Adrenoceptors: Introduction

General

Adrenoceptors (AR) are a group of nine 7-transmembrane receptors comprising 3 main types, α1, α2 and β, each with 3 subtypes that mediate the central and peripheral actions of the catecholamine hormones and neurotransmitters adrenaline (epinephrine) and noradrenaline (norepinephrine). Adrenoceptors are found in nearly all peripheral tissues and on many neuronal populations within the central nervous system. Both noradrenaline and adrenaline play important roles in the control of blood pressure, myocardial contractile rate and force, airway reactivity, and a variety of metabolic and central nervous system functions. Both catecholamines activate all adrenoceptor subtypes although noradrenaline displays some selectivity for β1-AR and adrenaline for β2-AR due to slightly higher affinity at their respective subtypes. Adrenaline and noradrenaline are important in the "fight or flight response" where blood is diverted from non-essential organs such as skin and gut to skeletal muscle, whilst maximising cardiac output, oxygenation and serum glucose and uptake into muscle. Thus, endogenous agonist actions at α1-AR increase vascular smooth muscle contraction in those blood vessels where these predominate and relaxation in blood vessels where β-ARs predominate. Stimulation of β-ARs in the heart increases heart rate and contractility and actions on β1-ARs on the juxta-glomerular apparatus in the kidney increases renin release. Stimulation of β2-AR in airways causes smooth muscle relaxation and bronchodilation. Activation of β2-AR also has other effects - tremor in skeletal smooth muscle and metabolic actions including a decrease is serum potassium, calcium, magnesium, phosphate, growth hormone, an increase in lactate, renin and lipolysis, and increased serum glucose due to breakdown of glycogen in the liver to release glucose as well as increasing glucose uptake into skeletal muscle. The only known relevant effect of β3-AR stimulation in adult humans is relaxation of smooth muscle in the urinary bladder.
The physiological agonists noradrenaline and adrenaline have some clinical uses: both are used to raise blood pressure in shock (septic or cardiogenic) and adrenaline is used to treat anaphylaxis and cardiac arrest. More selective agonists and antagonists interacting with adrenoceptors have proved useful in the treatment of a variety of diseases, including arrhythmias, ischaemic heart disease (post-myocardial infarction and angina pectoris), hypertension, portal hypertension, congestive heart failure, asthma, depression, benign prostatic hyperplasia, glaucoma, sedation, thyrotoxicosis, migraine and anxiety. Drugs acting at these receptors are also useful in several other therapeutic situations including premature labour (no-longer used) and opioid withdrawal, and as adjuncts to general anaesthetics.

Discovery of adrenaline and adrenoceptors
Oliver and Schäfer discovered in 1894 that ingestion of sheep adrenal gland caused adrenaline-mediated vasoconstriction of the radial artery, rapidly followed by more widespread vasoconstriction, increased blood pressure and ventricular contraction [114-115]. Subsequently, adrenaline was isolated as a pure crystalline substance [162], its formula determined in 1901 and it was named to distinguish it from other less active isolates from adrenal medulla. Adrenaline was first used clinically in 1903 in patients with asthma [25-26]. Sir Henry Dale pioneered the development of drugs acting selectively at adrenoceptors (ARs) and recognised that the actions of adrenaline were altered by preexposure to ergotoxine from a vasoconstrictor to a vasodilator response that indicated a mixed response under normal conditions. He suggested that ergotoxine caused a selective paralysis of myoneural junctions responsible for the vasoconstrictor response [40].
The history of the recognition of adrenoceptor subtypes began in 1948, with the division into two major types, α and β, based on their pharmacological characteristics (i.e., rank order of potency of agonists) [3]. Subsequently, both the α and β types were subdivided into α1, α2, β1 and β2 subtypes (for a more complete historical perspective, see [29]. Based on more recent pharmacological and molecular evidence, it is now clear that classification is based on three major types - α1, α2 and β - each of which is further divided into three subtypes [27].

α1-Adrenoceptors

α1-adrenoceptor subtypes
Building on an initial report that both phentolamine and WB 4101 inhibited [3H]prazosin binding in rat frontal cortex in a manner consistent with more than one type of receptor [16], Morrow and Creese [105] formally proposed a definition of α1A and α1B ARs based on the differential affinities of WB 4101 and phentolamine. At about the same time Johnson and Minneman [75] observed that the alkylating agent chloroethylclonidine selectively inactivated only half of the HEAT (BE2254) binding to α1-ARs in rat cerebral cortex. The inactivated sites corresponded to the α1B-AR subtype, based on low affinity for WB 4101 [68,104].
Three α1-AR subtypes have been identified by cloning and are now known as α1A, α1B and α1D. The α1B-AR from the DDT cell line (hamster smooth muscle) was cloned first [37], followed by what was thought to be a novel α1-AR from a bovine brain cDNA library identified as the α1C subtype [142]. Subsequently, it was shown that this clone is a species orthologue corresponding to the pharmacologically defined α1A subtype [57,70]. A third α1-AR was cloned from rat cortex and initially designated as an α1A-AR [94]. However, an identical recombinant rat α1-AR subtype was independently identified by Perez et al. [120] and denoted as the α1D-AR. This α1D-subtype was subsequently characterized functionally in tissues [81,122]. It appears that the α1D-AR signals less effectively upon agonist stimulation as compared to the other subtypes, perhaps because it exhibits spontaneous internalization [101].
Four isoforms of the α1A-AR have been reported. In addition to the originally reported sequence (α1A-1), two C-terminal variants (α1A-2 and α1A-3) produced by alterative splicing were reported in 1995 [71]. An additional splice variant (α1A-4) was subsequently reported [32]. No functional differences among these splice variants have been reported to date.
A fourth α1-AR subtype was postulated and designated as α1L based on low affinity for prazosin [116] and RS-17053. The evidence for the existence of the α1L-AR was supported by pharmacological data in several tissues, including human prostate, bladder neck and periurethral longitudinal muscle [55,106]. There is now strong evidence that the α1L subtype represents a particular conformational state of the α1A-AR [56] or due to differences in characteristics dependent on the tissue or assay environment [37].The α1L-AR mediated responses in the mouse prostate are essentially abolished in α1A-AR knockout mice [66].

α1-adrenoceptor expression
α1-AR are expressed in blood vessels, heart, urinary tract, brain, kidney and liver [29,56,64,103,111,123,128]. The individual subtypes have clearly defined roles in the brain, cardiovascular system and genitourinary system. In the brain, α1A-AR stimulation increases neurogenesis, learning and memory and improves mood whereas α1B-AR activation improves memory consolidation, exploratory activity and causes behavioural activation [4]. In the cardiovascular system α1A-AR have a dominant role in small densely innervated arteries in contrast to α1D-AR that are involved in contraction of large poorly innervated arteries and cause vessel hypertrophy [4]. In the heart, α1A-AR are protective and adaptive whereas α1B-AR activation causes maladaptive hypertrophy [4,111]. The dominant α-AR subtype in the genitourinary system is the α1A-AR that mediates contraction in the ureter, bladder, urethra and prostate but is also involved in fertility in the male [158].

α1-adrenoceptor signalling
The α1-ARs couple primarily to Gαq-proteins to promote calcium release and inositol phosphate production and also activate protein kinase C and mitogen activated protein kinases. Of the subtypes, α1A-ARs couple most efficiently to these pathways. α1A- and α1B-ARs can also couple to adenylyl cyclase to increase cAMP but with lower coupling efficacy [39,128].
The crystal structure of the α1B-AR has recently been determined [42].

Selectivity and clinical uses of drugs acting at α1-adrenoceptors
Agonists
The general α1-AR agonist phenylephrine is a common constituent of orally and nasally administered medicines for symptomatic relief of colds, coughs, influenza, allergies, sinusitis and bronchitis but can produce many side effects including increased blood pressure, dizziness, headache and tachycardia [4]. Oxymetazoline and xylometazoline (that display some α1A-AR selectivity but are complicated by significant actions at α2-AR and 5HT1B receptors [39], are also used as over-the-counter remedies for nasal congestion. The agonist A61603 is highly selective for the α1A-AR [39,52,128] although not used clinically. There are no selective agonists for α1B- or α1D-AR.

Antagonists
Non-selective α1-AR antagonists (e.g. doxazosin) are used to treat arterial hypertension. They are also useful for improving symptoms in benign prostatic hyperplasia (e.g. tamsulosin, terazosin). Prazosin, and increasingly doxazosin, are used to help symptoms in PTSD. Phentolamine and phenoxybenzamine (which has an ability to covalently bind to α1-AR making it "irreversible" [58]) are used to treat phaeochromocytoma. Labetalol is the drug of choice for hypertension in pregnancy. Several antidepressants and anti-psychotics have high α1A-AR affinity that may partly account for their blood pressure lowering side effects.
Several α1A-AR selective antagonists are available (e.g. silodosin, S(+)-niguldipine, SNAP5089, RS-100329 and Ro-70-0004) and are selective for α1A-ARs over the α1B- and α1D-AR subtypes [159]. The insurmountable antagonist ρ-Da1a is also α1A-AR subtype selective. There are no selective antagonists for α1B-AR. BMY-7378 shows selectivity for α1D-AR [129].
Both agonist and antagonist pharmacological characteristics are relatively well preserved across species.

