.C.; Chen, H.; Wambach, G. Effects of noradrenaline and neuropeptide Y on rat mesenteric microvessel contraction. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1996, 353, 314–323. [Google Scholar] [CrossRef]
  111. Chen, H.; Bischoff, A.; Schäfers, R.F.; Wambach, G.; Philipp, T.; Michel, M.C. Vasoconstriction of rat renal interlobar arteries by noradrenaline and neuropeptide Y. J. Auton. Pharmacol. 1997, 17, 137–146. [Google Scholar] [CrossRef] [PubMed]
  112. Rokosh, D.G.; Simpson, P.C. Knockout of the α1A/C-adrenergic receptor subtype: The α1A/C is expressed in resistance arteries and is required to maintain arterial blood pressure. Proc. Natl. Acad. Sci. USA 2002, 99, 9474–9479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tanoue, A.; Nasa, Y.; Koshimizu, T.; Shinoura, H.; Oshikawa, S.; Kawai, T.; Sunada, S.; Takeo, S.; Tsujimoto, G. The α1D-adrenergic receptor directly regulates arterial blood pressure via vasoconstriction. J. Clin. Investig. 2002, 109, 765–775. [Google Scholar] [CrossRef]
  114. Forster, C.; Carter, S.; Armstrong, P. Vascular smooth muscle responsiveness to noradrenaline and phenylephrine following experimental heart failure in dogs. Cardiovasc. Res. 1989, 23, 489–497. [Google Scholar] [CrossRef]
  115. Forster, C.; Armstrong, P.W. Pacing-induced heart failure in the dog: Evaluation of peripheral vascular alpha-adrenoceptor subtypes. J. Cardiovasc. Pharmacol. 1990, 16, 708–718. [Google Scholar] [CrossRef]
  116. Forster, C. Interaction of 5-methyl-urapidil with α1-adrenoceptors in canine blood vessels: Impact of pacing-induced heart failure. Eur. J. Pharmacol. 1996, 318, 55–63. [Google Scholar] [CrossRef]
  117. Le Tran, Y.; Forster, C. Chloroethylclonidine and alpha-adrenoceptor agonist interaction in blood vessels following heart failure. Eur. J. Pharmacol. 1997, 336, 177–185. [Google Scholar] [CrossRef]
  118. Lurie, K.G.; Tsujimoto, G.; Hoffman, B.B. Desensitization of alpha-1 adrenergic receptor-mediated vascular smooth muscle contraction. J. Pharmacol. Exp. Ther. 1985, 234, 147–152. [Google Scholar]
  119. Forster, C.; Campbell, P.M. Nifedipine inhibits responses to alpha-adrenoceptor stimulation in canine blood vessels: Impact of heart failure. Eur. J. Pharmacol. 1993, 242, 119–128. [Google Scholar] [CrossRef]
  120. Forster, C. Inhibition of vascular contractions to alpha-adrenoceptor agonists by polymyxin B: Impact of heart failure state. Eur. J. Pharmacol. 1995, 283, 241–250. [Google Scholar] [CrossRef]
  121. Bergdahl, A.; Valdemarsson, S.; Sun, X.Y.; Hedner, T.; Edvinsson, L. Enhanced Ca2+-induced contractions and attenuated alpha-adrenoceptor responses in resistance arteries from rats with congestive heart failure. Eur. J. Heart Fail. 2001, 3, 7–13. [Google Scholar] [CrossRef] [Green Version]
  122. Koida, S.; Ohyanagi, M.; Ueda, A.; Mori, Y.; Iwasaka, T. Mechanism of increased alpha-adrenoceptor-mediated contraction in small resistance arteries of rats with heart failure. Clin. Exp. Pharmacol. Physiol. 2006, 33, 47–52. [Google Scholar] [CrossRef]
  123. Trautner, S.; Amtorp, O.; Boesgaard, S.; Andersen, C.B.; Galbo, H.; Haunsoe, S.; Sheykhzade, M. Noradrenaline-induced increases in calcium and tension in skeletal muscle conductance and resistance arteries from rats with post-infarction heart failure. Eur. J. Pharmacol. 2006, 537, 143–154. [Google Scholar] [CrossRef]
  124. Schrier, R.W.; Abraham, W.T. Hormones and Hemodynamics in Heart Failure. N. Engl. J. Med. 1999, 341, 577–585. [Google Scholar] [CrossRef]
  125. Maze, M.; Spiss, C.K.; Tsujimoto, G.; Hoffman, B.B. Epinephrine infusion induces hyporesponsiveness of vascular smooth muscle. Life Sci. 1985, 37, 1571–1578. [Google Scholar] [CrossRef]
  126. Snavely, M.D.; Ziegler, M.G.; Insel, P.A. Subtype-Selective Down-Regulation of Rat Renal Cortical α-and β-Adrenergic Receptors by Catecholamines. Endocrinology 1985, 117, 2182–2189. [Google Scholar] [CrossRef]
  127. Wikberg, J.E.S.; Akers, M.; Caron, M.G.; Hagen, P.O. Norepinephrine-induced down regulation of alpha1 adrenergic receptors in cultured rabbit aorta smooth muscle cells. Life Sci. 1983, 33, 1409–1417. [Google Scholar] [CrossRef]
  128. Leeb-Lundberg, L.M.; Cotecchia, S.; DeBlasi, A.; Caron, M.G.; Lefkowitz, R.J. Regulation of adrenergic receptor function by phosphorylation. I. Agonist-promoted desensitization and phosphorylation of alpha 1-adrenergic receptors coupled to inositol phospholipid metabolism in DDT1 MF-2 smooth muscle cells. J. Biol. Chem. 1987, 262, 3098–3105. [Google Scholar]
  129. Townsley, M.I.; Pitts, V.H.; Ardell, J.L.; Zhao, Z.; Johnson, W.H., Jr. Altered pulmonary microvascular reactivity to norepinephrine in canine pacing-induced heart failure. Circ. Res. 1994, 75, 347–356. [Google Scholar] [CrossRef] [Green Version]
  130. Bergdahl, A.; Valdemarsson, S.; Pantev, E.; Ottosson, A.; Feng, Q.P.; Sun, X.Y.; Hedner, T.; Edvinsson, L. Modulation of vascular contractile responses to α1-and α2-adrenergic and neuropeptide Y receptor stimulation in rats with ischaemic heart failure. Acta Physiol. Scand. 1995, 154, 429–437. [Google Scholar] [CrossRef]
  131. Tamagawa, K.; Saito, T.; Oikawa, Y.; Maehara, K.; Yaoita, H.; Maruyama, Y. Alterations of alpha-adrenergic modulations of coronary microvascular tone in dogs with heart failure. J. Card. Fail. 2005, 11, 388–395. [Google Scholar] [CrossRef]
  132. Main, J.S.; Forster, C.; Armstrong, P.W. Inhibitory role of the coronary arterial endothelium to alpha-adrenergic stimulation in experimental heart failure. Circ. Res. 1991, 68, 940–946. [Google Scholar] [CrossRef] [Green Version]
  133. Stassen, F.R.M.; Fazzi, G.E.; Leenders, P.J.A.; Smits, J.F.M.; de Mey, J.G.R. Coronary Arterial Hyperreactivity and Mesenteric Arterial Hyporeactivity After Myocardial Infarction in the Rat. J. Cardiovasc. Pharmacol. 1997, 29, 780–788. [Google Scholar] [CrossRef]
  134. Feng, Q.; Sun, X.; Lu, X.; Edvinsson, L.; Hedner, T. Decreased responsiveness of vascular postjunctional alpha1-, alpha2-adrenoceptors and neuropeptide Y1 receptors in rats with heart failure. Acta Physiol. Scand. 1999, 166, 285–291. [Google Scholar] [CrossRef]
  135. Ahmadiasl, N.; Rostami, A.; Mohammadi, N.M.; Rajabi, F. Effects of noradrenaline and KCl on peripheral vessels in doxorubicin induced model of heart failure. Pathophysiology 2002, 8, 259–262. [Google Scholar] [CrossRef]
  136. Ramchandra, R.; Xing, D.T.; Matear, M.; Lambert, G.; Allen, A.M.; May, C.N. Neurohumoral interactions contributing to renal vasoconstriction and decreased renal blood flow in heart failure. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2019, 317, R386–R396. [Google Scholar] [CrossRef]
  137. Goldsmith, S.R.; Francis, G.S.; Cohn, J.N. Norepinephrine infusions in congestive heart failure. Am. J. Cardiol. 1985, 56, 802–804. [Google Scholar] [CrossRef]
  138. Borow, K.M.; Neumann, A.; Arensman, F.W.; Yacoub, M.H. Cardiac and peripheral vascular responses to adrenoceptor stimulation and blockade after cardiac transplantation. J. Am. Coll. Cardiol. 1989, 14, 1229–1238. [Google Scholar] [CrossRef] [Green Version]