α2-Adrenoceptors

α2-adrenoceptor subtypes
The evidence for α2-AR subtypes originally came from binding and functional studies in various tissues and cell lines, and more recently from cells transfected with the cDNA for the receptors . On the basis of these studies, three genetically distinct α2-AR subtypes have been defined- α2A, α2B and α2C. The α2A-AR, at which prazosin has a relatively low affinity and oxymetazoline a relatively high affinity, is found in for example in human platelets and HT29 cells [27,84]. The second subtype, α2B, was identified in neonatal rat lung and in NG108 cells [30]. This subtype has relatively high affinity for prazosin and a low affinity for oxymetazoline [30,156]. A third subtype, α2C, was originally identified in an opossum kidney (OK) cell line and subsequently cloned from human kidney [107,131]. This subtype also has relatively high affinity for prazosin and a low affinity for oxymetazoline, but is pharmacologically distinct from the α2B subtype [20]. The α2D, that was described in the rat salivary [98] and bovine pineal gland [144] and cloned from the rat [88] are species orthologues of the human α2A subtype. The species orthologue of the human α2A-ARs found in the rat, mouse and cow has significantly different antagonist pharmacology, but the agonist pharmacology appears to be similar.
Additional α2-AR subtypes have been identified in fish, including five receptor genes in the zebrafish and as many as eight in the pufferfish [134]. In the zebrafish, three of the subtypes are similar to those found in mammals (orthologues, the same gene in different species), whereas the other two are not found in mammals, but are paralogues (duplicated genes in the same species). The significance of the numerous receptor subtypes in non-mammalian species is not well understood [28].

α2-adrenoceptor expression
α2-AR are widely distributed in the brain and in the periphery [47,51,90,95,121]. Although originally identified and regarded as prejunctional autoreceptors (i.e. receptors on presynaptic neurones that when stimulated by noradrenaline in the synapse act as a negative feedback mechanism inhibiting further noradrenaline release from the neurone), it rapidly became evident that α2-AR are located both pre and post-junctionally [18]. α2A-AR are located in the locus coeruleus and other noradrenergic cell bodies involved in controlling sympathetic outflow, brainstem, cerebral cortex, hippocampus, septum, hypothalamic and amygdaloid nuclei and spinal cord [95]. α2C-AR are largely located in caudate putamen, olfactory tubercle, hippocampus and cerebral cortex [95]. More recent detailed studies in human prefrontal cortex show that the dominant (87%) receptors are post-synaptic α2A-AR, with the remaining α2C-AR (13%) being located both pre and post-synaptically (60/40) [51]. α2B-AR are weakly expressed solely in the thalamus [51]. All three α2-AR subtypes are widely distributed in the periphery being present in the heart, blood vessels, lung, kidney, pancreas, gastrointestinal tract, adrenal gland, spleen and platelets [47,121,126].

α2-adrenoceptor signalling
α2-AR are primarily Gi/Go coupled receptors, so that activation of α2-AR causes an inhibition of adenylyl cyclase, inhibition of L-type voltage-gated Ca2+ channels (thus raising intracellular calcium), increased Na+/H+ exchange and opening K+ channels (K-ATP), stimulate phospholipase α2 and ERK1/2 phosphorylation [92,95]. In some circumstances αA2- and α2B-AR can also couple to Gαs to increase adenylyl cyclase activity and cAMP accumulation [46,48], although this Gαs-coupling requires high receptor expression, high concentrations of high efficacy agonists and is of lower coupling efficacy. The physiological significance of this mechanism is unknown [127]. α-AR are important for control of blood pressure, analgesia, sedation, platelet aggregation and hypothermia [129].
The structures of the three α2-AR subtypes have recently been determined [33,130,163].

Selectivity and clinical uses of drugs acting at α2-adrenoceptors
Agonists
The α2-AR subtypes are activated by the endogenous ligands, (-)-adrenaline and (-)-noradrenaline but no known clinical actions are associated with these effects. The non-selective partial agonist clonidine was one of the first selective α2-AR agonists to be developed and is a centrally acting anti-hypertensive that alters baroreflex control to cause hypotension and bradycardia [74,119,124]. These properties are shared by other α2-AR partial agonists, e.g. dexmedetomidine and xylazine. These compounds, particularly dexmedetomidine, a very potent α2-AR agonist, are now mainly used for sedation in an intensive care setting including as an adjunct to other agents as it causes less respiratory depression and is useful for reducing delirium, nausea and agitation, and even allows arousal or cooperative sedation allowing neurosurgery in awake patients [60,63,89,154]. It is also useful for minimising withdrawal symptoms from opioids, benzodiazepines, alcohol and nicotine. The activity of dexmedetomidine at α2-ARs is mainly associated with its affinity for all three α2-AR subtypes [127] but it and similar drugs may also be used to control agitation associated with schizophrenia or bipolar disorder. Xylazine has been used in veterinary medicine for >50 years for its analgesic and sedative effects in a variety of species including cats, dogs and horses but has been superseded to some extent by dexmedetomidine which is more potent. An important advantage conferred by these compounds is that the sedation associated with their use is easily reversible [15]. Brimonidine (UK14304) (UK14304) is a non-selective full agonist that has vasoconstrictor and anti-inflammatory properties that make it useful for the treatment of facial erythema in rosacea and glaucoma where it reduces aqueous humour production whist increasing its outflow [2,74,119,124-125]. Guanabenz is a partial agonist that displays some α2A-AR selectivity but also activates α2B-AR and has no agonist actions at α2C-AR although it has affinity for this subtype [74]. It is used mainly as a centrally acting anti-hypertensive perhaps reflecting its actions on the two dominant α2-AR subtypes in the CNS. Guanfacine, another partial agonist at all 3 α2-AR subtypes, is again somewhat α2A-AR selective, and is mainly used in patients with ADHD as it avoids the cardiovascular and hypertensive issues of other ADHD medications [87,110]. Lofexidine is now used predominantly to treat the symptoms of opiate withdrawal [151]. It has an interesting pharmacological profile appearing in GTPγs binding assays to be a selective agonist at α2B-AR yet in binding studies displaying high affinity for all 3 subtypes [44,74]. Among other agonists active at α2-ARs, tizanidine is used to relieve muscle spasticity [63], brimonidine (UK14304) is used for glaucoma where it reduces aqueous humour production whilst increasing its outflow [2], and brimonidine and oxymetazoline are used as topical vasoconstrictors in rosacea [113]. Several anti-Parkinsonian drugs such as lisuride, roxindole and terguride have high potency competing for α2-AR binding [101]. The agonist xylazine has recently emerged in the North American illegal drug markets as a common admixture with synthetic opioids particularly fentanyl and is associated with a marked increase in the number of fatalities associated with drug overdose. While opioid antagonists such as naloxone can rapidly reverse the effects of fentanyl, they do not counteract the sedation, bradycardia and hypotension due to xylazine. There are no highly selective α2A-AR agonists, and no α2B or α2C-AR selective agonists.

Antagonists
There are several high affinity non-selective α2-AR antagonists (e.g. yohimbine, rauwolscine, RX821002, atipamezole and RS79948), although only atipamezole has found a clinical use for reversing the effects of xylazine and dexmedetomidine in veterinary medicine. This role has yet to be established in human medicine. Of the antagonists, BRL 44408 displays some selectivity for α2A-AR and MK-912 and JP1302 for the α2C-AR [126]. Highly selective and potent α2C-AR antagonists are emerging- ORM-10921 and ORM-12741 - that cross the BBB and have potential utility for treatment of cognitive dysfunction and neuropsychiatric symptoms. There are no α2B-AR selective antagonists.

β-Adrenoceptors

β-adrenoceptor subtypes
In 1967, Lands and coworkers [86], comparing rank orders of potency of agonists in a manner similar to that of Ahlquist, concluded that there were two subtypes of the β-AR. The β1-AR, the dominant receptor in heart was equally sensitive to noradrenaline and adrenaline, whereas the β2-AR, responsible for relaxation of vascular, uterine, and airway smooth muscle, was more sensitive to adrenaline than noradrenaline.
Subsequently it became apparent that not all of the β-AR-mediated responses can be classified as either β1 or β2, suggesting the existence of at least one additional subtype [7,23] that was subsequently identified and cloned [50]. The β3-AR is less sensitive to the commonly used β-AR antagonists and was referred to as the "atypical" β-adrenoceptor. However, not all "atypical β-AR responses" could be explained by the β3-AR, so for example there was a suggestion of a fourth β-AR, localized in cardiac tissues of various species [77], activated with low potency by noradrenaline and adrenaline, and blocked by β-AR antagonists such as bupranolol and CGP 20712A [61,135]. Although some of the pharmacology overlapped with the β3-AR, the receptor-mediated responses remained in β3-AR knockout mice [79-80]. Furthermore, this pharmacology was not seen in β1-AR knockout mice. CGP 12177 was originally described as an agonist at the putative β4-AR but is now listed as a partial agonist at the β1-AR in heart. Thus, CGP 12177 binds as a neutral antagonist inhibiting the catecholamines and many agonists with subnanomolar affinity, but has agonist actions at high concentration (30 nM) making this incompatible with a single site of action. Cellular studies demonstrated that whenever the β1-AR was present (rat or human), the secondary site of activation was also present [9,13,65,85,117]. Thus the agonist actions of CGP 12177 result from an action at a non-catecholamine site on the β1-AR [14]. It is now accepted that there are two active sites or conformations of the β1-AR, a catecholamine conformation where most agonists have their actions and readily blocked by most β-AR antagonists, and a secondary site where CGP 12177 (and several other compounds) are agonists and where responses require considerably higher concentrations of β-AR antagonists for inhibition. This secondary conformation also explains the two-component dose-response curves seen with several compounds (e.g. pindolol, alprenolol) [11,13,153]. The secondary site involves amino acids at the extracellular end of TM4. Although pharmacological responses can be demonstrated from this secondary conformation in many species, including human [78], and is conserved across species [14] its clinical relevance is unknown.
The β2-AR was cloned from man [35,82] using probes derived from the hamster β2-AR [45]. However, it proved difficult to clone the β1-AR using this breakthrough, because the human β2-AR cDNA did not cross-hybridize with the β1-AR. The approach that worked utilised the related 5-HT1A receptor [54], that was isolated using the β2-AR cDNA as a probe [83] and then used to probe a human placental cDNA library and identify the β1-AR [59]. The β3-AR was subsequently cloned [50] using the coding regions of the turkey β1-AR and the human β2-AR to screen a human genomic library. Splice variants of the β3-AR have been reported in mouse [53].