  139. Angus, J.A.; Ferrier, C.P.; Sudhir, K.; Kaye, D.M.; Jennings, G.L. Impaired contraction and relaxation in skin resistance arteries from patients with congestive heart failure. Cardiovasc. Res. 1993, 27, 204–210. [Google Scholar] [CrossRef]
  140. Barrett-O’Keefe, Z.; Lee, J.F.; Ives, S.J.; Trinity, J.D.; Witman, M.A.H.; Rossman, M.J.; Groot, H.J.; Sorensen, J.R.; Morgan, D.E.; Nelson, A.D.; et al. α-Adrenergic receptor regulation of skeletal muscle blood flow during exercise in heart failure patients with reduced ejection fraction. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2019, 316, R512–R524. [Google Scholar] [CrossRef]
  141. Wilson, J.R.; Matthai, W.; Lanoce, V.; Frey, M.; Ferraro, N. Effect of experimental heart failure on peripheral sympathetic vasoconstriction. Am. J. Physiol. 1988, 254, H727–H733. [Google Scholar] [CrossRef]
  142. Mulder, P.; Compagnon, P.; Devaux, B.; Richard, V.; Henry, J.P.; Elfertak, L.; Wimart, M.C.; Thibout, E.; Comoy, E.; Mace, B.; et al. Response of large and small vessels to alpha and beta adrenoceptor stimulation in heart failure: Effect of angiotensin converting enzyme inhibition. Fundam. Clin. Pharmacol. 1997, 11, 221–230. [Google Scholar] [CrossRef]
  143. McMillon, R.K.; Townsley, M.I. Alpha-adrenergic vasoreactivity of canine intrapulmonary bronchial arteries in pacing-induced heart failure. Am. J. Physiol. 1999, 277, H1392–H1402. [Google Scholar]
  144. Indolfi, C.; Maione, A.; Volpe, M.; Rapacciuolo, A.; Esposito, G.; Ceravolo, R.; Rendina, V.; Condorelli, M.; Chiariello, M. Forearm vascular responsiveness to alpha 1- and alpha 2-adrenoceptor stimulation in patients with congestive heart failure. Circulation 1994, 90, 17–22. [Google Scholar] [CrossRef] [Green Version]
  145. Schwinn, D.A.; McIntyre, R.W.; Hawkins, E.D.; Kates, R.A.; Reves, J.G. alpha 1-Adrenergic responsiveness during coronary artery bypass surgery: Effect of preoperative ejection fraction. Anesthesiology 1988, 69, 206–217. [Google Scholar] [CrossRef] [PubMed]
  146. Teerlink, J.R.; Gray, G.A.; Clozel, M.; Clozel, J.P. Increased vascular responsiveness to norepinephrine in rats with heart failure is endothelium dependent. Dissociation of basal and stimulated nitric oxide release. Circulation 1994, 89, 393–401. [Google Scholar] [CrossRef] [Green Version]
  147. Van Tassell, B.W.; Rondina, M.T.; Huggins, F.; Gilbert, E.M.; Munger, M.A. 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. 2008, 156, 315–321. [Google Scholar] [CrossRef] [Green Version]
  148. Mey, J.D.; Vanhoutte, P.M. Uneven distribution of postjunctional alpha 1-and alpha 2-like adrenoceptors in canine arterial and venous smooth muscle. Circ. Res. 1981, 48, 875–884. [Google Scholar] [CrossRef] [Green Version]
  149. Van Vessem, M.E.; Petrus, A.H.J.; Palmen, M.; Braun, J.; Schalij, M.J.; Klautz, R.J.M.; Beeres, S.L.M.A. Vasoplegia after Restrictive Mitral Annuloplasty for Functional Mitral Regurgitation in Patients with Heart Failure. J. Cardiothorac. Vasc. Anesth. 2019, 33, 3273–3280. [Google Scholar] [CrossRef] [PubMed]
  150. Argenziano, M.; Chen, J.M.; Choudhri, A.F.; Cullinane, S.; Garfein, E.; Weinberg, A.D.; Smith, C.R., Jr.; Rose, E.A.; Landry, D.W.; Oz, M.C. Management of vasodilatory shock after cardiac surgery: Identification of predisposing factors and use of a novel pressor agent. J. Thorac. Cardiovasc. Surg. 1998, 116, 973–980. [Google Scholar] [CrossRef] [Green Version]
  151. Orozco Vinasco, D.M.; Triana Schoonewolff, C.A.; Orozco Vinasco, A.C. Vasoplegic syndrome in cardiac surgery: Definitions, pathophysiology, diagnostic approach and management. Rev. Esp. Anestesiol. Reanim. 2019, 66, 277–287. [Google Scholar] [CrossRef]
  152. Van Vessem, M.E.; Palmen, M.; Couperus, L.E.; Stijnen, T.; Berendsen, R.R.; Aarts, L.; de Jonge, E.; Klautz, R.J.M.; Schalij, M.J.; Beeres, S. Vasoplegia after Surgical Left Ventricular Restoration: 2-Year Follow-Up. Ann. Thorac. Surg. 2018, 106, 1371–1378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Tecson, K.M.; Lima, B.; Lee, A.Y.; Raza, F.S.; Ching, G.; Lee, C.H.; Felius, J.; Baxter, R.D.; Still, S.; Collier, J.D.G.; et al. Determinants and Outcomes of Vasoplegia Following Left Ventricular Assist Device Implantation. J. Am. Heart Assoc. 2018, 7, e008377. [Google Scholar] [CrossRef] [Green Version]
  154. Stassen, F.R.; Maas, R.G.; Schiffers, P.M.; Janssen, G.M.; de Mey, J.G. A positive and reversible relationship between adrenergic nerves and alpha-1A adrenoceptors in rat arteries. J. Pharmacol. Exp. Ther. 1998, 284, 399–405. [Google Scholar]
  155. Bristow, M.R.; Feldman, A.M.; Adams, K.F., Jr.; Goldstein, S. Selective versus nonselective beta-blockade for heart failure therapy: Are there lessons to be learned from the COMET trial? J. Card. Fail. 2003, 9, 444–453. [Google Scholar] [CrossRef]
  156. Furberg, C.D.; Wright, J.T.; Davis, B.R.; Cutler, J.A.; Alderman, M.; Black, H.; Cushman, W.; Grimm, R.; Haywood, J.; Leenen, F.; et al. Major Cardiovascular Events in Hypertensive Patients Randomized to Doxazosin vs Chlorthalidone; The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). JAMA 2000, 283, 1967–1975. [Google Scholar]
  157. O’Connell, T.D.; Swigart, P.M.; Rodrigo, M.C.; Ishizaka, S.; Joho, S.; Turnbull, L.; Tecott, L.H.; Baker, A.J.; Foster, E.; Grossman, W.; et al. Alpha1-adrenergic receptors prevent a maladaptive cardiac response to pressure overload. J. Clin. Investig. 2006, 116, 1005–1015. [Google Scholar] [CrossRef]
  158. Cohn, J.N. The Vasodilator-Heart Failure Trials (V-HeFT). Mechanistic data from the VA Cooperative Studies. Introduction. Circulation 1993, 87, VI1–VI4. [Google Scholar]
  159. O’Connell, T.D.; Ishizaka, S.; Nakamura, A.; Swigart, P.M.; Rodrigo, M.C.; Simpson, G.L.; Cotecchia, S.; Rokosh, D.G.; Grossman, W.; Foster, E.; et al. The α1A/C- and α1B-adrenergic receptors are required for physiological cardiac hypertrophy in the double-knockout mouse. J. Clin. Investig. 2003, 111, 1783–1791. [Google Scholar] [CrossRef]
  160. Kukin, M.L.; Kalman, J.; Mannino, M.; Freudenberger, R.; Buchholz, C.; Ocampo, O. Combined alpha-beta blockade (doxazosin plus metoprolol) compared with beta blockade alone in chronic congestive heart failure. Am. J. Cardiol. 1996, 77, 486–491. [Google Scholar] [CrossRef]
  161. Tham, T.C.; Guy, S.; McDermott, B.J.; Shanks, R.G.; Riddell, J.G. The dose dependency of the alpha- and beta-adrenoceptor antagonist activity of carvedilol in man. Br. J. Clin. Pharmacol. 1995, 40, 19–23. [Google Scholar] [CrossRef] [Green Version]
  162. Kubo, T.; Azevedo, E.R.; Newton, G.E.; Parker, J.D.; Floras, J.S. Lack of evidence for peripheral alpha1-adrenoceptor blockade during long-term treatment of heart failure with carvedilol. J. Am. Coll. Cardiol. 2001, 38, 1463. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) study flow chart.