β-adrenoceptor expression
β-AR subtypes are important therapeutic targets and are widely distributed in peripheral organs and tissues and in the CNS. β1-AR are present in the heart where stimulation increases heart rate (chronotropy), force of contraction (inotropy), rate of conduction through the AV node (dromotropy) and relaxation during diastole (lusitropy) [77] and relaxation of the coronary arteries. In the kidney, β1-AR stimulation increases renin release from the juxtaglomerular apparatus (JGA) [29]. Other peripheral human tissues with significant populations of β1-AR include adipose tissue and salivary gland. In brain, β1-AR are present in the cerebral cortex, hippocampus, amygdala, pineal, putamen and accumbens.
β2-AR have a wide distribution in the human body including lung, arteries, skeletal muscle, tongue, heart [148], adipose tissue, bone marrow, spleen, gall bladder, brain and adipose tissue. β2-AR are also present in many immune cells. In the lung, β2-AR are present in airway smooth muscle where activation results in the important clinical response of bronchodilation. β2-AR are also present in airway epithelial cells, goblet ells, type II pneumocytes and inflammatory cells (e.g. mast cells and eosinophils). In arteries and veins β2-AR stimulation causes vasodilatation and in the heart positive inotropic and chronotropic responses. Within the heart, 80% of all β-AR are of the β1-AR subtype [24], but the 20% β2-AR in myocardium are more concentrated in the AV node, AV bundle, Purkinje tissue and papillary muscle where they may comprise up to 50-80% of total β-AR present [49,147-148]. In brain, β2-AR are highly expressed in the substantia nigra and hippocampus with more modest expression in amygdala, thalamus, locus coeruleus and spinal cord. In hippocampus, agonist stimulation of the β2-AR located on astrocytes promote glucose uptake and glutamate production and stimulation promotes memory consolidation [31,62]. In bone marrow and spleen, β2-AR are associated with modulation of immune functions and in adipose tissue influence lipolysis. β2-AR activation also produces increased glucose uptake and anabolic effects in skeletal muscle and glycogenolysis and gluconeogenesis in the liver. High expression of β2-AR also occurs in reproductive tissues in the male with significant expression in the penis, prostate and epididymis and in the female in breast, vagina, placenta and uterus where functions are more obscure but likely involve relaxation of smooth muscle.
β3-AR have a localised distribution in humans being present in urinary and gall bladder, with lower expression in fat, intestine and brain [149]. In the urinary bladder β3-AR activation relaxes the detrusor muscle and increases bladder capacity, whereas in gastrointestinal tissues β3-AR mediate relaxation [133]. In females, high concentrations of β3-AR are expressed in the ovary, fallopian tubes, uterine endometrium and placenta [149].
There are considerable differences between rodent and human β3-AR. Rodent β3-AR have several splice variants and ligands display markedly different pharmacological characteristics. β3-AR are highly expressed in rodent white (WAT) and particularly brown (BAT) adipose tissue where they are very important in mediating rodent lipolysis and thermogenesis respectively (the expression and function of β3-AR in these tissues in humans is less important) [140].

β-adrenoceptor signalling
All three β-AR subtypes couple to Gs-proteins to activate adenylyl cyclase. Stimulation of adenylyl cyclase causes the conversion of ATP into cAMP that activates protein kinase A, that in turn phosphorylates several substrates, for example L-type Ca2+ channels. In addition to the PKA pathway cAMP also activates Epac (exchange protein directly activated by cAMP) that modulates cell-type specific protein-protein and protein-lipid interactions that control fine-tuning of important biologic responses [141]. In heart, β1-AR stimulation increases cardiac output, and in blood vessels β1-AR mediated vasodilation. In lungs, β2-AR stimulation of smooth muscle results in bronchodilation and in blood vessels, vasodilatation. In skeletal muscle β2-AR mediated cAMP signalling cases tremor and is also associated with mTORC2 activation that promotes GLUT4 translocation and increased glucose uptake. In adipocytes β3-AR activation leads to phosphorylation and activation of the hormone sensitive lipase and perilipins [36]. Other kinases including ERK1/2, p38MAPK and AMP kinases are also activated and PKA may be involved [36].
Prolonged activation of β2-AR and activation of β3-AR also promotes coupling to Gi in some systems to produce adenylyl cyclase inhibition that reduces the conversion of ATP to cAMP. βγ subunits from Gi are also likely involved in ERK1/2 phosphorylation following β-AR activation [67]. There is also stimulation of guanylyl cyclase (GC) that causes an increase in cGMP levels, and subsequent activation of protein kinase G.
The human β2-AR structure was solved by X-ray crystallography: a 3.4 Ä structure of the wild type β2-adrenoceptor in complex with a conformationally sensitive Fab [82]; and a 2.4 Ä structure of a β2-AR engineered to facilitate crystal formation [34]. This was the first 7-transmembrane receptor for a hormone or neurotransmitter to have its crystal structure solved, and it provided a relevant template for homology models of closely related monoamine and other 7-transmembrane receptors. Agonist, antagonist and G protein receptor complexes have been solved for β1- and β2-AR and a β3-AR structure has been recently described [1,5,43,72,91,93,96-97,102,109,118,132,139,146,150,155,157,161].

Selectivity and clinical uses of drugs acting at β-adrenoceptors
Agonists
Adrenaline and noradrenaline are used clinically to support blood pressure in cases of shock (septic and cardiogenic). Adrenaline is also used in anaphylaxis and in cardiac arrest. The non-selective β-AR agonist isoprenaline is also used to increase heart rate as a bridge to a permanent pacing system in those with profound bradycardia. (-)-Ro 363 is the only β-AR agonist that displays significant selectivity for the β1-AR subtype. Noradrenaline and denopamine display a minor degree of selectivity for β1-AR. Some effort was put into developing somewhat selective β1-AR partial agonists (ligands with intrinsic sympathomimetic activity ISA) with a view to the treatment of heart failure. However clinical trials with xamoterol revealed an increase in mortality in severe heart failure [38]. Xamoterol, denopamine and dobutamine have varying degrees of β1-AR selectivity and previously have been used over short periods to maintain cardiac function.
Considerable effort has been put into developing selective β2-AR agonists, over the last 100 years, that are first line treatments for amelioration of asthma and COPD. Salbutamol and terbutaline are examples of short acting β2-AR agonists (SABAs), salmeterol and formoterol are long acting agonists (LABAs) and indacaterol, olodaterol and vilanterol are ultra-long acting agonists (ultra-LABAs) [19]. Development of β-agonists for asthma was not always straightforward with certain preparations being linked in epidemics of increased asthma death rates. There are several non- or less selective high efficacy β-agonists not in clinical use e.g., fenoterol and cimaterol. Clenbuterol is included on the World Anti-Doping Authority's list of banned substances due to its popularity as a drug used by bodybuilders to increase skeletal muscle mass and for weight loss [76]. However, there is evidence to show that clenbuterol may have potential for the treatment of type II diabetes [76,136].
The species orthologs of the human β3-ARs found in the rat, mouse, and cow have significantly different agonist pharmacology that has proved problematic for drug development. For example, BRL 37344 is a potent full agonist at rodent β3-AR but a weak partial agonist at the human β3-AR [6] and carazolol has a much higher affinity for the bovine receptor [108], whereas [125I]CYP has lower affinity [143]. Carazolol is a potent high efficacy partial agonist at β3-AR but is also a potent partial agonist at β1-AR and a highly potent antagonist at β2-AR [11]. Since the β3-AR largely lacks sites required for receptor phosphorylation it is less susceptible to desensitisation than the other β-AR subtypes. However, desensitisation can occur associated with mRNA and protein down regulation of receptor and post receptor signalling proteins and there is also evidence for cell specific events [112]. Ligands acting at β3-AR can also display ligand-directed signalling [137-138]. Vibegron, and mirabegron are selective β3-AR agonists that potently activate human β3-AR [73]. The identification of high levels of β3-AR mRNA in bladder together with a relaxation response to β3-AR agonists suggested that they could be useful for the treatment of overactive bladder syndrome [99]. Vibegron and mirabegron have been successfully introduced as treatments for this condition [99]. The availability of these human β3-AR selective compounds has also recreated interest in possible metabolic effects of these compounds [41].
The human β3-AR appears to exist in at least two agonist conformations [8], in a similar manner to the β1-AR, due to some residues not conserved in the β2-AR. There is no evidence for a secondary conformation of the β2-AR. At the human β3-AR fenoterol stimulates responses utilising the catecholamine conformation whereas alprenolol, SR58230A and CGP12177 utilise a secondary conformation [8].

Antagonists
propranolol is described as a non-selective β-AR antagonist but is slightly selective for β2-AR and has very low affinity for β3-AR. Carvedilol is a non-selective β-AR and α1-AR antagonist used to treat cardiac failure. It has been suggested to be a β-arrestin biased agonist [160] but this characteristic has been challenged [17] with evidence supporting low efficacy activation of Gs coupled β2-ARs as the explanation. Other β-antagonists commonly used to reduce arrhythmias, in ischaemic heart disease and in heart failure are bisoprolol, metoprolol and nebivolol. Most so-called cardioselective (β1-AR selective) β-AR antagonists such as atenolol and metoprolol display only modest selectivity for β1-AR (bisoprolol is somewhat better) and are associated with side effects due to significant blockade of β2-AR [10]. CGP 20712A is the most selective β1-AR antagonist available [10] and is widely used in in vitro studies but is unsuitable for clinical use. NDD-713 and NDD-825 are high-affinity, β1-AR selective ligands devoid of agonist activity, off-target effects, and toxicology issues, but with good distribution, metabolism and elimination properties [12]. These ligands are largely devoid of β2-AR-mediated adverse effects and may be beneficial in patients with cardiovascular and respiratory disease or limb ischemia. Some β-blockers have other anti-arrhythmic properties (e.g. sotalol, used for paroxysmal AF). The main action of propafenone (class 1C antiarrhythmic) is to block voltage gated Na+ channels (see the Voltage-gated sodium channels family in the Ion Channels section of this website for further details) but is also a weak β-AR antagonist. Labetolol is the β-AR antagonist of choice in pregnancy. Esmolol is a serum esterase sensitive β-AR antagonist and as such only has a short duration of action. It is used in medical emergencies in the operating theatre and in intensive care settings.
ICI 118551 is the most selective β2-AR antagonist currently available but is only used in in vitro and in animal studies [10]. Some clinically available compounds (e.g. timolol) that are used for ischaemic heart disease and glaucoma are somewhat β2-AR selective whilst others used for similar conditions (e.g. betaxolol) are marginally β1-AR selective. There are increasing suggestions that β-AR antagonists (most commonly aimed at β2-AR) may reduce the growth and spread of some cancers.
A number of antagonists such as nadolol, tertatolol and propranolol can behave in some systems as agonists at the β3-AR [21-22,145]. Propranolol is primarily a β1-/β2-AR antagonist with lower affinity for β3-AR. SR59230A that is often described as a selective antagonist at β3-AR displays little selectivity and has similar potency at all 3 subtypes (in fact higher affinity at β2-AR) [11,100] - what it does have is reasonable potency at blocking β3-AR. L-748337 is currently the most selective antagonist at the human β3-AR [11,99,152].

References

Show »

1. Abiko LA, Grahl A, Grzesiek S. (2019) High Pressure Shifts the β1-Adrenergic Receptor to the Active Conformation in the Absence of G Protein. J Am Chem Soc, 141 (42): 16663-16670. [PMID:31564099]

2. Adkins JC, Balfour JA. (1998) Brimonidine. A review of its pharmacological properties and clinical potential in the management of open-angle glaucoma and ocular hypertension. Drugs Aging, 12 (3): 225-41. [PMID:9534022]

3. Ahlquist RP. (1948) A study of the adrenotropic receptors. Am J Physiol, 153 (3): 586-600.

4. Akinaga J, García-Sáinz JA, S Pupo A. (2019) Updates in the function and regulation of α1 -adrenoceptors. Br J Pharmacol, 176 (14): 2343-2357. [PMID:30740663]

5. Alegre KO, Paknejad N, Su M, Lou JS, Huang J, Jordan KD, Eng ET, Meyerson JR, Hite RK, Huang XY. (2021) Structural basis and mechanism of activation of two different families of G proteins by the same GPCR. Nat Struct Mol Biol, 28 (11): 936-944. [PMID:34759376]

6. Arch JR. (2011) Challenges in β(3)-Adrenoceptor Agonist Drug Development. Ther Adv Endocrinol Metab, 2 (2): 59-64. [PMID:23148171]

7. Arch JR, Ainsworth AT, Cawthorne MA, Piercy V, Sennitt MV, Thody VE, Wilson C, Wilson S. (1984) Atypical beta-adrenoceptor on brown adipocytes as target for anti-obesity drugs. Nature, 309 (5964): 163-5. [PMID:6325935]

8. Baker JG. (2005) Evidence for a secondary state of the human beta3-adrenoceptor. Mol Pharmacol, 68 (6): 1645-55. [PMID:16129733]

9. Baker JG. (2005) Site of action of beta-ligands at the human beta1-adrenoceptor. J Pharmacol Exp Ther, 313 (3): 1163-71. [PMID:15716385]

10. Baker JG. (2005) The selectivity of beta-adrenoceptor antagonists at the human beta1, beta2 and beta3 adrenoceptors. Br J Pharmacol, 144 (3): 317-22. [PMID:15655528]

11. Baker JG. (2010) The selectivity of beta-adrenoceptor agonists at human beta1-, beta2- and beta3-adrenoceptors. Br J Pharmacol, 160 (5): 1048-61. [PMID:20590599]

12. Baker JG, Gardiner SM, Woolard J, Fromont C, Jadhav GP, Mistry SN, Thompson KSJ, Kellam B, Hill SJ, Fischer PM. (2017) Novel selective β1-adrenoceptor antagonists for concomitant cardiovascular and respiratory disease. FASEB J, 31 (7): 3150-3166. [PMID:28400472]

13. Baker JG, Hall IP, Hill SJ. (2003) Agonist actions of "beta-blockers" provide evidence for two agonist activation sites or conformations of the human beta1-adrenoceptor. Mol Pharmacol, 63 (6): 1312-21. [PMID:12761341]

14. Baker JG, Proudman RG, Hill SJ. (2014) Identification of key residues in transmembrane 4 responsible for the secondary, low-affinity conformation of the human β1-adrenoceptor. Mol Pharmacol, 85 (5): 811-29. [PMID:24608857]

15. Barends CR, Absalom A, van Minnen B, Vissink A, Visser A. (2017) Dexmedetomidine versus Midazolam in Procedural Sedation. A Systematic Review of Efficacy and Safety. PLoS One, 12 (1): e0169525. [PMID:28107373]

16. Battaglia G, Shannon M, Borgundvaag B, Titeler M. (1983) Properties of [3H]prazosin-labeled alpha 1-adrenergic receptors in rat brain and porcine neurointermediate lobe tissue. J Neurochem, 41 (2): 538-42. [PMID:6308163]

17. Benkel T, Zimmermann M, Zeiner J, Bravo S, Merten N, Lim VJY, Matthees ESF, Drube J, Miess-Tanneberg E, Malan D et al.. (2022) How Carvedilol activates β2-adrenoceptors. Nat Commun, 13 (1): 7109. [PMID:36402762]

18. Berthelsen S, Pettinger WA. (1977) A functional basis for classification of alpha-adrenergic receptors. Life Sci, 21 (5): 595-606. [PMID:20542]

19. Billington CK, Penn RB, Hall IP. (2017) β2 Agonists. Handb Exp Pharmacol, 237: 23-40. [PMID:27878470]

20. Blaxall HS, Murphy TJ, Baker JC, Ray C, Bylund DB. (1991) Characterization of the alpha-2C adrenergic receptor subtype in the opossum kidney and in the OK cell line. J Pharmacol Exp Ther, 259 (1): 323-9. [PMID:1656026]

21. Blin N, Camoin L, Maigret B, Strosberg AD. (1993) Structural and conformational features determining selective signal transduction in the beta 3-adrenergic receptor. Mol Pharmacol, 44 (6): 1094-104. [PMID:7903415]

22. Blin N, Nahmias C, Drumare MF, Strosberg AD. (1994) Mediation of most atypical effects by species homologues of the beta 3-adrenoceptor. Br J Pharmacol, 112 (3): 911-9. [PMID:7921620]

23. Bond RA, Clarke DE. (1988) Agonist and antagonist characterization of a putative adrenoceptor with distinct pharmacological properties from the alpha- and beta-subtypes. Br J Pharmacol, 95: 723-734. [PMID:2905184]

24. Bristow MR, Ginsburg R, Umans V, Fowler M, Minobe W, Rasmussen R, Zera P, Menlove R, Shah P, Jamieson S et al.. (1986) Beta 1- and beta 2-adrenergic-receptor subpopulations in nonfailing and failing human ventricular myocardium: coupling of both receptor subtypes to muscle contraction and selective beta 1-receptor down-regulation in heart failure. Circ Res, 59 (3): 297-309. [PMID:2876788]

25. Bullowa J, Kaplan D. (1903) Treatment of asthmatic attacks: on the hypodermatic use of adrenalin chloride in the treatment of asthmatic attacks. Med News, 83: 787-90.

26. Burnett J. (1903) Adrenalin: a short account of its therapeutic applications. The Medical Times and Hospital Gazette, June 20: 385-7.

27. Bylund DB. (1988) Subtypes of alpha 2-adrenoceptors: pharmacological and molecular biological evidence converge. Trends Pharmacol Sci, 9 (10): 356-61. [PMID:2855960]

28. Bylund DB. (2005) Alpha-2 adrenoceptor subtypes: are more better?. Br J Pharmacol, 144 (2): 159-60. [PMID:15655520]

29. Bylund DB, Eikenberg DC, Hieble JP, Langer SZ, Lefkowitz RJ, Minneman KP, Molinoff PB, Ruffolo Jr RR, Trendelenburg U. (1994) International Union of Pharmacology nomenclature of adrenoceptors. Pharmacol Rev, 46 (2): 121-36. [PMID:7938162]

30. Bylund DB, Ray-Prenger C, Murphy TJ. (1988) Alpha-2A and alpha-2B adrenergic receptor subtypes: antagonist binding in tissues and cell lines containing only one subtype. J Pharmacol Exp Ther, 245 (2): 600-7. [PMID:2835476]

31. Catus SL, Gibbs ME, Sato M, Summers RJ, Hutchinson DS. (2011) Role of β-adrenoceptors in glucose uptake in astrocytes using β-adrenoceptor knockout mice. Br J Pharmacol, 162 (8): 1700-15. [PMID:21138422]

32. Chang DJ, Chang TK, Yamanishi SS, Salazar FH, Kosaka AH, Khare R, Bhakta S, Jasper JR, Shieh IS, Lesnick JD et al.. (1998) Molecular cloning, genomic characterization and expression of novel human alpha1A-adrenoceptor isoforms. FEBS Lett, 422 (2): 279-83. [PMID:9490024]

33. Chen X, Xu Y, Qu L, Wu L, Han GW, Guo Y, Wu Y, Zhou Q, Sun Q, Chu C et al.. (2019) Molecular Mechanism for Ligand Recognition and Subtype Selectivity of α2C Adrenergic Receptor. Cell Rep, 29 (10): 2936-2943.e4. [PMID:31801061]

34. Cherezov V, Rosenbaum DM, Hanson MA, Rasmussen SG, Thian FS, Kobilka TS, Choi HJ, Kuhn P, Weis WI, Kobilka BK et al.. (2007) High-resolution crystal structure of an engineered human beta2-adrenergic G protein-coupled receptor. Science, 318 (5854): 1258-65. [PMID:17962520]

35. Chung FZ, Lentes KU, Gocayne J, Fitzgerald M, Robinson D, Kerlavage AR, Fraser CM, Venter JC. (1987) Cloning and sequence analysis of the human brain beta-adrenergic receptor. Evolutionary relationship to rodent and avian beta-receptors and porcine muscarinic receptors. FEBS Lett, 211 (2): 200-6. [PMID:3026848]

36. Collins S. (2011) β-Adrenoceptor Signaling Networks in Adipocytes for Recruiting Stored Fat and Energy Expenditure. Front Endocrinol (Lausanne), 2: 102. [PMID:22654837]

37. Cotecchia S, Schwinn DA, Randall RR, Lefkowitz RJ, Caron MG, Kobilka BK. (1988) Molecular cloning and expression of the cDNA for the hamster alpha 1-adrenergic receptor. Proc Natl Acad Sci USA, 85 (19): 7159-63. [PMID:2845398]

38. Cruickshank JM. (1993) The xamoterol experience in the treatment of heart failure. Am J Cardiol, 71 (9): 61C-64C. [PMID:8465800]

39. da Silva Junior ED, Sato M, Merlin J, Broxton N, Hutchinson DS, Ventura S, Evans BA, Summers RJ. (2017) Factors influencing biased agonism in recombinant cells expressing the human α1A -adrenoceptor. Br J Pharmacol, 174 (14): 2318-2333. [PMID:28444738]

40. Dale HH. (1906) On some physiological actions of ergot. Am J Physiol, 34 (3): 163-206.

41. Dehvari N, Sato M, Bokhari MH, Kalinovich A, Ham S, Gao J, Nguyen HTM, Whiting L, Mukaida S, Merlin J et al.. (2020) The metabolic effects of mirabegron are mediated primarily by β3 -adrenoceptors. Pharmacol Res Perspect, 8 (5): e00643. [PMID:32813332]

42. Deluigi M, Morstein L, Schuster M, Klenk C, Merklinger L, Cridge RR, de Zhang LA, Klipp A, Vacca S, Vaid TM et al.. (2022) Crystal structure of the α1B-adrenergic receptor reveals molecular determinants of selective ligand recognition. Nat Commun, 13 (1): 382. [PMID:35046410]

43. DeVree BT, Mahoney JP, Vélez-Ruiz GA, Rasmussen SG, Kuszak AJ, Edwald E, Fung JJ, Manglik A, Masureel M, Du Y et al.. (2016) Allosteric coupling from G protein to the agonist-binding pocket in GPCRs. Nature, 535 (7610): 182-6. [PMID:27362234]

44. Diamanti E, Del Bello F, Carbonara G, Carrieri A, Fracchiolla G, Giannella M, Mammoli V, Piergentili A, Pohjanoksa K, Quaglia W et al.. (2012) Might the observed α(2A)-adrenoreceptor agonism or antagonism of allyphenyline analogues be ascribed to different molecular conformations?. Bioorg Med Chem, 20 (6): 2082-90. [PMID:22341244]

45. Dixon RA, Kobilka BK, Strader DJ, Benovic JL, Dohlman HG, Frielle T, Bolanowski MA, Bennett CD, Rands E, Diehl RE et al.. (1986) Cloning of the gene and cDNA for mammalian beta-adrenergic receptor and homology with rhodopsin. Nature, 321 (6065): 75-9. [PMID:3010132]

46. Eason MG, Kurose H, Holt BD, Raymond JR, Liggett SB. (1992) Simultaneous coupling of alpha 2-adrenergic receptors to two G-proteins with opposing effects. Subtype-selective coupling of alpha 2C10, alpha 2C4, and alpha 2C2 adrenergic receptors to Gi and Gs. J Biol Chem, 267 (22): 15795-801. [PMID:1322406]

47. Eason MG, Liggett SB. (1993) Human alpha 2-adrenergic receptor subtype distribution: widespread and subtype-selective expression of alpha 2C10, alpha 2C4, and alpha 2C2 mRNA in multiple tissues. Mol Pharmacol, 44 (1): 70-5. [PMID:7688069]

48. Eason MG, Liggett SB. (1995) Identification of a Gs coupling domain in the amino terminus of the third intracellular loop of the alpha 2A-adrenergic receptor. Evidence for distinct structural determinants that confer Gs versus Gi coupling. J Biol Chem, 270 (42): 24753-60. [PMID:7559592]

49. Elnatan J, Molenaar P, Rosenfeldt FL, Summers RJ. (1994) Autoradiographic localization and quantitation of beta 1- and beta 2-adrenoceptors in the human atrioventricular conducting system: a comparison of patients with idiopathic dilated cardiomyopathy and ischemic heart disease. J Mol Cell Cardiol, 26 (3): 313-23. [PMID:7913135]

50. Emorine LJ, Marullo S, Briend-Sutren MM, Patey G, Tate K, Delavier-Klutchko C, Strosberg AD. (1989) Molecular characterization of the human beta 3-adrenergic receptor. Science, 245 (4922): 1118-21. [PMID:2570461]

51. Erdozain AM, Brocos-Mosquera I, Gabilondo AM, Meana JJ, Callado LF. (2019) Differential α2A- and α2C-adrenoceptor protein expression in presynaptic and postsynaptic density fractions of postmortem human prefrontal cortex. J Psychopharmacol, 33 (2): 244-249. [PMID:30255728]

52. Evans BA, Broxton N, Merlin J, Sato M, Hutchinson DS, Christopoulos A, Summers RJ. (2011) Quantification of functional selectivity at the human α(1A)-adrenoceptor. Mol Pharmacol, 79 (2): 298-307. [PMID:20978120]

53. Evans BA, Papaioannou M, Hamilton S, Summers RJ. (1999) Alternative splicing generates two isoforms of the beta-3 adrenoceptor which are differentially expressed in mouse tissues. Br J Pharmacol, 127: 1525-1531. [PMID:10455305]

54. Fargin A, Raymond SR, Lohse MJ, Kobilka BK, Caron MG, Lefkowitz RJ. (1988) The genomic clone G-21 which resembles aβ-adrenergic receptor sequence encodes the 5-HT1Areceptor. Nature, 335: 358-360. [PMID:3138543]

55. Ford AP, Arredondo NF, Blue Jr DR, Bonhaus DW, Jasper J, Kava MS, Lesnick J, Pfister JR, Shieh IA, Vimont RL et al.. (1996) RS-17053 (N-[2-(2-cyclopropylmethoxyphenoxy)ethyl]-5-chloro-alpha, alpha-dimethyl-1H-indole-3-ethanamine hydrochloride), a selective alpha 1A-adrenoceptor antagonist, displays low affinity for functional alpha 1-adrenoceptors in human prostate: implications for adrenoceptor classification. Mol Pharmacol, 49 (2): 209-15. [PMID:8632751]

56. Ford AP, Daniels DV, Chang DJ, Gever JR, Jasper JR, Lesnick JD, Clarke DE. (1997) Pharmacological pleiotropism of the human recombinant alpha1A-adrenoceptor: implications for alpha1-adrenoceptor classification. Br J Pharmacol, 121 (6): 1127-35. [PMID:9249248]

57. Ford AP, Williams TJ, Blue DR, Clarke DE. (1994) Alpha 1-adrenoceptor classification: sharpening Occam's razor. Trends Pharmacol Sci, 15 (6): 167-70. [PMID:7916507]

58. Frang H, Cockcroft V, Karskela T, Scheinin M, Marjamäki A. (2001) Phenoxybenzamine binding reveals the helical orientation of the third transmembrane domain of adrenergic receptors. J Biol Chem, 276 (33): 31279-84. [PMID:11395517]

59. Frielle T, Collins S, Daniel KW, Caron MG, Lefkowitz RJ, Kobilka BK. (1987) Cloning of the cDNA for the human β1-adrenergic receptor. Proc Natl Acad Sci USA, 84: 7920-7924. [PMID:2825170]

60. Gaertner J, Fusi-Schmidhauser T. (2022) Dexmedetomidine: a magic bullet on its way into palliative care-a narrative review and practice recommendations. Ann Palliat Med, 11 (4): 1491-1504. [PMID:35400162]

61. Galitzky J, Langin D, Verwaerde P, Montastruc JL, Lafontan M, Berlan M. (1997) Lipolytic effects of conventional beta 3-adrenoceptor agonists and of CGP 12,177 in rat and human fat cells: preliminary pharmacological evidence for a putative beta 4-adrenoceptor. Br J Pharmacol, 122 (6): 1244-50. [PMID:9401793]

62. Gibbs ME, Summers RJ. (2005) Contrasting roles for beta1, beta2 and beta3-adrenoceptors in memory formation in the chick. Neuroscience, 131 (1): 31-42. [PMID:15680689]

63. Giovannitti Jr JA, Thoms SM, Crawford JJ. (2015) Alpha-2 adrenergic receptor agonists: a review of current clinical applications. Anesth Prog, 62 (1): 31-9. [PMID:25849473]

64. Graham RM, Perez DM, Hwa J, Piascik MT. (1996) alpha 1-adrenergic receptor subtypes. Molecular structure, function, and signaling. Circ Res, 78 (5): 737-49. [PMID:8620593]

65. Granneman JG. (2001) The putative beta4-adrenergic receptor is a novel state of the beta1-adrenergic receptor. Am J Physiol Endocrinol Metab, 280 (2): E199-202. [PMID:11158920]

66. Gray K, Short J, Ventura S. (2008) The alpha1A-adrenoceptor gene is required for the alpha1L-adrenoceptor-mediated response in isolated preparations of the mouse prostate. Br J Pharmacol, 155 (1): 103-9. [PMID:18552869]

67. Hadi T, Barrichon M, Mourtialon P, Wendremaire M, Garrido C, Sagot P, Bardou M, Lirussi F. (2013) Biphasic Erk1/2 activation sequentially involving Gs and Gi signaling is required in beta3-adrenergic receptor-induced primary smooth muscle cell proliferation. Biochim Biophys Acta, 1833 (5): 1041-51. [PMID:23388888]

68. Han C, Abel PW, Minneman KP. (1987) Heterogeneity of alpha 1-adrenergic receptors revealed by chlorethylclonidine. Mol Pharmacol, 32 (4): 505-10. [PMID:2890094]

69. Hein P, Michel MC. (2007) Signal transduction and regulation: are all alpha1-adrenergic receptor subtypes created equal?. Biochem Pharmacol, 73 (8): 1097-106. [PMID:17141737]

70. Hieble JP, Bylund DB, Clarke DE, Eikenburg DC, Langer SZ, Lefkowitz RJ, Minneman KP, Ruffolo Jr RR. (1995) International Union of Pharmacology. X. Recommendation for nomenclature of alpha 1-adrenoceptors: consensus update. Pharmacol Rev, 47 (2): 267-70. [PMID:7568329]

71. Hirasawa A, Shibata K, Horie K, Takei Y, Obika K, Tanaka T, Muramoto N, Takagaki K, Yano J, Tsujimoto G. (1995) Cloning, functional expression and tissue distribution of human alpha 1c-adrenoceptor splice variants. FEBS Lett, 363 (3): 256-60. [PMID:7737411]

72. Huang J, Chen S, Zhang JJ, Huang XY. (2013) Crystal structure of oligomeric β1-adrenergic G protein-coupled receptors in ligand-free basal state. Nat Struct Mol Biol, 20 (4): 419-25. [PMID:23435379]

73. Igawa Y, Michel MC. (2013) Pharmacological profile of β3-adrenoceptor agonists in clinical development for the treatment of overactive bladder syndrome. Naunyn Schmiedebergs Arch Pharmacol, 386 (3): 177-83. [PMID:23263450]

74. Jasper JR, Lesnick JD, Chang LK, Yamanishi SS, Chang TK, Hsu SA, Daunt DA, Bonhaus DW, Eglen RM. (1998) Ligand efficacy and potency at recombinant alpha2 adrenergic receptors: agonist-mediated [35S]GTPgammaS binding. Biochem Pharmacol, 55 (7): 1035-43. [PMID:9605427]

75. Johnson RD, Minneman KP. (1987) Differentiation of α1-adrenergic receptors linked to phosphatidylinositol turnover and cyclic AMP accumulation in rat brain. Mol Pharmacol, 31: 239-246. [PMID:2436033]

76. Kalinovich A, Dehvari N, Åslund A, van Beek S, Halleskog C, Olsen J, Forsberg E, Zacharewicz E, Schaart G, Rinde M et al.. (2020) Treatment with a β-2-adrenoceptor agonist stimulates glucose uptake in skeletal muscle and improves glucose homeostasis, insulin resistance and hepatic steatosis in mice with diet-induced obesity. Diabetologia, 63 (8): 1603-1615. [PMID:32472192]

77. Kaumann AJ. (1997) Four beta-adrenoceptor subtypes in the mammalian heart. Trends Pharmacol Sci, 18 (3): 70-6. [PMID:9133774]

78. Kaumann AJ, Molenaar P. (1997) Modulation of human cardiac function through 4 beta-adrenoceptor populations. Naunyn Schmiedebergs Arch Pharmacol, 355 (6): 667-81. [PMID:9205950]

79. Kaumann AJ, Molenaar P. (2008) The low-affinity site of the beta1-adrenoceptor and its relevance to cardiovascular pharmacology. Pharmacol Ther, 118 (3): 303-36. [PMID:18501968]

80. Kaumann AJ, Preitner F, Sarsero D, Molenaar P, Revelli JP, Giacobino JP. (1998) (-)-CGP 12177 causes cardiostimulation and binds to cardiac putative beta 4-adrenoceptors in both wild-type and beta 3-adrenoceptor knockout mice. Mol Pharmacol, 53 (4): 670-5. [PMID:9547357]

81. Kenny BA, Chalmers DH, Philpott PC, Naylor AM. (1995) Characterization of an alpha 1D-adrenoceptor mediating the contractile response of rat aorta to noradrenaline. Br J Pharmacol, 115 (6): 981-6. [PMID:7582530]

82. Kobilka BK, Dixon RA, Frielle T, Dohlman HG, Bolanowski MA, Sigal IS, Yang-Feng TL, Francke U, Caron MG, Lefkowitz RJ. (1987) cDNA for the human beta 2-adrenergic receptor: a protein with multiple membrane-spanning domains and encoded by a gene whose chromosomal location is shared with that of the receptor for platelet-derived growth factor. Proc Natl Acad Sci USA, 84 (1): 46-50. [PMID:3025863]

83. Kobilka BK, Frielle T, Collins S, Yang-Feng T, Kobilka TS, Francke U, Lefkowitz RJ, Caron MG. (1987) An intronless gene encoding a potential member of the family of receptors coupled to guanine nucleotide regulatory proteins. Nature, 329 (6134): 75-9. [PMID:3041227]

84. Kobilka BK, Matsui H, Kobilka TS, Yang-Feng TL, Francke U, Caron MG, Lefkowitz RJ, Regan JW. (1987) Cloning, sequencing, and expression of the gene coding for the human platelet alpha 2-adrenergic receptor. Science, 238 (4827): 650-6. [PMID:2823383]

85. Konkar AA, Zhu Z, Granneman JG. (2000) Aryloxypropanolamine and catecholamine ligand interactions with the beta(1)-adrenergic receptor: evidence for interaction with distinct conformations of beta(1)-adrenergic receptors. J Pharmacol Exp Ther, 294 (3): 923-32. [PMID:10945842]

86. Lands AM, Arnold A, McAuliff JP, Luduena FP, Brown Jr TG. (1967) Differentiation of receptor systems activated by sympathomimetic amines. Nature, 214 (5088): 597-8. [PMID:6036174]

87. Langer SZ. (2015) α2-Adrenoceptors in the treatment of major neuropsychiatric disorders. Trends Pharmacol Sci, 36 (4): 196-202. [PMID:25771972]

88. Lanier SM, Downing S, Duzic E, Homcy CJ. (1991) Isolation of rat genomic clones encoding subtypes of the alpha 2-adrenergic receptor. Identification of a unique receptor subtype. J Biol Chem, 266 (16): 10470-8. [PMID:1645350]

89. Lee S. (2019) Dexmedetomidine: present and future directions. Korean J Anesthesiol, 72 (4): 323-330. [PMID:31220910]

90. Lehto J, Hirvonen MM, Johansson J, Kemppainen J, Luoto P, Naukkarinen T, Oikonen V, Arponen E, Rouru J, Sallinen J et al.. (2015) Validation of [(11) C]ORM-13070 as a PET tracer for alpha2c -adrenoceptors in the human brain. Synapse, 69 (3): 172-81. [PMID:25530024]

91. Leslie AG, Warne T, Tate CG. (2015) Ligand occupancy in crystal structure of β1-adrenergic G protein-coupled receptor. Nat Struct Mol Biol, 22 (12): 941-2. [PMID:26643842]

92. Limbird LE. (1988) Receptors linked to inhibition of adenylate cyclase: additional signaling mechanisms. FASEB J, 2 (11): 2686-95. [PMID:2840317]

93. Liu X, Ahn S, Kahsai AW, Meng KC, Latorraca NR, Pani B, Venkatakrishnan AJ, Masoudi A, Weis WI, Dror RO et al.. (2017) Mechanism of intracellular allosteric β2AR antagonist revealed by X-ray crystal structure. Nature, 548 (7668): 480-484. [PMID:28813418]

94. Lomasney JW, Cotecchia S, Lorenz W, Leung WY, Schwinn DA, Yang-Feng TL, Brownstein M, Lefkowitz RJ, Caron MG. (1991) Molecular cloning and expression of the cDNA for the alpha 1A-adrenergic receptor. The gene for which is located on human chromosome 5. J Biol Chem, 266 (10): 6365-9. [PMID:1706716]

95. MacDonald E, Kobilka BK, Scheinin M. (1997) Gene targeting--homing in on alpha 2-adrenoceptor-subtype function. Trends Pharmacol Sci, 18 (6): 211-9. [PMID:9227000]

96. Manglik A, Kim TH, Masureel M, Altenbach C, Yang Z, Hilger D, Lerch MT, Kobilka TS, Thian FS, Hubbell WL et al.. (2015) Structural Insights into the Dynamic Process of β2-Adrenergic Receptor Signaling. Cell, 161 (5): 1101-11. [PMID:25981665]

97. Masureel M, Zou Y, Picard LP, van der Westhuizen E, Mahoney JP, Rodrigues JPGLM, Mildorf TJ, Dror RO, Shaw DE, Bouvier M et al.. (2018) Structural insights into binding specificity, efficacy and bias of a β2AR partial agonist. Nat Chem Biol, 14 (11): 1059-1066. [PMID:30327561]

98. Michel AD, Loury DN, Whiting RL. (1989) Differences between the alpha 2-adrenoceptor in rat submaxillary gland and the alpha 2A-and alpha 2B-adrenoceptor subtypes. Br J Pharmacol, 98 (3): 890-7. [PMID:2556205]

99. Michel MC, Korstanje C. (2016) β3-Adrenoceptor agonists for overactive bladder syndrome: Role of translational pharmacology in a repositioning clinical drug development project. Pharmacol Ther, 159: 66-82. [PMID:26808167]

100. Michel MC, Seifert R. (2015) Selectivity of pharmacological tools: implications for use in cell physiology. A review in the theme: Cell signaling: proteins, pathways and mechanisms. Am J Physiol, Cell Physiol, 308 (7): C505-20. [PMID:25631871]

101. Millan MJ, Maiofiss L, Cussac D, Audinot V, Boutin JA, Newman-Tancredi A. (2002) Differential actions of antiparkinson agents at multiple classes of monoaminergic receptor. I. A multivariate analysis of the binding profiles of 14 drugs at 21 native and cloned human receptor subtypes. J Pharmacol Exp Ther, 303 (2): 791-804. [PMID:12388666]

102. Miller-Gallacher JL, Nehmé R, Warne T, Edwards PC, Schertler GF, Leslie AG, Tate CG. (2014) The 2.1 Å resolution structure of cyanopindolol-bound β1-adrenoceptor identifies an intramembrane Na+ ion that stabilises the ligand-free receptor. PLoS One, 9 (3): e92727. [PMID:24663151]

103. Minneman KP. (1988) Alpha 1-adrenergic receptor subtypes, inositol phosphates, and sources of cell Ca2+. Pharmacol Rev, 40 (2): 87-119. [PMID:2853370]

104. Minneman KP, Han C, Abel PW. (1988) Comparison of α1-adrenergic receptor subtypes distinguished by chloroethylclonidine and WB4101. Mol Pharmacol, 33: 509-514. [PMID:2835650]

105. Morrow AL, Creese I. (1986) Characterization of alpha 1-adrenergic receptor subtypes in rat brain: a reevaluation of [3H]WB4104 and [3H]prazosin binding. Mol Pharmacol, 29 (4): 321-30. [PMID:3010073]

106. Muramatsu I, Oshita M, Ohmura T, Kigoshi S, Akino H, Gobara M, Okada K. (1994) Pharmacological characterization of alpha 1-adrenoceptor subtypes in the human prostate: functional and binding studies. Br J Urol, 74 (5): 572-8. [PMID:7530120]

107. Murphy TJ, Bylund DB. (1988) Characterization of alpha-2 adrenergic receptors in the OK cell, an opossum kidney cell line. J Pharmacol Exp Ther, 244 (2): 571-8. [PMID:2894455]

108. Méjean A, Guillaume JL, Strosberg AD. (1995) Carazolol: a potent, selective beta 3-adrenoceptor agonist. Eur J Pharmacol, 291 (3): 359-66. [PMID:8719421]

109. Nagiri C, Kobayashi K, Tomita A, Kato M, Kobayashi K, Yamashita K, Nishizawa T, Inoue A, Shihoya W, Nureki O. (2021) Cryo-EM structure of the β3-adrenergic receptor reveals the molecular basis of subtype selectivity. Mol Cell, 81 (15): 3205-3215.e5. [PMID:34314699]

110. Newcorn JH, Krone B, Dittmann RW. (2022) Nonstimulant Treatments for ADHD. Child Adolesc Psychiatr Clin N Am, 31 (3): 417-435. [PMID:35697393]

111. O'Connell TD, Jensen BC, Baker AJ, Simpson PC. (2014) Cardiac alpha1-adrenergic receptors: novel aspects of expression, signaling mechanisms, physiologic function, and clinical importance. Pharmacol Rev, 66 (1): 308-33. [PMID:24368739]

112. Okeke K, Angers S, Bouvier M, Michel MC. (2019) Agonist-induced desensitisation of β3 -adrenoceptors: Where, when, and how?. Br J Pharmacol, 176 (14): 2539-2558. [PMID:30809805]

113. Okwundu N, Cline A, Feldman SR. (2021) Difference in vasoconstrictors: oxymetazoline vs. brimonidine. J Dermatolog Treat, 32 (2): 137-143. [PMID:31294643]

114. Oliver G, Schäfer EA. (1894) On the physiological action of extracts of the suprarenal capsule. J Physiol, 16: 1-4.

115. Oliver G, Schäfer EA. (1895) On the Physiological Action of Extracts of Pituitary Body and certain other Glandular Organs: Preliminary Communication. J Physiol (Lond.), 18 (3): 277-9. [PMID:16992253]

116. Oshita M, Kigoshi S, Muramatsu I. (1991) Three distinct binding sites for [3H]-prazosin in the rat cerebral cortex. Br J Pharmacol, 104 (4): 961-5. [PMID:1687370]

117. Pak MD, Fishman PH. (1996) Anomalous behavior of CGP 12177A on beta 1-adrenergic receptors. J Recept Signal Transduct Res, 16 (1-2): 1-23. [PMID:8771528]

118. Pandey S, Saha S, Shukla AK. (2020) Transmitting the Signal: Structure of the β1-Adrenergic Receptor-Gs Protein Complex. Mol Cell, 80 (1): 3-5. [PMID:33007256]

119. Peltonen JM, Pihlavisto M, Scheinin M. (1998) Subtype-specific stimulation of [35S]GTPgammaS binding by recombinant alpha2-adrenoceptors. Eur J Pharmacol, 355 (2-3): 275-9. [PMID:9760042]

120. Perez DM, Piascik MT, Graham RM. (1991) Solution-phase library screening for the identification of rare clones: isolation of an alpha 1D-adrenergic receptor cDNA. Mol Pharmacol, 40 (6): 876-83. [PMID:1661838]

121. Perälä M, Hirvonen H, Kalimo H, Ala-Uotila S, Regan JW, Akerman KE, Scheinin M. (1992) Differential expression of two alpha 2-adrenergic receptor subtype mRNAs in human tissues. Brain Res Mol Brain Res, 16 (1-2): 57-63. [PMID:1334200]

122. Piascik MT, Guarino RD, Smith MS, Soltis EE, Saussy Jr DL, Perez DM. (1995) The specific contribution of the novel alpha-1D adrenoceptor to the contraction of vascular smooth muscle. J Pharmacol Exp Ther, 275 (3): 1583-9. [PMID:8531132]

123. Piascik MT, Perez DM. (2001) Alpha1-adrenergic receptors: new insights and directions. J Pharmacol Exp Ther, 298 (2): 403-10. [PMID:11454900]

124. Pihlavisto M, Sjöholm B, Scheinin M, Wurster S. (1998) Modulation of agonist binding to recombinant human alpha2-adrenoceptors by sodium ions. Biochim Biophys Acta, 1448 (1): 135-46. [PMID:9824686]

125. Piwnica D, Rosignoli C, de Ménonville ST, Alvarez T, Schuppli Nollet M, Roye O, Jomard A, Aubert J. (2014) Vasoconstriction and anti-inflammatory properties of the selective α-adrenergic receptor agonist brimonidine. J Dermatol Sci, 75 (1): 49-54. [PMID:24802713]

126. Proudman RGW, Akinaga J, Baker JG. (2022) The affinity and selectivity of α-adrenoceptor antagonists, antidepressants and antipsychotics for the human α2A, α2B, and α2C-adrenoceptors and comparison with human α1 and β-adrenoceptors. Pharmacol Res Perspect, 10 (2): e00936. [PMID:35224877]

127. Proudman RGW, Akinaga J, Baker JG. (2022) The signaling and selectivity of α-adrenoceptor agonists for the human α2A, α2B and α2C-adrenoceptors and comparison with human α1 and β-adrenoceptors. Pharmacol Res Perspect, 10 (5): e01003. [PMID:36101495]

128. Proudman RGW, Baker JG. (2021) The selectivity of α-adrenoceptor agonists for the human α1A, α1B, and α1D-adrenoceptors. Pharmacol Res Perspect, 9 (4): e00799. [PMID:34355529]

129. Proudman RGW, Pupo AS, Baker JG. (2020) The affinity and selectivity of α-adrenoceptor antagonists, antidepressants, and antipsychotics for the human α1A, α1B, and α1D-adrenoceptors. Pharmacol Res Perspect, 8 (4): e00602. [PMID:32608144]

130. Qu L, Zhou Q, Xu Y, Guo Y, Chen X, Yao D, Han GW, Liu ZJ, Stevens RC, Zhong G et al.. (2019) Structural Basis of the Diversity of Adrenergic Receptors. Cell Rep, 29 (10): 2929-2935.e4. [PMID:31801060]

131. Regan JW, Kobilka TS, Yang-Feng TL, Caron MG, Lefkowitz RJ, Kobilka BK. (1988) Cloning and expression of a human kidney cDNA for an alpha 2-adrenergic receptor subtype. Proc Natl Acad Sci USA, 85 (17): 6301-5. [PMID:2842764]

132. Ring AM, Manglik A, Kruse AC, Enos MD, Weis WI, Garcia KC, Kobilka BK. (2013) Adrenaline-activated structure of β2-adrenoceptor stabilized by an engineered nanobody. Nature, 502 (7472): 575-579. [PMID:24056936]

133. Roberts SJ, Papaioannou M, Evans BA, Summers RJ. (1997) Functional and molecular evidence for beta 1-, beta 2- and beta 3- adrenoceptors in human colon. Br J Pharmacol, 120 (8): 1527-35. [PMID:9113375]

134. Ruuskanen JO, Xhaard H, Marjamäki A, Salaneck E, Salminen T, Yan YL, Postlethwait JH, Johnson MS, Larhammar D, Scheinin M. (2004) Identification of duplicated fourth alpha2-adrenergic receptor subtype by cloning and mapping of five receptor genes in zebrafish. Mol Biol Evol, 21 (1): 14-28. [PMID:12949138]

135. Sarsero D, Molenaar P, Kaumann AJ. (1998) Validity of (-)-[3H]-CGP 12177A as a radioligand for the 'putative beta4-adrenoceptor' in rat atrium. Br J Pharmacol, 123 (3): 371-80. [PMID:9504376]

136. Sato M, Dehvari N, Oberg AI, Dallner OS, Sandström AL, Olsen JM, Csikasz RI, Summers RJ, Hutchinson DS, Bengtsson T. (2014) Improving type 2 diabetes through a distinct adrenergic signaling pathway involving mTORC2 that mediates glucose uptake in skeletal muscle. Diabetes, 63 (12): 4115-29. [PMID:25008179]

137. Sato M, Horinouchi T, Hutchinson DS, Evans BA, Summers RJ. (2007) Ligand-directed signaling at the beta3-adrenoceptor produced by 3-(2-Ethylphenoxy)-1-[(1,S)-1,2,3,4-tetrahydronapth-1-ylamino]-2S-2-propanol oxalate (SR59230A) relative to receptor agonists. Mol Pharmacol, 72 (5): 1359-68. [PMID:17717109]

138. Sato M, Hutchinson DS, Evans BA, Summers RJ. (2008) The beta3-adrenoceptor agonist 4-[[(Hexylamino)carbonyl]amino]-N-[4-[2-[[(2S)-2-hydroxy-3-(4-hydroxyphenoxy)propyl]amino]ethyl]-phenyl]-benzenesulfonamide (L755507) and antagonist (S)-N-[4-[2-[[3-[3-(acetamidomethyl)phenoxy]-2-hydroxypropyl]amino]-ethyl]phenyl]benzenesulfonamide (L748337) activate different signaling pathways in Chinese hamster ovary-K1 cells stably expressing the human beta3-adrenoceptor. Mol Pharmacol, 74 (5): 1417-28. [PMID:18684840]

139. Sato T, Baker J, Warne T, Brown GA, Leslie AG, Congreve M, Tate CG. (2015) Pharmacological Analysis and Structure Determination of 7-Methylcyanopindolol-Bound β1-Adrenergic Receptor. Mol Pharmacol, 88 (6): 1024-34. [PMID:26385885]

140. Schena G, Caplan MJ. (2019) Everything You Always Wanted to Know about β3-AR * (* But Were Afraid to Ask). Cells, 8 (4). [PMID:30995798]

141. Schmidt M, Dekker FJ, Maarsingh H. (2013) Exchange protein directly activated by cAMP (epac): a multidomain cAMP mediator in the regulation of diverse biological functions. Pharmacol Rev, 65 (2): 670-709. [PMID:23447132]

142. Schwinn DA, Lomasney JW, Lorenz W, Szklut PJ, Fremeau Jr RT, Yang-Feng TL, Caron MG, Lefkowitz RJ, Cotecchia S. (1990) Molecular cloning and expression of the cDNA for a novel alpha 1-adrenergic receptor subtype. J Biol Chem, 265 (14): 8183-9. [PMID:1970822]

143. Severi C, Tattoli I, Romano G, Corleto VD, Delle Fave G. (2004) Beta3-adrenoceptors: relaxant function and mRNA detection in smooth muscle cells isolated from the human colon. Can J Physiol Pharmacol, 82 (7): 515-22. [PMID:15389299]

144. Simonneaux V, Ebadi M, Bylund DB. (1991) Identification and characterization of alpha 2D-adrenergic receptors in bovine pineal gland. Mol Pharmacol, 40 (2): 235-41. [PMID:1652052]

145. Strosberg AD. (1997) Structure and function of the beta 3-adrenergic receptor. Annu Rev Pharmacol Toxicol, 37: 421-50. [PMID:9131260]

146. Su M, Zhu L, Zhang Y, Paknejad N, Dey R, Huang J, Lee MY, Williams D, Jordan KD, Eng ET et al.. (2020) Structural Basis of the Activation of Heterotrimeric Gs-Protein by Isoproterenol-Bound β1-Adrenergic Receptor. Mol Cell, 80 (1): 59-71.e4. [PMID:32818430]

147. Summers RJ, Molenaar P, Stephenson JA. (1987) Autoradiographic localization of receptors in the cardiovascular system. TiPS, 8 (7): 272-276. DOI: 10.1016/0165-6147(87)90202-1

148. Summers RJ, Molnaar P, Russell F, Elnatan J, Jones CR, Buxton BF, Chang V, Hambley J. (1989) Coexistence and localization of beta 1- and beta 2-adrenoceptors in the human heart. Eur Heart J, 10 Suppl B: 11-21. [PMID:2553407]

149. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A et al.. (2015) Proteomics. Tissue-based map of the human proteome. Science, 347 (6220): 1260419. [PMID:25613900]

150. Ul-Haq Z, Saeed M, Halim SA, Khan W. (2015) 3D structure prediction of human β1-adrenergic receptor via threading-based homology modeling for implications in structure-based drug designing. PLoS One, 10 (4): e0122223. [PMID:25860348]

151. Urits I, Patel A, Zusman R, Virgen CG, Mousa M, Berger AA, Kassem H, Jung JW, Hasoon J, Kaye AD et al.. (2020) A Comprehensive Update of Lofexidine for the Management of Opioid Withdrawal Symptoms. Psychopharmacol Bull, 50 (3): 76-96. [PMID:32733113]

152. van Wieringen JP, Michel-Reher MB, Hatanaka T, Ueshima K, Michel MC. (2013) The new radioligand [(3)H]-L 748,337 differentially labels human and rat β3-adrenoceptors. Eur J Pharmacol, 720 (1-3): 124-30. [PMID:24183974]

153. Walter M, Lemoine H, Kaumann AJ. (1984) Stimulant and blocking effects of optical isomers of pindolol on the sinoatrial node and trachea of guinea pig. Role of beta-adrenoceptor subtypes in the dissociation between blockade and stimulation. Naunyn Schmiedebergs Arch Pharmacol, 327 (2): 159-75. [PMID:6092972]

154. Weerink MAS, Struys MMRF, Hannivoort LN, Barends CRM, Absalom AR, Colin P. (2017) Clinical Pharmacokinetics and Pharmacodynamics of Dexmedetomidine. Clin Pharmacokinet, 56 (8): 893-913. [PMID:28105598]

155. Weichert D, Kruse AC, Manglik A, Hiller C, Zhang C, Hübner H, Kobilka BK, Gmeiner P. (2014) Covalent agonists for studying G protein-coupled receptor activation. Proc Natl Acad Sci U S A, 111 (29): 10744-8. [PMID:25006259]

156. Weinshank RL, Zgombick JM, Macchi M, Adham N, Lichtblau H, Branchek TA, Hartig PR. (1990) Cloning, expression, and pharmacological characterization of a human alpha 2B-adrenergic receptor. Mol Pharmacol, 38 (5): 681-8. [PMID:2172775]

157. Weiss DR, Ahn S, Sassano MF, Kleist A, Zhu X, Strachan R, Roth BL, Lefkowitz RJ, Shoichet BK. (2013) Conformation guides molecular efficacy in docking screens of activated β-2 adrenergic G protein coupled receptor. ACS Chem Biol, 8 (5): 1018-26. [PMID:23485065]

158. White CW, Choong YT, Short JL, Exintaris B, Malone DT, Allen AM, Evans RJ, Ventura S. (2013) Male contraception via simultaneous knockout of α1A-adrenoceptors and P2X1-purinoceptors in mice. Proc Natl Acad Sci U S A, 110 (51): 20825-30. [PMID:24297884]

159. Williams TJ, Blue DR, Daniels DV, Davis B, Elworthy T, Gever JR, Kava MS, Morgans D, Padilla F, Tassa S et al.. (1999) In vitro alpha1-adrenoceptor pharmacology of Ro 70-0004 and RS-100329, novel alpha1A-adrenoceptor selective antagonists. Br J Pharmacol, 127 (1): 252-8. [PMID:10369480]

160. Wisler JW, DeWire SM, Whalen EJ, Violin JD, Drake MT, Ahn S, Shenoy SK, Lefkowitz RJ. (2007) A unique mechanism of beta-blocker action: carvedilol stimulates beta-arrestin signaling. Proc Natl Acad Sci U S A, 104 (42): 16657-62. [PMID:17925438]

161. Xu X, Kaindl J, Clark MJ, Hübner H, Hirata K, Sunahara RK, Gmeiner P, Kobilka BK, Liu X. (2021) Binding pathway determines norepinephrine selectivity for the human β1AR over β2AR. Cell Res, 31 (5): 569-579. [PMID:33093660]

162. Yamashima T. (2003) Jokichi Takamine (1854-1922), the samurai chemist, and his work on adrenalin. J Med Biogr, 11 (2): 95-102. [PMID:12717538]

163. Yuan D, Liu Z, Kaindl J, Maeda S, Zhao J, Sun X, Xu J, Gmeiner P, Wang HW, Kobilka BK. (2020) Activation of the α2B adrenoceptor by the sedative sympatholytic dexmedetomidine. Nat Chem Biol, 16 (5): 507-512. [PMID:32152538]

How to cite this page

To cite this family introduction, please use the following:

Database page citation (select format):