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) study flow chart.
Cells 09 02412 g001
Figure 2. The prototypical signaling pathway of α1-ARs. CA: catecholamines, DAG: diacylglycerol, IP3: inositol triphosphate, PKC: protein kinase C, PLC: phospholipase C.
Figure 2. The prototypical signaling pathway of α1-ARs. CA: catecholamines, DAG: diacylglycerol, IP3: inositol triphosphate, PKC: protein kinase C, PLC: phospholipase C.
Cells 09 02412 g002
Figure 3. Differential expressions of α1-ARs in the cardiovascular system. NA: noradrenaline.
Figure 3. Differential expressions of α1-ARs in the cardiovascular system. NA: noradrenaline.
Cells 09 02412 g003
Table 1. Summary of the changes in cardiac mRNA studies for total α1-adrenoceptors (AR) and their subtypes in chronic heart failure.
Table 1. Summary of the changes in cardiac mRNA studies for total α1-adrenoceptors (AR) and their subtypes in chronic heart failure.
mRNASpeciesTissueα1A-AR α1B-ARα1D-ARTotal α1-AR
Fischer et al. [43]humanLV, RV, LA, RA-n/a
Jensen et al. [41] humanLV and RV XXX
Monto et al. [17]humanLV and RVX XXn/a
Brattelid et al. [2]ratLVX Xn/a
↑: increased; ↓: decreased; X: unchanged; n/a: no data available; LV, left ventricle; RV, right ventricle; LA, left atrium; RA, right atrium.
Table 2. Summary of cardiac α1-adrenoceptor (AR) protein expression in chronic heart failure.
Table 2. Summary of cardiac α1-adrenoceptor (AR) protein expression in chronic heart failure.
BindingSpeciesTissueα1A-ARα1B-ARα1D-ARTotal α1-AR
Böhm et al. [49]humanpapillaryn/an/an/aX
Bristow et al. [50]humanLV and RVn/an/an/aX
Corr et al. [36]catventriclen/an/an/a
Dixon et al. [54]ratLVn/an/an/a
Erdmann and Böhm [51]humanpapillaryn/an/an/aX
Fischer et al. [43]humanLV, RV, LA, RAn/a
Gopalakrishnan et al. [32]ratLV and RVn/an/an/aX
Hwang et al. [45]humanexplanted heartsn/an/an/a↑ (in ICM)
X (in DCM)
Jensen et al. [41]humanLV and RVX (↑ in RV)XundetectedX
Karliner et al. [33]GPwhole heartn/an/an/a
Limas et al. [52]humanLV and RVn/an/an/a↓ (in severe HF)
X (in mild)
Litwin et al. [25]ratLVn/an/an/aX
Maisel et al. [34]GPLVn/an/an/a
Meggs et al. [26]ratLV and RVn/an/an/aX
Re et al. [37]dogLV, RV, LA, RAn/an/an/a
Shi et al. [53]humanLVXn/a
Sjaastad et al. [55]ratLV and RVn/an/an/aX
Steinfath et al. [46]humanLVn/an/an/a
Watanebe et al. [35]hamsterwhole heartn/an/an/aSlight↓
Zhao et al. [30]ratRA and LVn/an/an/a
Rowley et al. [56]ratatrian/an/an/aX
Vago et al. [47]humanLVn/an/an/a
GP: guinea pig, ↑: increased; ↓: decreased; X: unchanged; n/a: no data available; LV, left ventricle; RV, right ventricle; LA, left atrium; RA, right atrium.
Table 3. Summary of changes in cardiac α1-adrenoceptor (AR)-mediated responses in chronic heart failure.
Table 3. Summary of changes in cardiac α1-adrenoceptor (AR)-mediated responses in chronic heart failure.
SpeciesPreparationα1-AR-mediated Responses
Böhm et al. [49]humanpapillaryX
Erdmann and Böhm [51]humanpapillaryX
Litwin et al. [25]ratpapillaryX
Sjaastad et al. [55]ratpapillaryX
Cowley et al. [29]mousetrabeculaα1A–med↑
α1B–med X
Janssen et al. [60] humantrabeculaslight ↓
Steinfath et al. [46]humantrabecula
Wang et al. [27]mousetrabeculain RV switch from NIE to PIE
in LV X
Skomedal et al. [81] humantrabeculaPIE
Rowley et al. [56]ratatria
↑: increased; ↓: decreased; X: unchanged; RV: rigt ventricle; LV: left ventricle; NIE: negative inotropic effect; PIE: positive inotropic effect.
Table 4. Summary of changes in vascular α1-adrenoceptor (AR)-mediated responses in chronic heart failure.
Table 4. Summary of changes in vascular α1-adrenoceptor (AR)-mediated responses in chronic heart failure.
SpeciesPreparation/Parameter Studiedα1-AR-Mediated Responses
Goldsmith et al. [137] humanIn vivo hemodynamic responses
Wilson et al. [141] dogIn vivo hindlimb vascular responsesX
Schwinn et al. [145]humanIn vivo BP responseX (awake)/
↓ (on fentanyl anesthesia)
Borow et al. [138] humanIn vivo BP response
Forster et al. [114] dogDPA and SV
Forster et al. [115] dogDPA and SV
Main et al. [132] dogLAD and LCX
Forster et al. [119] dogDPA and SV
Angus et al. [139] humanSmall arteries from gluteal skin biopsy
Townsley et al. [129] dogPulmonary arterial and venous pressure response
Indolfi et al. [144] humanIn-vivo forearm blood flow responseX
Teerlink et al. [146] ratThoracic aorta↑ intact vessels/
↓ denuded vessels
Forster et al. [120] dogDPA and SV↑ significant only in SV
Bergdahl et al. [130] ratBasilar, femoral, and renal artery and iliac vein↑ arteries (NS)
↓ vein
Forster et al. [116] dogDPA and SV
Stassen et al. [102] ratMSA
Stassen et al. [133] ratThoracic aorta, coronary arteries, MSAX thoracic aorta/
↑ coronary arteries (NS)/↓ MSA
Mulder et al. [142] ratAbdominal aorta, Femoral artery, MSA X
Le Tran et al. [117] dogDPA and SV↑ (significant only for NA in DPA)
Martinez et al. [105] ratAorta, Carotid arteryX
McMillon et al. [143] dogIntrapulmonary bronchial vesselsX
Feng et al. [134] ratIn vivo BP response
Bergdahl et al. [121] ratMSA
Ahmadiasl et al. [135] rabbitThoracic aorta, left renal artery and vein, lateral saphenous artery and vein, vena cava↓ significant only in vena cava
Tamagawa et al. [131] dogLAD/ In vivo coronary pressure-flow relationship response
Trautner et al. [123] ratFemoral artery (2nd order side branches) and muscle resistance arteriesX
Koida et al. [122] ratFemoral resistance artery (small branch)
Van Tassel et al. [147]humanIn vivo BP response↑ with up-titration of carvedilol
Ramchandra et al. [136] sheepRenal vessels/In vivo renal vasoconstrictor response
Barrett-O’Keefe et al. [140] humanCFA and CFV/In vivo leg blood flow response ↓at rest
BP: blood pressure, CFA: common femoral artery, CFV: common femoral vein, DPA: dorsal pedal artery, LAD: left anterior descending coronary artery, LCX: left circumflex coronary artery, MSA: mesenteric small artery, NS: not significant, SV: saphenous vein, ↑: increase, ↓: decrease, X: similar.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kaykı-Mutlu, G.; Papazisi, O.; Palmen, M.; Danser, A.H.J.; Michel, M.C.; Arioglu-Inan, E. Cardiac and Vascular α1-Adrenoceptors in Congestive Heart Failure: A Systematic Review. Cells 2020, 9, 2412. https://doi.org/10.3390/cells9112412

AMA Style

Kaykı-Mutlu G, Papazisi O, Palmen M, Danser AHJ, Michel MC, Arioglu-Inan E. Cardiac and Vascular α1-Adrenoceptors in Congestive Heart Failure: A Systematic Review. Cells. 2020; 9(11):2412. https://doi.org/10.3390/cells9112412

Chicago/Turabian Style

Kaykı-Mutlu, Gizem, Olga Papazisi, Meindert Palmen, A. H. Jan Danser, Martin C. Michel, and Ebru Arioglu-Inan. 2020. "Cardiac and Vascular α1-Adrenoceptors in Congestive Heart Failure: A Systematic Review" Cells 9, no. 11: 2412. https://doi.org/10.3390/cells9112412

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop