Abstract
The discovery and characterization of the signal cascades of the β-adrenergic receptors have made it possible to effectively target the receptors for drug development. β-Adrenergic receptors are a class A rhodopsin type of G protein-coupled receptors (GPCRs) that are stimulated mainly by catecholamines and therefore mediate diverse effects of the parasympathetic nervous system in eliciting “fight or flight” type responses. They are detectable in several human tissues where they control a plethora of physiological processes and therefore contribute to the pathogenesis of several disease conditions. Given the relevance of the β-adrenergic receptor as a molecular target for many pathological conditions, this comprehensive review aims at providing an in-depth exploration of the recent advancements in β3-adrenergic receptor research. More importantly, we delve into the prospects of the β3-adrenergic receptor as a therapeutic target across a variety of clinical domains.
Keywords: β1, β2, β3-adrenoceptors; adrenaline; cancer; cardiovascular disease; discovery; eye diseases; metabolic disorders; pathophysiology; pharmacology; therapeutics
1. Introduction
A plethora of biomarker identification and the development of therapeutic drugs have been made possible by the discovery and characterization of G protein-coupled receptors (GPCRs) most especially β-adrenergic receptors which are the best characterized with distinctive features that make them suitable as drug targets. GPCRs and GPCR-related genes constitute ∼17% of the total protein-coding genome and are considered the most important drug targets in medicine and physiology representing 12% of all drug targets [1, 2]. Over the past few decades, in-depth studies on receptor structure, subtypes, cellular localization, receptor activation, and canonical/noncanonical signaling cascades have made it possible to effectively use β3-adrenoceptors as potential targets for treating a variety of harmful conditions ranging from metabolic disorders to cardiovascular and pulmonary disorders. Adrenergic receptors also known as adrenoceptors constitute a rhodopsin class A type of GPCRs that stimulate the sympathetic nervous system (SNS) [3]. Agonists of adrenoceptors include catecholamines (e.g., epinephrine and norepinephrine) and some medications (e.g., β-blockers and α1 and β3 agonists) that are used for the treatment of high blood pressure and asthma among others [4, 5].
The human genome contains 9 adrenoceptor genes that encode 9 different adrenoceptors of which 2 major groups (α- and β-) have been recognized. In 1948, they were grouped into 3 distinct families (i.e., α1-, α2-, and β-adrenoceptors) based on their pharmacological characteristics, sequence similarity, localization, and signaling mechanisms [3, 6]. More recently, each family has further been subdivided into 3 subtypes based on their affinities to adrenergic agonists and antagonists (Figure 1). The α1-adrenoceptor family is predominantly coupled to Gq proteins and has been subdivided into α1A, α1B, and α1D while the α2-adrenoceptor family is usually coupled to Gi/o proteins and has likewise been subdivided into α2A, α2B, and α2C subtypes (Figure 1) [7]. On the other hand, though β1-, β2-, and β3-adrenoceptors of the β-type (based on their affinities to adrenergic agonists and antagonists) are coupled to Gs proteins, β2- and β3-subtypes may also couple Gi/o proteins [8–12]. Ligand or agonist binding on adrenergic receptors therefore trigger intracellular cascade of events through direct interaction of a specific α subunit of the G protein (guanine nucleotide-binding protein) which is dependent on the particular receptor family (Figure 2). Each G protein comprises of 3 distinct subunits named α, β, and γ and is therefore a heterotrimeric complex.
Figure 1.

Classification of adrenergic receptors.
Figure 2.

Major transduction mechanisms of the adrenoceptors. Different adrenoceptor types interact with different G protein types and therefore induce specific intracellular effects. Stimulation of an α1-adrenoceptor leads to conformational changes in the receptor that result in interaction with and subsequent dissociation of the Gαq subunit from the βγ subunits of the heterotrimeric G protein which in turn interacts with and activates phospholipase C (PLC) (left). Active PLC catalyzes the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol 1,4,5-triphosphate (IP3) and diacylglycerol (DAG). Binding of ligands to β-adrenoceptors which are normally coupled to Gαs results in interaction with and subsequent activation of adenylyl cyclase (AC) (right panel). Active AC then synthesizes cyclic adenosine monophosphate (cAMP) from adenosine triphosphate (ATP). However, when α2-adrenoceptor is stimulated (middle panel), it is the Gαi/o subunit that dissociates from the βγ subunits to inhibit AC activity, thus leading to reduced intracellular cAMP concentration. Both β2- and β3-subtypes may also engage the Gαi/o protein.
Adrenoceptors are expressed by most cells of the human body. They are present in almost all peripheral tissues as well as several neurons of the central nervous system (CNS) [13]. Precisely, α1A is predominantly expressed in the cerebral cortex, cerebellum, heart, liver, prostate, and lymphocytes (Table 1). The α1B subtype is expressed in the spleen, kidney, endothelial cells, and osteoblasts and predominantly in the heart accounting for 70%–80% of the cardiac adrenoceptors, whereas α1D is expressed in the cerebral cortex, carotid artery, aorta, lymphocytes, and heart [14–16]. The α2 subtypes are expressed in the brain, spleen, heart, kidney, and liver (Table 1). β3-adrenoceptors are predominantly found in brown adipose tissue, the gut, urinary bladder, and gallbladder. However, both β1- and β2-subtypes are located in the brain, lungs, liver, kidney, spleen, lymphocytes, and skin [16, 17]. Specific subtype activation therefore has a distinct function. For instance, the β1-subtype stimulation results in increased heart rate and cardiac contractility (Table 1). In addition, β1-subtype is reported to enhance cardiac dysfunction following myocardial infarction once over stimulated [18]. The β2-subtype regulates presynaptic norepinephrine release, vasodilation, ventricular function, and bronchodilation among others [19].
Table 1.
Human major tissue distribution and physiological functions of adrenoceptor subtypes.
| AR subtype | Tissue distribution | Physiological function |
|---|---|---|
| α1A | Cerebral cortex, cerebellum, heart, liver, prostate, lymphocytes | Contraction of urethral smooth muscle, contraction of arterial smooth muscle |
| α1B | Spleen, kidney, endothelial cells, heart osteoblasts, lymphocytes, somatic veins, and arteries | Contraction of arteries/veins, osteoblastic cell proliferation, promotes cardiac remodeling |
| α1D | Cerebral cortex, carotid artery, aorta, lymphocytes, heart, prostate, blood, vessels, bladder, platelets | Ureteral muscle contraction, contraction of arteries, vasoconstriction of aorta and coronary artery |
| α2A | Brain, kidney, liver, heart, spleen, aorta, lung, skeletal muscle, platelets sympathetic neurons, pancreas | Inhibition of presynaptic norepinephrine release, vasoconstriction of vessels in skeletal muscle, involved in baroreceptor reflex, sedation, hypotension, analgesic |
| α2B | Brain, kidney, liver, heart, lung, aorta, skeletal muscle, spleen | Vasoconstriction |
| α2C | Brain, kidney, liver, heart, spleen, aorta, lung, skeletal muscle, platelets, pancreas, coronary and CNS vessels, sympathetic neurons | Inhibition of presynaptic norepinephrine release, analgesic, sedation |
| β1 | Brain, lungs, liver, kidney, spleen, skin, adipose tissue, lymphocytes, coronary arteries, intestinal muscle | Increase heart rate (conduction, contractility, automaticity), renin release, lipolysis, vasodilation, relaxation of intestinal muscle |
| β2 | Brain, lungs, liver, lymphocytes, skin, heart, eye, vascular smooth muscle, uterus, bladder, adipocytes, sympathetic terminal | Vasodilation, bronchodilation, bladder and uterus relaxation, lipolysis, glycogenolysis, positive ionotropic/chronotropic, norepinephrine release |
| β3 | Adipose tissue, brown tissue, gut, urinary bladder, gall bladder, eye, small intestine | Thermogenesis, lipolysis, myometrium relaxation, vasodilation of coronary arteries, negative ionotropic effect |
Abbreviations: AR, adrenoceptor; CNS: central nervous system.
The β3-adrenoceptor is the most recently discovered of all adrenoceptor subtypes and has received less attention than both β1- and β2-adrenoceptors and therefore is the least studied so far among the β-subtypes [20]. To date, no literature has reported a crystal structure of the β3-adrenoceptor [21]. Perhaps, lack of the most suitable technology, lack of high affinity and high-avidity antibodies, animal and cellular models, and interspecies differences may explain this drought of information [22]; however, a substantial amount of literature over the past 3 decades has unraveled the β3 subfamily as an emerging target for novel pharmacological approaches in several clinical disciplines which has aroused a lot of interest among many scientists. Though not found in the Protein Data Bank (PDB), Nagiri et al. [23] recently reported a cryogenic electron microscopy structure of the β3-adrenoceptor bound Gs protein with agonist mirabegron revealing the molecular basis for β-adrenoceptor subtype selectivity that will allow the design of more selective drugs with fewer adverse effects. For instance, pharmacological agonists such as SR 58611, BRL 37344, and CGP 12177 activate the β3-adrenoceptor but have little effect on β1- and β2-adrenoceptors [24]. On the other hand, some agonists such as mirabegron (YM178) and solabegron (GW427353) are species-dependent and have higher affinity for the human β3‐adrenoceptor than that of rodent. Conversely, others such as BRL 27344, ritobegron (KUC-7483), and CL 316243 apparently have higher affinity for the rodent than the human β3‐adrenoceptor [20, 25]. Compared to both β1 and β2, β3‐adrenoceptor requires higher quantities of catecholamines to be stimulated [22].
β3‐adrenoceptors as forementioned relatively have a more restricted expression pattern in humans compared with β1‐ and β2‐adrenoceptors [26]. β3‐Adrenoceptors also have a unique ligand recognition profile for several antagonists including propranolol previously considered to inhibit all β‐adrenoceptor subtypes [27]. Besides, β3‐adrenoceptor agonists are currently preferred for development of therapeutics for metabolic diseases since norepinephrine and other nonselective derivatives have adverse side effects on cardiovascular tissue which have limited their use.
In this review, we provide a current insight on the structure, signaling, and functions of the β-adrenoceptors. We particularly explore the emerging potential of the β3-adrenoceptor as a versatile therapeutic target, comprehensively shedding more light on its implications for cancer therapeutics, metabolic disorders, neurological conditions, pulmonary disorder, asthma, and chronic obstructive pulmonary disease (COPD) among others.
2. Discovery of the β3-Adrenoceptor
The history of the discovery of the β-adrenoceptors is well documented by several scientists [28, 29]. Briefly, in the latter parts of the 1800s, John Newport Langley (1852–1925) first postulated the concept of specific receptors on the cell that bind drugs or substances and by so doing initiate biological effects or instead inhibit downstream cellular functions [30, 31]. Paul Ehrlich (1854–1915) about the same time observed that these receptors were selective [32]. In 1897, John Jacob Abel (1857–1938) successfully isolated an impure form of would be “epinephrine” from the adrenal gland of sheep that regulated blood pressure as an agonist on these receptors [17, 33, 34]. However, these concepts were not fully accepted by the scientific community until in 1948 when Alquist [6] clarified adrenaline's actions on 2 distinct receptors, the so called α- and β-receptors, and further established the concept that a single sympathetic mediator produced both excitatory and inhibitory responses. Lands and colleagues [35] in 1967 suggested subdividing the β-type adrenoceptors into 2 broad categories β1- and β2- based on differential order of potencies against 12 agonists and antagonists in several organ bath preparations [35]. Barely 5 years after this subclassification, scientists provided experimental evidence to demonstrate that some β‐adrenergic–like responses were not mediated by either β1 - or β2-adrenoceptors [36], such as lipolytic responses in rat white adipose tissue [37], smooth muscle relaxation responses in rat colon [38], and human urinary bladder [39], which looked to be mediated by β‐adrenoceptors based on the order of potency of catecholamines. It was not until 1984 when Arch [40] provided the first evidence for the existence of β3-adrenoceptors by demonstrating a new class of β-adrenoceptor ligands that selectively induced lipolysis in rat brown adipocytes. They showed that a series of β-adrenoceptor ligands (BRL 26830A, BRL 33725A, and BRL 35135A) had remarkable antiobesity, antidiabetic, and thermogenic effects on mice β3-adrenoceptor with severe obesity and diabetes rather than classical β-adrenoceptors. However, the existence of a third subtype, the β3‐adrenoceptor, only became fully accepted after it was cloned in 1989 by Emorine et al. [41]. Since its discovery, research over the past 3 decades has unraveled new insights relating the β3-adrenoceptor signaling pathway (Figure 3) and β3-adrenoceptor–mediated functions (Table 1). Recently, for instance, involvement of the β3-adrenoceptor in urine concentration mechanism, fat mass reduction, and inflammatory processes has been described giving more opportunities for new therapeutic applications, and hopefully more details will be discovered in the near future [42–45]. A proposed fourth subtype (β4-adrenoceptor) has remained controversial to date [46, 47].
Figure 3.

β 3-Adrenoceptor signaling and desensitization. Stimulation of the β3-adrenergic receptor through catecholamine binding results in a rise in intracellular cAMP concentration which binds and activates PKA. Activated PKA phosphorylates and activates lipases (e.g., HSL, PLIN, and CGI-58) to increase adipocyte lipolysis releasing free fatty acids (FFAs) and glycerol. PKA also phosphorylates CREB which translocates into the nucleus. CREB and ATF-2 (also known as CRE; cAMP response element) together form a transcription factor that regulates expression of thermogenic genes. In homologous desensitization, cAMP also activates the EPAC/RAP pathway which is synergized by tumor necrosis factor receptor (TNFR) activation (i.e., heterologous desensitization) through TNF-α binding. Activated RAP2A in turn activates PLC which hydrolyzes PIP2 into IP3 and DAG. IP3's action on endoplasmic reticulum increases intracellular Ca2+ concentration which induces transcription of TRIB1 gene. Resulting TRIB1 protein recruits COP-1, an E3 Ub ligase whose activity leads to the degradation of CEBPα and subsequent downregulation of ADRB3 gene expression.
3. Gene and Protein Structure
3.1. The β3-Adrenoceptor Gene
The β3-adrenoceptor gene (ADRB3 or BETA3AR) is located on the short arm of chromosome 8 in humans (8p12–p11.2; cytogenetic location: 8p11.23). Besides humans, β3-adrenoceptor has also been identified in other mammalian species including the rat [48], mouse [49], bovine, sheep, goat [50], and dog [51]. The mouse and human β3-adrenoceptor genes are 81% identical with the highest homology of 94% within the transmembrane (TM) domains and lowest at the C-terminus and third intracellular loop [20]. The mRNA transcript-start site for both mouse and human ADRB3 genes is between 150 and 200 nucleotides upstream of the ATG translation start codon. Compared with β1- and β2- which contain 1 exon each, the β3-adrenoceptor genes comprise 2 exons (Table 2). In humans, a large exon (1.4 kb) encodes the first 402 residues. The first 388 amino acid residues of the mouse β3-adrenoceptor are encoded by this exon [52]. In humans, ADRB3 gene is made of 1 intron which is absent in both β1- and β2-adrenoceptors. The second exon (700 bp) in humans encodes the 6 C-terminus residues of the β3-subtype. In the mice, the second exon which is 68 bp encodes the 12 C-terminus residues while the third exon contains the β3-adrenoceptor mRNA 3′ untranslated region [52]. The rat ortholog was found to contain 3 exons and 2 introns [53] having an extra exon and intron over that of humans. As such, the human, mouse, and rat β3-adrenoceptor primary sequences differ markedly in length and essentially at the C-terminus region of the receptor [52]. Three subtypes of β3-adrenoceptors (A, B, and C forms) have been described resulting from an alternative splicing of the mRNA which arises from alternative promoters or polyadenylation signals in a tissue-dependent manner [40]. The primary sequence of form A consists of 396 amino acids, whereas forms B and C, respectively, contain additional 12 and 6 amino acids besides the C-terminus amino acid [49, 54]. The ADRB3 gene product (the β3-adrenoceptor) belongs to a family of β-adrenergic receptors (Figure 1) that mediate catecholamine-induced signaling. The receptor is mainly located in the adipose tissue where it is involved in the regulation of lipolysis and thermogenesis. The structure and function of the receptor has now been widely characterized.
Table 2.
Characteristic differences between human β-adrenoceptors.
| Property | Receptor subtype | ||
|---|---|---|---|
| β 1 | β 2 | β 3 | |
| No. of amino acids | 477 | 413 | 408 |
| No. of introns | — | — | 1 |
| No. of exons | 1 | 1 | 2 |
| Phosphorylation by PKA and β-ARK | Yes | Yes | No |
| Effector enzyme | AC | AC | AC, NO synthase |
| G protein type | Gs | Gs or Gi | Gs or Gi |
| Sequence homology | 54% with β2 | 46% with β3 | 51% with β1 |
Abbreviations: AC, adenylyl cyclase; NO, nitric oxide.
3.2. Protein Structure of the β3-Adrenoceptor
The β3-adrenoceptor is a 408 amino acid glycoprotein found in membranes of several cell types and tissues. The extracellular N-terminus of the β3-adrenoceptor, like β1- and β2-, is located outside the cell membrane and is glycosylated (Figure 2). It is required for ligand recognition and binding [55]. Characteristic of all members of the GPCR superfamily, adrenoceptors contain 7 TM helices (TM1-TM7) made of ∼22–28 hydrophobic amino acids which stretch across the cell membrane in a serpentine fashion to form a stable structure within the lipid bilayer. The structure therefore has 3 intracellular and 3 extracellular loops connecting the 7 TM domains. Ligands bind to the TM helices in the crevices formed by the helices. Specific amino acid residues in each helix interact with ligands, allowing for precise recognition and binding. This region (the binding pocket) is highly selective, thereby enabling the receptor to differentiate between ligands [55]. Ligand binding and subsequent activation of the receptor are essentially dependent on the formation of a disulfide bond between Cys110 and Cys189, respectively, located within the second and third extracellular loops [21, 24]. The intracellular loops connect the cytoplasmic ends of adjacent helices between TM helices. Conformational changes in these loops facilitate interaction with G protein subunits, resulting in stimulation of downstream intracellular signaling cascades upon ligand binding [55, 56].
The C-terminus of the receptor is intracellular. It is essential for receptor signaling and regulation [56]. Various proteins bind to the C-terminus, including G proteins and other signaling molecules, modulating receptor activity and downstream signaling events. Interactions at the C-terminus are required for the receptor to effectively transmit signals [55]. β-Adrenoceptors are coupled mostly to Gs-protein (rarely to Gi-protein) and stimulate activation of the effector enzyme (second messenger), AC (Figure 2). An initial computer modeling revealed an image of the ligand-binding site and β3 receptor-ligand interactions (source: https://www.uni-graz.at/%7Ebinder/science/b3adrenoceptors.html) [22]. Asp117 (within TM3) was identified to be involved in ligand binding via mutagenesis. Other amino acids implicated in ligand binding include Ser169 (TM4), Ser209, Ser212 (TM5), and Phe309 (TM6). TM2 and 7 are required for Gαs subunit binding as well as receptor activation possibly through Asp83 (TM2) and Tyr336 (TN7) [22, 56].
Like most GPCRs, the β-adrenoceptors are modified by Cys palmitoylation referred to as S-palmitoylation (Figure 2, right panel). Protein S-palmitoylation is a covalent and reversible lipid post-translational modification of a eukaryotic protein in which the lipid is specifically targeted for Cys residues of the protein. S-palmitoylation is catalyzed by palmitoyltransferases in mammals in which palmitate is invariably attached covalently to a specific Cys residue of a protein forming a thioester bond. The reverse reaction in which the lipid is removed from the protein is catalyzed by acyl-protein thioesterases. S-palmitoylation mostly occurs in membrane proteins such as cell surface receptor proteins. Essentially, S-palmitoylation increases hydrophobicity of proteins, thereby enhancing their membrane association. Additionally, S-palmitoylation is thought to enhance subcellular localization, protein–protein interactions, and binding capacities of a protein.
Both human and mouse β-receptors are modified at the canonical S-palmitoylation site, i.e., Cys358 (located in the fourth intracellular loop) within the C-terminal tail; however, additional sites such as Cys361 in mouse and Cys153, 292, 363 in human β3-receptors which are differentially S-palmitoylated have recently been described [57]. Palmitoylation of Cys361 residue has been demonstrated to mediate ligand-induced activation of AC (Figure 4) [58]. The pharmacology of β-adrenoceptors has been extensively reviewed [20, 59–62]. Apart from the structural distinction between β3- and β1-/β2-receptors, β3's pharmacological profile is also somewhat different. β3-Adrenoceptor shares 51% amino acid sequence similarity with the β1- and 46% similarity with β2-adrenoceptor, whereas β1 and β2 share 54% sequence homology (Table 2) [24, 63–65]. The main distinction between β3- and β1-/β2-adrenoceptors is that the third intracellular loop and the C-terminus domain of the β3-adrenoceptor lack phosphorylatable Ser and Thr sites for cAMP-dependent protein kinase A (PKA) and other GPCR kinases such as the β-receptor kinases (β-ARKs) present in β1/β2-adrenergic receptors (Table 2). PKA and β-ARK–dependent phosphorylation of these residues is required for rapid receptor desensitization and subsequent downregulation (Figure 4, Section 4.1) because it induces conformational changes at the C-terminal tail that prevents receptor-G protein binding and instead promotes β-arrestin binding of C-terminal tail of the receptor which triggers the internalization process [1]. To activate AC, the G protein must first interact with the C-terminal tail of the receptor which enables the G protein to exchange ADP for ATP with a consequential dissociation of the Gαs subunit from the βγ complex. Interaction of the Gαs subunit with AC activates AC. Activated AC then catalyzes the synthesis of cAMP from ATP which in turn activates several downstream cascades. Phosphorylation of receptor by PKA and β-ARK prevents this receptor-G protein coupling, thereby downregulating the receptor. This phenomenon makes the β3-adrenoceptor comparatively more resistant to agonist-induced desensitization [28, 64, 66, 67] as PKA and β-ARK–dependent phosphorylation of sites is absent in the receptor.
Figure 4.

β-Adrenoceptor activation and regulation. When intracellular cAMP concentration rises resulting from receptor activation, cAMP molecules bind the regulatory subunits of PKA resulting in the release of active catalytic subunits of PKA which migrate into the nucleus where they catalyze phosphorylation of CREB. Once phosphorylated, CREB, a signal-regulated transcription factor, recruits a coactivator CBP which together induce gene transcription upon binding CRE. Activated PKA subunits may phosphorylate the receptor directly on Ser and Thr residues of intracellular loops or indirectly via phosphorylation of β-ARK. Phosphorylation activates β-ARK which in turn phosphorylates Ser and Thr residues of the β-adrenoceptor upon recognizing its stimulated configuration which decouples the Gs subunit from the receptor by steric exclusion. β-Arrestins bind the phosphorylated receptor. This prevents the receptor from further interaction with the Gs subunit of the trimeric G protein and acts as an adapter to initiate receptor endocytosis, subsequently sequestering and desensitizing the receptor (right panel).
Interestingly, this characteristic, among others, renders the β3-adrenoceptor a unique therapeutic target for treatment of several chronic conditions [27, 68]. Structural homology between β1- and β2-receptors is 54% (Table 2) [24]. Structural homologies of β3-adrenoceptor of the human, bovine, monkey, hamster, guinea pig, rat, and mouse are considerably higher (∼80%–90%) [69]. These homologies are somewhat restricted to the TM domains, the membrane-bound proximal regions of the intracellular loops, and extracellular regions of the receptor responsible for ligand binding [63, 64]. β-Adrenoceptor signaling is mediated by AC activation via G proteins following ligand binding of receptor.
4. β-Adrenoceptor Signaling
The SNS is one of the 3 anatomically distinct divisions of the autonomic nervous system responsible for regulation of a plethora of homeostatic mechanisms including cardiac function (heart rate acceleration), gastrointestinal responses to food, urinary bladder contraction, thermoregulation, constriction of blood vessels, sweating, activation of goose bumps, pupil dilation (focusing), airway reactivity, glycogenolysis, and dilation of blood vessels in skeletal muscles [70–72]. Its main function is stimulation of the “fight-or-flight response” of the body. Catecholamines such as epinephrine (adrenaline) synthesized and released by the adrenal medulla, norepinephrine (noradrenaline) produced by sympathetic nerve endings (to a small extend) but majorly by the adrenal medulla, and dopamine synthesized in the substantia nigra, ventral tegmental area, and hypothalamus of the brain constitute the main stimulators of the SNS [73, 74]. These catecholamines circulate throughout the body and act on adrenergic receptors to elicit tissue-dependent responses.
When a specific catecholamine or agonist approaches a β-adrenoceptor, it interacts with the extracellular domain causing the receptor to activate [55]. The interaction allows the receptor to distinguish between different ligands, thereby initiating the specific corresponding signaling process [56, 75]. Ligand binding elicits complex changes in the conformation of the receptor resulting in receptor activation and subsequent initiation of the downstream intracellular signaling pathways (Figure 4). This change is pivotal in receptor activation because it exposes specific regions of the receptor downstream enabling them to likewise interact with a specific heterotrimeric G protein type within the intracellular compartment of the cell membrane. This interaction takes place at the intracellular loops as well as the C-terminus and is highly selective (Figure 4). β3-Adrenoceptor signals mostly through the activation of the Gαssubunit which in turn dissociates from the Gβγ subunit complex and interacts with and activates AC. Active AC then catalyzes the conversion of ATP to cAMP leading to a cytosolic rise in cAMP concentration [75]. However, β2/β3-adrenoceptor may also engage the Gαi/o to downregulate AC activity and consequential reduction in cAMP levels.
G proteins (of which there are over 1000 types encoded in humans) are signaling molecules that transmit activation signals from receptors (i.e., GPCRs) to various intracellular effectors that ultimately result in a diverse range of cellular responses [54]. They are so called because they bind guanine nucleotides mainly guanosine diphosphate (GDP) and guanosine triphosphate (GTP) [76, 77]. As cytosolic cAMP levels rise, cAMP molecules bind the regulatory subunits of PKA leading to its activation and cAMP-dependent phosphorylation of gated ion channels [1, 78, 79]. Conformational changes in PKA due to cAMP binding result in the release of the regulatory subunits from PKA's catalytic subunits leading to its activation. Activated PKA catalytic subunits then migrate into the nucleus and phosphorylate CREB (cAMP response element-binding gene regulatory protein). Once phosphorylated, CREB recruits a coactivator CBP (CREB-binding protein) which together stimulate gene transcription upon binding CRE (CREB-binding element) (Figure 4).
4.1. β3-Adrenoceptor Desensitization
Receptor desensitization refers to a decreased responsiveness of a receptor following chronic or repeated exposure to an agonist and is a general characteristic of most GPCRs including α2- and β-adrenoceptors [80]. β1- and β2-adrenoceptors undergo classical β-arrestin–mediated desensitization (Figure 4), minutes after agonist stimulation to prevent hormonal overload [64]. Receptor desensitization is dependent on adrenoceptor phosphorylation by PKA, protein kinase C (PKC), or G protein linked receptor kinases (GRKs, β-ARKs) [1, 81]. Primarily, β-ARKs which recognize the agonist-occupied configuration of the receptors phosphorylate Ser and Thr residues of α2-, β1-, or β2-adrenoceptors. Once phosphorylated, the receptor binds an arrestin family of proteins with high affinity promoting uncoupling of the receptor from the stimulatory G protein (Gαs). This prevents the receptor from further interaction with Gαs or serves as an adapter to couple receptor clathrin-coated pits to induce receptor-mediated endocytosis. These interactions ensure reduced receptor response and increased internalization and sequestration resulting from acute or chronic agonist exposure [73, 82–84].
Previous investigations in contrast demonstrated that β3-adrenoceptor fails to desensitize following acute exposure to agonists which is attributable to the lack of most of the structural determinants (i.e., the C-terminal β-arrestin–binding motif and the key phosphorylation sites) (Section 3.2, Table 2) that contribute to agonist-induced receptor internalization and consequent desensitization in α2-, β1-, and β2-adrenoceptors [63, 64, 66, 67, 85]. The β3-adrenoceptor, however, has been shown to desensitize only after long-term agonist-induced activation periods (hours to days) in some cell types which is suggestive of the involvement of cell- and/or species-specific mechanisms [86]. How then is the β3-adrenoceptor desensitized? What are the molecular alterations involved? Many studies have speculated about different desensitization mechanisms. Some have demonstrated a downregulation of β3‐adrenoceptor mRNA through decreased transcription of the ADRB3 gene by exposure to some ligands such as tumor necrosis factor alpha (TNF-α) [28, 87–89]. However, whether reduced β3‐adrenoceptor mRNA synthesis necessarily translates into changes in the expression of the functional receptor protein remains controversial [28, 90]. This is because β3‐adrenoceptor–mediated function can diminish without any receptor protein downregulation suggesting that changes at the post‐receptor level may be contributing to receptor desensitization [91–94].
When stimulated, the β3‐adrenergic receptor interactions with G proteins subsequently result in a rise in intracellular cAMP concentration which bind and activate PKA (Figure 3). Phosphorylation of hormone-sensitive lipase (HSL) and perilipin (PLIN) by activated PKA increases lipolysis [94, 95] and also stimulates thermogenic gene (FGF21, IRF4, ZFP516, etc.) transcription via activation of the ATF-2 (activating transcription factor 2) and the CREB [96, 97].
Recently, Valentine et al. [85] provided a model for the signaling events that ultimately lead to both homologous (which is dependent on β3‐adrenoceptor activation) and heterologous (which is dependent on the inflammatory cytokine, TNF-α) desensitization of the β3‐adrenoceptor in adipocytes. They demonstrated that both homologous and heterologous signal cascades merge at the EPAC (exchange protein activated by cAMP)-RAP2A (Ras-related protein Rap-2a)-PI-PLC (phosphoinositide-phospholipase C) pathway to activate a cascade of transcriptional events which lead to the repression of the ADRB3 gene expression through targeted degradation of CEBPα (CCAAT enhancer binding protein alpha) by Ca2+ dependent induction of TRIB1 (Tribbles pseudokinase 1) gene expression in adipocytes in response to TNF-α or high-fat diet (HFD) exposure. TRIB1 recruits COP-1 (constitutive photomorphogenesis protein 1), an E3 Ub ligase whose activity results in the degradation of CEBPα and subsequent downregulation of ADRB3 gene expression [98] leading to catecholamine resistance of the receptor (Figure 3). Therefore, this pathway may be therapeutically relevant in the treatment of obesity and other related metabolic diseases [85, 99, 100].
5. Pathophysiology and Therapeutic Potential of the β-Adrenoceptor
β-Receptors are detectable in several human tissues. They mediate catecholamine signaling to regulate a surfeit of homeostatic functions of the SNS in the myocardium, retina, myometrium, adipose tissue, gallbladder, brain, urinary bladder, and blood vessels among others [20]. As such, β-adrenoceptors have become attractive drug targets. Agonists and antagonists of adrenergic receptors are being targeted for the treatment of a variety of pathologies including overactive bladder, heart failure, metabolic syndromes (obesity, type 2 diabetes, etc.), hypertension, glaucoma, hypotension, angina pectoris, asthma, different cancers (colon carcinoma, breast cancer, etc.), premature labor, cachexia, control of colon motility, anxiety, and depressive disorders. They are used as adjuncts to general anesthetics, as vasoconstrictors, bronchodilators, etc. In addition, they are targeted for opioid withdrawal, for relaxation of uterine smooth muscle, relief of allergic states (including anaphylaxis), and for CNS stimulation.
5.1. Targeting the β-Adrenoceptor in Urinary Disorders
Urinary disorders, encompassing the syndrome of overactive bladder (nocturia, urgency, with or without incontinence) including stress incontinence, significantly impact patients' quality of life. Generally, lower urinary tract dysfunctions increase in the elderly and postmenopausal populations and are attributable to hormone imbalances which may induce changes in bladder contractile and/or relaxing mechanisms. The bladder detrusor muscles relax during storage to accommodate rising volumes of urine to acceptable pressure while the neck of the bladder and urethra contract in order to resist involuntary urine leakage. However, during voluntary micturition (urine expulsion), the bladder neck and urethra muscles instead relax enabling the detrusor muscles of the bladder to contract in order to void urine without much resistance [101]. Detrusor muscle relaxation can be induced by stimulation of all 3 adrenergic receptor subtypes. However, previous studies identified the β3-adrenoceptor as the most important adrenoceptor in human detrusor muscle relaxation [102, 103]. Since then, researchers have focused on developing innovative therapies aimed at restoring normal urinary function [101]. Later, researchers demonstrated that sympathetic system–mediated relaxation of the urinary bladder [104, 105] is dependent on cAMP-mediated β3-adrenoceptor-Gs coupled signaling in mammals such as humans, monkeys, and dogs [106–108]. β3-Adrenoceptors which are expressed in the smooth muscles of the detrusor mediate muscle relaxation, thereby facilitating the filling capacity of the bladder [109, 110]. Location of β3-adrenoceptors, considerably the most prevalent of the 3 subtypes in the lower urinary tract and urethra, is strategic for regulation of the urothelium and prostate function [111, 112]. β3-Adrenoceptor–specific agonists used clinically today capitalize on this mechanism to improve overactive bladder conditions to prevent urinary incontinence [113]. As such, selective β3-adrenoceptor agonists, e.g., BRL 37344, CL 316243, FK175, or YM178, enhance bladder function by decreasing voiding frequency [114–116].
Similarly, Kullman et al. [115] assessed the ability of β3-adrenoceptor agonists to treat bladder dysfunction (i.e., voiding frequency, urgency, and incontinence). Using Sprague Dawley rat ovariectomy model, they demonstrated that depletion of ovarian hormone increased voiding frequency and reduced bladder volume by about 25% in awake rats and induced irregular cystometrograms in urethane-anesthetized rats even though RT-PCR results of bladder tissue indicated no significant difference in the expression patterns of all 3 β-subtypes compared with SHAM rats. Immunostaining showed localization of the β3-subtype in urothelium and detrusor muscle indicative of strategic localization for effective regulation of detrusor muscle relaxation. In addition, they confirmed the selectivity of BRL 37344, TAK-677, and FK175 to β3-subtype in CHOK1 cells overexpressing all 3 β-subtypes by assessing the relative potency of cAMP concentrations. They provided evidence that intravenous administration of 0.1–500 μg/kg of the β3-adrenoceptor agonists in urethane-anesthetized rats decreased voiding frequency, bladder pressure, and amplitude of bladder contractions while 10−2–10−4 M reduced baseline tone as well as spontaneous contractions. Intraperitoneal administration of 5 mg/kg BRL 37344 or TAK-677 in awake rats reduced voiding frequency by as much as 40%–70%. The results therefore indicate that ovariectomy-induced bladder dysfunction (increased voiding and reduced bladder capacity) is not associated with β3-subtype expression pattern nor affects bladder inhibitory effects of the β3-subtype agonists and is confirmative of the potential of β3-subtype selective agonists to be developed for the treatment of hyperactive bladder disease in the elderly especially in postmenopausal populations.
So far, mirabegron has proved to be the most promising β3-adrenoceptor agonist over the past 30 years exhibiting good efficacy and tolerability and has currently successfully passed Phase III clinical trials in adults [117, 118]. Reports indicate that mirabegron has been authorized in Japan for the treatment of overactive bladder conditions [119]. Of interest also is TRK-380, another selective human β3-adrenoceptor agonist discovered 2 decades ago which induces isolated detrusor strip relaxation of several mammalian species [120]. TRK-380-induced relaxation of mammalian bladder strips is apparently solely mediated by β3-adrenoceptor and does have an effect on both resting and contractile responses. Additionally, TRK-380 has limited or no activity on β1 nor β2-subtypes compared with mirabegron and may have less cardiovascular effects without any major sight effects on urethral tone [68, 101]. BRL 37344 and CL 316243 which are species specific and therefore more efficient in rodents than in humans have been withdrawn [121, 122].
5.2. Targeting the β-Adrenoceptor in Endothelial Cell, Vascular Smooth Muscle Cell, and Cardiovascular Disease
Being essential regulators of vascular physiology, the β-adrenoceptors are involved in the pathology of such conditions as hypertension, atherosclerosis, and heart failures and are therefore potentially druggable for therapeutics in this regard. Generally, cAMP-mediated β-subtype signaling in smooth muscle cells often leads to muscle relaxation (i.e., vasodilation, bronchodilation (airways), uterine muscle relaxation, gastrointestinal tract relaxation, and neurotransmitter release) [123, 124]. Tissue specificity of β-subtype distribution is a major determinant controlling mechanical functions of the corresponding smooth muscle. The β2-subtype, for example, is abundantly expressed in airway smooth muscle. Increase in intracellular muscle cell cAMP concentration mediated by β-adrenoceptor stimulation due to binding of the receptor by a specific agonist is the established key trigger that leads to smooth muscle relaxation. Increased cellular cAMP level leads to activation of PKA. Active PKA then phosphorylates IP3 receptor of sarcoplasmic reticulum causing Ca2+-influx which in turn activates and opens Ca2+-dependent K+ channels such as the MaxiK (large-voltage dependant Ca2+-activated K+) channel. PKA and CaMKII (Ca2+ /calmodulin-dependent protein kinase II) also downregulate myosin light-chain kinase (MLCK; a Ser/Thr kinase that phosphorylates smooth muscle myosin on Ser19 resulting in smooth muscle contraction) by phosphoylating it on Ser1760, thereby, decreasing contractility and promoting smooth muscle relaxation [122, 125, 126]. Smooth muscle myosin lacks intrinsic myosin ATPase and therefore relies on posttranslational modification; i.e., phosphorylation on its Ser19 by MLCK to display activity [127]. MLCK is a Ca2+/CaM-dependent kinase and therefore is most simply activated by high intracellular Ca2+ ion concentration [127]. β-Adrenoceptor–stimulated smooth muscle relaxation without changes in cAMP concentration has also been reported. In airway smooth muscle, Gαs can directly interact with and activate MaxiK channels via β2-subtype stimulation. Gastrointestinal tract smooth muscle relaxation via cAMP-independent relaxation mechanism resulting from β3-adrenoceptor activation apparently leads to opening of delayed rectified K+ channels rather than the MaxiK channels [122] and are therefore druggable.
Cardiovascular diseases constitute a global leading cause of mortality and morbidity with a high 5-year mortality rate of 42.3%. Recent global statistics report that about 26 million adults suffer from heart failure [128, 129]. The main GPCR subtypes predominantly expressed in mammalian hearts are β1- and β2-adrenergic receptors. Cardiomyocytes, cardiac fibroblasts, endothelial, and vascular smooth muscle cells are the most abundant types of cells in the mammalian heart. Cardiomyocytes make up 30% of all heart cells and are the principal components of the myocardium (i.e., the muscle layer of the heart) that provides contractile force [130]. In humans, β1- and β2-adrenoceptors are the principal regulators of cardiovascular function, respectively, constituting ∼80% and ∼20% of all β-adrenoceptors in cardiomyocytes under physiological conditions [62, 131]. The β2-subtype is mainly expressed in cardiac fibroblasts, endothelial, and vascular smooth muscle cells. The stoichiometry of β1- and β2-adrenoceptors is reported to change to ∼60:40 during myocardial infarction, and a marked increase in Gi/o protein is primarily suggestive of a selective downregulation of β1- and a significant upregulation of β2-adrenergic gene expression [132]. For this reason, β-adrenoceptors play vital roles in the pathophysiology of heart disease and are therefore essential drug targets [133, 134].
Besides the canonical β-adrenoceptor agonists (catecholamines), some hormones and growth factors (Figures 3 and 4) such as vasopressin [135], insulin [136], TNF-α [137], and prostaglandin E [138], may also modulate the receptor function. Stimulated β1-adrenoceptor mediates accumulation of cAMP via Gαs-AC interaction consequently leading to upregulation of PKA activity, which in turn phosphorylates intracellular signaling molecules (Ca2+ channels) responsible for intracellular Ca2+ regulation in cardiomyocytes including phospholamban (PLB), T- and L-type Ca2+ channel (T-/L-TCC), and ryanodine receptor (RyR) and other cation channels involved in depolarization and subsequent generation of action potentials (e.g., the delayed rectifier K+ channel IKs) as well as proteins involved in regulation of contractile machinery (e.g., cardiac troponin I and cardiac myosin-binding protein C) [139]. Binding of only cAMP activates cyclic nucleotide-gated Na+ channels causing Na+ influx leading to rapid depolarization. This creates action potentials at a faster rate than just opening of T- and L-TCC in order to produce an increase in inotropy (force of contraction), chronotropy (heart rate), dromotropy (rate of conduction), and lusitropy (relaxation of myocardium during diastole) essentially providing an increase in cardiac output [132, 140, 141].
Furthermore, binding of Ca2+ to RyR of cardiac sarcoplasmic reticulum allows release of more Ca2+ ions into the cytoplasm during an action potential resulting in an increased inotropism [72]. Overstimulation of the β1-adrenoceptor which engages only Gαs mediates cardiotoxic signals such as cardiomyocyte apoptosis, cardiac hypertrophy, and eventually heart failure [21]. During heart failure, excessive sympathetic stimulation through Gαs results in the activation of the cardiotoxic β1-adrenoceptor-CaMKII pathway in parallel with PKA. Both enzymes have many common Ca2+ ion homeostatic protein substrates and other substrates targeted at regulation of gene transcription. As such, β1-adrenoceptor blockade is one effective way of treating heart failure. In addition, β2-adrenoceptor–linked upregulation of GRK2 and Gαi is observed in heart failure. The β2-adrenoceptor couples dually to both Gαs and Gαi proteins. Gαi pathway on the contrary activates GRK2 phosphorylation of the receptor eventually leading to β-arrestin binding and subsequent receptor desensitization (Figure 4). Therefore, the Gαi pathway opposes the Gαs coupled β-adrenoceptor positive inotropism to reduce heart rate and myocardial contractility which is beneficial. This way the Gαi pathway exhibits antiapoptotic and cardioprotective properties [18, 142, 143].
β-Blockers constitute one of the most widely prescribed drugs for the treatment of cardiovascular disorders such as hypertension and myocardial infarction [25]. β-Blockers (e.g., carvedilol, bisoprolol, and metoprolol) have proven effective and have shown potential of reducing morbidity and mortality in myocardial infarction [144–146]. The primary function of β-blockers (as the name suggests) in the cardiovascular system is downregulation of β-receptor signaling to basal levels to prevent harmful overactivation of Gαs proteins in heart cells [131]. However, β-receptor oversuppression may cause constriction of blood vessels and bronchi which may be side effects in the use of β-blockers where there is a global inhibition of β2-adrenoceptors in other tissues [147]. The β-arrestin–pathway biased β-receptor agonist, carvedilol especially, preferentially stimulates β-arrestin–mediated pathways while exhibiting inverse agonism toward Gαs signaling [148]. Carvedilol-stimulated β-receptor triggers a wide range of signaling events such as microRNA processing, epidermal growth factor receptor (EGFR) transactivation, and extracellular regulated kinase (ERK) stimulation. β-Arrestin–mediated EGFR transactivation apparently has a cardioprotective effect as it regulates epithelial tissue development and homeostasis. This has been demonstrated in transgenic mice overexpressing a mutant β1-receptor that lacked GRK phosphorylation sites and so lacked ability to initiate β-arrestin–dependent β-receptor downregulation, in which case apoptosis and heart dilation were enhanced [134, 149]. Therefore, it seems that β-arrestin–dependent β-adrenergic receptor signaling is advantageous for the heart.
Recent reports suggest that the β3-adrenoceptor is cardioprotective as they are expressed in cardiac and pulmonary vascular tissues and are upregulated in cardiac diseases. Moreover, β3-adrenoceptors produce negative inotropic effects. Previous investigations had established that the canonical β-adrenoceptor pathway in cardiac function via Ca2+ influx is L-TCC phosphorylation mediated by PKA which is in turn activated by intracellular rise in cAMP. The effect of β3-adrenoceptor stimulation on cardiac L-TCC Ca2+ current which underlies the plateau phase of the action potential for contraction in left ventricular myocytes was investigated by Zhang et al. [150]. Myocytes obtained from isoproterenol-induced male Sprague Dawley rat heart failure were used to demonstrate that the β3-adrenoceptor agonist BRL 37344 causes a 21% dose-dependent decrease in L-TCC Ca2+ current under normal physiological conditions and even greater inhibition (31%) in a failing heart at the same dosage. While a similar inhibition pattern of L-TCC Ca2+ current was observed with nadolol (an antagonist of both β1- and β2-subtypes), this inhibitory action was abolished by L748337 (a highly selective antagonist of β3-adrenoceptor) but not nadolol [150]. This implies that stimulation of positive ionotropic response by BRL 37344 through β1- and β2-subtype activation of L-TCC is mediated mainly by the classical cAMP/PKA mechanistic pathway but was abolished following BRL 37344 superfusion due to β3-adrenoceptor stimulation and not by β1- or β2-subtype activation. When endothelial nitric oxide synthase (eNOS) was inhibited with L-NAME (L-NG-Nitro arginine methyl ester) in the presence of BRL 37344, the inhibitory effect on L-TCC was abrogated indicative of the involvement of the NO pathway in β3-adrenoceptor–mediated negative ionotropic response [150]. When myocytes were incubated with pertussis toxin, the inhibitory function of L-TCC was again abolished by BRL 37344 indicating that pertussis toxin might have inhibited ADP-ribosylation of Gi/oα protein subunit, thereby negatively regulating PKG and allowing L-TCC to function. Zhang et al. [150] then concluded that β3-adrenoceptor inhibits L-TCC function in both normal and heart failing myocytes, and that β3-adrenoceptor–mediated inhibition of myocyte L-TCC is aggravated during heart failure. They further concluded that the mechanism by which β3-adrenoceptor inhibits L-TCC function may involve the, pertussis sensitive G protein and partially, mediated by NOS-dependent pathway.
It is now known that the β3-adrenoceptor is positively coupled to the Gαs subunit (in the human atrium, Figure 5(a)) which activates AC upon stimulation and therefore results in the synthesis of cAMP. Intracellular rise in cAMP results in binding of cAMP to PKA and activates PKA which in turn phosphorylates L-TCC. L-TCC opens to allow Ca2+ influx upon phosphorylation by PKA. On the other hand, the β3-adrenoceptor is negatively coupled to eNOS apparently through Gi/oα subunit of the G protein (in the human ventricle). Gi/oα subunit interacts with and activates eNOS which synthesizes NO from Arg (Figures 5(b) and 6). NO in turn binds and activates soluble guanylyl cyclase (sGC) resulting in the synthesis of cGMP. PKG is activated upon binding cGMP and phosphorylates L-TCC to inhibit Ca2+ influx [151, 152]. This pathway is cardioprotective and is pharmacologically relevant as this inhibitory ability may prevent overactivity of myocytes in heart failure. As such, the β3-adrenoceptor is of high interest for new therapeutic approaches for the treatment of heart failure.
Figure 5.

Mechanism by which β3-adrenoceptor mediates regulation of L-TCC Function. (a) Stimulation of L-TCC via cAMP/PKA pathway. (b) Inhibition of L-TCC via eNOS/PKA pathway. β3-Adrenoceptor activation may upregulate intracellular Ca2+ depolarization through positive regulation of L-TCC function to allow Ca2+ entry into myocytes (a). This is achieved via activation of the canonical pathway leading to intracellular increase in cAMP, subsequent activation of PKA, and phosphorylation of L-TCC by activated PKA. However, the β3-adrenoceptor is negatively coupled to eNOS apparently through Gαi/o subunit of the G protein (b). Interaction of Gαi/o with eNOS leads to activation of eNOS and synthesis of NO. NO then binds and activates sGC resulting in the synthesis of cGMP. PKG is activated upon binding cGMP and phosphorylates L-TCC to inhibit Ca2+ influx.
Figure 6.

Mechanism of nitric oxide-mediated vasodilation and pathogenesis of endothelial dysfunction in vascular disease. In response to agonist/bradykinin/catecholamine stimulation of the β-adrenergic receptor which results in intracellular rise in cAMP and Ca2+ concentrations, cAMP and Ca2+/calmodulin bind and activate eNOS by disrupting its interaction with caveolin. Activated dimer of eNOS (coupled) then catalyzes the synthesis of NO by electron transfer through electron carriers to reduce O2. The critical cofactor, tetrahydrobiopterin (BH4), acts as the electron donor to reduce and activate O2 which enables oxidation of L-Arg to NO releasing citrulline as a by-product under physiological conditions. NO diffuses into the smooth muscle cells to activate cGS initiating a cascade of events leading to vascular smooth muscle relaxation. Uncoupling of eNOS results in the synthesis of O2− a harmful free radical rather than antiatherosclerotic NO which is helpful. Uncoupling of eNOS is attributable to the oxidative depletion of BH4 to dihydrobiopterin (BH2), depletion of L-Arg (substrate), or accumulation of its analog (asymmetrical dimethylarginine) as well as eNOS S‐glutathionylation. Under oxidative stress conditions, BH4 is oxidized by O2− and even more strongly by NOO− to BH2. NOO− is produced by scavenging of NO by O2−. Unfortunately, BH2 promotes functional dimeric eNOS uncoupling, thereby resulting in a vicious cycle of increasing harmful oxidants and oxidative stress. This phenomenon of excessive O2− and oxidative stress leading to high ratio of BH2:BH4 is a major risk factor of cardiovascular disease pathophysiology.
Ferreira et al. [153] demonstrated that β3-adrenoceptor agonists attenuate myocardial remodeling in rat models. Similarly, results published by Kamiya et al. [154] suggest that chronic infusion of a β3-adrenoceptor agonist, BRL 37344, attenuates cardiac fibrosis and ameliorates diastolic dysfunction independent of blood pressure in an angiotensin II (Ang II)–induced hypertensive C57BL/6 J mouse model. Previous evidence had suggested an upregulation of β3-adrenoceptor expression in a heart that is failing in both man and animal models [155, 156]. Therefore, Kamayi and colleagues assessed the chronic effect of BRL 37344 in Ang II–induced cardiomyopathy mice and found that left ventricular end-diastolic pressure and end-diastolic pressure volume were significantly higher in Ang II–treated mice than controls; however, this increase was prevented in Ang II + BRL 37344–treated mice. Though heart rate was not different among the 3 groups (i.e., controls, Ang II only, and Ang II + BRL 37344), systolic blood pressure was significantly elevated in Ang II and Ang II + BRL 37344–treated mice. These results indicate improvement in myocardial stiffness induced by BRL 37344 treatment. Additionally, despite the fact that the left ventricular hypertrophy was stimulated in Ang II–treated mice and that BRL 37344 failed to prevent this, BRL 37344 however inhibited synthesis of collagen I/III, cardiac fibrosis, and lung congestion unlike Ang II only treated mice. This signifies cardioprotective effects of BRL 37344 which is thought to be associated with downregulation of transforming growth factor-β1 (TGFβ1) expression and subsequent phosphorylated SMAD2/3 mediated signaling [154]. Thus, chronic infusion of β3-adrenoceptor agonists may have potential therapeutic benefits for the treatment of heart failure.
5.3. Targeting β-Adrenoceptor in Arterial Hypertension
Together with their vasodilatory effects, β3-adrenoceptor agonists are a promising target in pulmonary arterial hypertension. Hypertension (i.e., high blood pressure) is a physical condition in which the blood vessels have persistently raised pressure to limits that can result in cardiovascular complications. Pulmonary artery hypertension is caused by endothelial dysfunction which is characterized by not only the inability of endothelial cells to vasodilate appropriately but also endothelial inflammatory activation. Hypertension stimulates proliferation of the vessel wall and narrowing of the lumen of the vessel. Persistent hypertension may result in lesions of the vessel walls which lead to vascular remodeling and ultimately development of atherosclerosis. In persistent hypertension, aneurysms (bulges) may occur in weakened vessels anywhere within the circulation, most especially around the aorta, and may result in vessel rupture posing a life-threatening situation for the patient [157]. Endothelial dysfunction is therefore associated with the development of several cardiovascular disorders such as heart failure and diabetes mellitus type 2. Current drugs used for treatment of pulmonary artery hypertension target dysfunctional endothelin (ET-1), NO, or prostacyclin (PGI2) pathways; however, overall survival with the disease remains poor (Figure 6) [158]. They most exclusively act as pulmonary vasodilators with poor safety profiles and so are inefficient.
The sympathoadrenal system regulates arterial blood pressure by controlling the vasculature, kidney, and heart rate [159]. Increased catecholamine secretion is reported to contribute to the pathogenesis of hypertension [160] suggesting that β-adrenoceptors may play a key role in the pathogenesis and maintenance of hypertension. Current knowledge on the molecular and biochemical basis of pathogenesis indicates a decline of the vascular β2-adrenoceptor signaling which leads to an impairment in vessel vasorelaxation while maintaining normal vasocontraction. This phenomenon is age-related. The underlying dysfunction is a consequential reduction in β-adrenoceptor–mediated cAMP synthesis in response to agonist activation and as such is associated with conditions involving altered cAMP synthesis such as hypertension, arterial insufficiency, orthostatic hypotension, and atherosclerosis [161, 162]. To date, no change in any specific factor can explain the impairment in β-adrenoceptor–mediated signaling suggesting that it may be as a result of multiple factors. Apparently, this impairment is linked to upregulated overexpression and activity of GRK2—the enzyme responsible for phosphorylation and subsequent desensitization of β-adrenoceptors; however, this mechanism is not fully understood [163].
Catecholamines and β-adrenoceptors contribute to the synthesis of NO. NO is synthesized by eNOS (a Ca2+/calmodulin-dependent enzyme) using BH4 as a cofactor. NO initiates a cascade of events that lead to smooth muscle cell relaxation and blood vessel dilation (Figure 6). Once synthesized, NO diffuses through the vascular endothelium into vascular smooth muscle cells where it reacts with Fe bound to the active site of sGC. NO-Fe interaction activates sGC. Active sGC in turn synthesizes cyclic guanosine monophosphate (cGMP, a second messenger) from GTP. Protein kinases such as protein kinase G (PKG) are activated by binding cGMP. Active protein kinases phosphorylate Ca2+ channels resulting in their closure to prevent further Ca2+ ion entry into the cell. These channels also enhance Ca2+ transport back into intracellular stores (sarcoplasmic reticulum), resulting in a reduction in cytosolic Ca2+ concentration [164]. PKG phosphorylates Ser/Thr residues of myosin-light-chain phosphatase (MLCP) to activate MLCP whose major function is to dephosphorylate the regulatory light chain (RLC) of the motor protein myosin-II and so negatively regulates actomyosin-based contractility to relax vascular smooth muscle and reduce blood pressure (Figure 6). This notwithstanding, other pathways which are independent of Ca2+ but involve phosphorylation of Ser residues of eNOS by some protein kinases have also been identified. One of such pathways involves β2-adrenoceptor in which eNOS is activated by kinases such as PKA and protein kinase B (AKT/PKB) [1]. Other functions of NO include inhibition of platelet aggregation and regulation of gene transcription among others.
The major cause of endothelial dysfunction is decreased NO bioavailability. However, there is evidence to suggest that increased reactive oxygen species (ROS) bioavailability, increased consumption of NO by ROS, and oxidative stress also contribute to the pathogenesis of endothelial dysfunction which is a common hallmark of cardiovascular disease [1, 165, 166]. Diabetes, angiotensin II, congestive heart failure, dyslipidemia (hypercholesterolemia), organic nitrates, asymmetrical dimethylarginine, aging, and smoking increase vascular oxidative stress which triggers eNOS dysfunction by enhancing uncoupling of the active dimeric eNOS. As eNOS uncouples, it synthesizes predominantly superoxides (O2−) rather than NO, thereby increasing the amount of harmful free radicals which intensify cellular oxidative stress while reducing concentration of NO. The biochemical and molecular mechanisms underlying eNOS uncoupling have been reviewed in detail elsewhere [1, 167, 168]. Bioavailability of NO is dependent on the balance of factors leading to its synthesis and degradation. Reduced expression of eNOS or eNOS activity is the major cause of low intracellular NO. However, NO is also scavenged by O2− which leads to decrease in NO's half-life in the vasculature implying that increased oxidative stress is implicated in the pathogenesis of cardiovascular disease [169]. More importantly, O2− reacts with NO to produce peroxynitrite radical (ONOO−) which further aggravates oxidative stress situation and drives pathological changes. ONOO− also strongly inhibits PGI2 signaling (Figure 6).
Being major downstream regulators of intracellular cAMP synthesis, the β-adrenoceptors have been targeted for the management of hypertension, angina, postmyocardial infarction risk, congestive heart failure, tremor, arrhythmias, and chronic obstructive pulmonary disorders [170, 171]. So far, nebivolol has demonstrated potential as a novel target for treatment of pulmonary artery hypertension in animal models [172]. For instance, Perros et al. [172] demonstrated that nebivolol (a β2- and β3-adrenoceptor agonist and a β1-antagonist) downregulates overexpression of growth factors and proinflammatory mediators leading to reduced vascular remodeling and inflammation. Nebivolol also stimulates endothelium-derived NO release leading to pulmonary vasodilation which attenuates the hemodynamic severity of pulmonary artery hypertension, reduces right ventricular hypertrophy, and removes ROS during oxidative stress [173–175]. Since increased catecholamine secretion is reported to contribute to pathogenesis of hypertension and that β-adrenoceptor function is depressed in hypertension as aforementioned, the success of nebivolol might be in part due to the activation of β-arrestin signaling [176–178]. The molecular basis for mechanism of nebivolol action is that it antagonizes the Gαs-coupling of β2-subtype and stimulates GRK-mediated phosphorylation of the receptor, thereby promoting recruitment of β-arrestins and ERK1/2 phosphorylation leading to receptor desensitization (Figure 4) [179]. Vasodilation property of nebivolol is achieved by several mechanisms including NO release, Ca2+ influx blockade through L-TCC, and antioxidative effect among others [180]. The effect of nebivolol may be a resultant effect of β-adrenoceptor subtypes rather than a single specific subtype as metoprolol (a β1-adrenoceptor selective blocker) does not yield similar outcomes. Nevertheless, these features of β-blockers especially of the third generation suggest therapeutic potential with better safety profiles than conventional blockers in the treatment of heart failure and pulmonary artery hypertension.
5.4. Targeting the β-Adrenergic Receptor in Obesity and Related Metabolic Disorders
Obesity is defined as BMI > 30 kg/m2. Obesity increases the risk of developing related metabolic disorders such as type 2 diabetes mellitus, atherosclerosis, hypertension, coronary heart disease, and cancer. Unfortunately, obesity has become a pandemic affecting about a third of the world's population [181]. β-Adrenoceptors particularly the β3-subtype have been targeted for the treatment of obesity and type 2 diabetes mellitus [182]. Earlier investigations showed that β-adrenoceptor blockade counter regulates decreased plasma glucose concentration after insulin-induced hypoglycemia but does not interfere with other hormonal (e.g., cortisol, glucagon, and growth hormone) responses to hypoglycemia. Scientists showed that propranolol exhibited ability to prevent epinephrine from initiating peripheral insulin resistance by raising plasma glucose levels in humans [183]. This is because epinephrine increases glycogenolysis, through a cascade of phosphorylations initiated by β-adrenoceptor binding, inactivates glycogenesis, and activates HSL and phosphorylase kinase [184, 185]. Since then, the choice of β-blockers for treating obesity and type 2 diabetes has remained controversial emanating from various meta-analyses because of 3 major reasons including development of insulin resistance, masking hypoglycemia which is more characteristic of nonselective β-blockers compared with selective ones, and development of dyslipidemia. β-Blockers seem to increase plasma triglyceride levels but reduce HDL cholesterol levels [186].
Later, the idea to potentially use β3-adrenoceptor agonists (e.g., GP-12177) for the treatment of obesity and type 2 diabetes mellitus stemmed from β3-subtype's regulatory function on lipolysis. Brown adipocytes contain multiple mitochondria and exclusively express uncoupling protein 1 (UCP1) which essentially ameliorate energy usage through thermogenesis while white adipocytes store excess energy as fat. Therefore, brown tissue activation with concomitant beiging (i.e., conversion of white adipose tissue to beige fat) increases energy expenditure and is a potentially beneficial strategy for treatment of obesity and related metabolic diseases [187, 188]. Beiging can be activated by cold climate, exercise, or pharmacological activation of the β3-adrenoceptor [189]. Mainly expressed on the surfaces of brown and white adipocytes in mammals, the β3-adrenoceptor is principally accountable for activation of brown adipocytes as well as the induction of beiging in white adipose tissue under cold temperature or adrenoceptor agonist stimulation. In addition, β3-adrenoceptor promotes release of insulin and glucose uptake by cells besides thermogenesis which ultimately lead to reduction in body weight [190]. Dysfunction of ADRB3 gene therefore could lead to insulin resistance and obesity [191]. β3-Adrenoceptor agonists such as mirabegron are potent activators of brown adipose tissue thermogenesis as well as white adipose tissue beiging in rodents [192, 193]. Mirabegron is already approved at 50 mg for treatment of overactive bladder disorders. However, to achieve significant thermogenic effect requires higher doses of the order of 200 mg which is accompanied by several adverse effects. This among others is attributable for inability to commercially develop β3-adrenoceptor agonists for treatment of obesity and metabolic syndromes at this stage [194, 195].
Several polymorphic variations of the ADRB3 gene exist that have been shown to be associated with many disease pathologies. The most common variant is the Trp64Arg mutation. Some investigations suggest that β3-adrenoceptor polymorphism (Trp64Arg mutation) is associated with insulin resistance and body weight gain [196, 197] which can lead to obesity and type 2 diabetes. Similar investigations also suggest that various metabolic phenotypes (including increased BMI) are associated with this Trp64Arg mutation in the ADRB3 gene [198–200]. However, even though some investigators have reported controversial outcomes [201–203], more recently, meta-analysis carried out by Wang et al. [204] is in consonance with earlier suggestions that Trp64Arg mutation in the ADRB3 gene might actually cause insulin resistance. Wang and colleagues [204] analyzed data from 8 papers using 1586 subjects who had been examined and found that Trp64Arg variant had a positive correlation with insulin level and therefore concluded that there might be an association between Trp64Arg and insulin resistance. They further stated that this correlation may be affected by the type of blood sample, obesity, and ethnicity and agreed that dietary ingredients among others may affect the degree of insulin sensitivity [205]. As β3-adrenoceptor signaling is essential for activation of brown adipocyte thermogenesis and lipolysis of white adipose tissue, β3-adrenoceptor dysfunction due to this mutation may account for various metabolic diseases including insulin resistance and obesity leaving affected individuals prone to developing type 2 diabetes [182, 183]. As such, β3-adrenoceptor agonists have potential to be developed for the treatment of insulin resistance and related metabolic disorders.
Additionally, Chen et al. [206] recently evaluated the effect of corylin (a flavonoid extract from Psoralea corylifolia L.) on browning and obesity. Chen and colleagues showed that corylin induces increased overexpression of beige- or browning-specific marker genes (cited1, hox9, pgc1α, prdm16, and UCP1) in 3 T3 L1 adipocytes, white adipose tissues, and brown adipose tissues which ultimately increase the browning process with subsequential increase in lipolysis and thermogenesis. Using HFD and DIO (diet-induced obesity) male C57BL/6 mice, they further demonstrated that corylin strikingly reduces body weight and fat accumulation and increases insulin resistance, mitochondrial biogenesis, and β-oxidation. As the effect of corylin was abrogated by treatment with EX527, (a sirtuin 1 (SIRT1) inhibitor) and L-748337 (a β3-adrenoceptor antagonist), the molecular mechanisms employed by corylin to enhance browning and lipolysis of adipocytes most likely involves SIRT1 and β3-adrenoceptor, suggesting that corylin may be therapeutically relevant for the treatment of obesity with both SIRTI and β3-adrenoceptor dependent pathways playing a vital role. Previous evidence indicated that SIRT1 an NAD+-dependent protein deacetylase attenuates obesity, promotes fat mobilization, augments mitochondrial metabolism, regulates glucose metabolism, and also inhibits inflammatory response [206–208].
In a slightly different approach, assessment of adipose tissue oxygen consumption and type 2 deiodinase protein expression levels in male C57BL/6J mice after treatment with liraglutide and the β3-adrenoceptor agonist, CL 316243, revealed remarkable loss of body weight in the mice suggesting potential for combined therapeutic development in the treatment of obesity and type 2 diabetes [209]. Liraglutide is a long-acting agonist of glucagon-like peptide 1 (GLP-1) receptor and shares 97% sequence homology with native GLP-1. Liraglutide has over the past decade been approved as a lifestyle therapy in the management of obesity [210]. Weight loss resulting from liraglutide (GLP-1A) signaling is largely attributable to loss of appetite and energy consumption. Scientists have demonstrated that GLP-1/1A receptors in adipocytes may induce brown adipose tissue thermogenesis and browning of white adipose tissue [211, 212]. Additionally, Oliveira et al. [209] established that liraglutide not only increases interscapular brown adipose tissue oxygen consumption but also further exhibits additive effects by enhancing β3-adrenoceptor–induced oxygen consumption in interscapular brown adipose tissue as well as inguinal white adipose tissue in mice with concomitant increase in UCP1 and type 2 deiodinase expression. This implies that liraglutide complements effects of β3-adrenoceptor–induced thermogenesis and increases type 2 deiodinase activity in brown adipose tissue suggesting that activation of brown adipose tissue depot may result from intracellular thyroid hormone activation via deiodinase activity which upregulates adrenergic signaling in adipocytes through the thyroid hormone receptor alpha (TRα) [209, 213]. Moreover, TRα-mediated signaling also in turn increases expression of thermogenesis-related genes [214].
The effect of green tea (Camellia sinensis) extract (GT) on β3-adrenoceptor–mediated regulation of white and beige adipose tissue lipolysis was investigated in [215]. Earlier studies had established that administration of GT potentially increases energy expenditure, lipid mobilization, and fat burning with concomitant reduction of body weight in obese animal models [216, 217]. Sousa-Filho et al. [215] used wild type (WT) and β3-adrenoceptor knockout (β3KO) male mice fed with standard diet (SD) or HFD, in 6 groups, treated with or without 0.5 g/kg body weight of GT for 12 weeks (i.e., (WT + SD and β3KO + SD), (WT + HFD and β3KO + HFD), and (WT + HFD + GT and β3KO + HFD + GT)). Histological analysis showed that GT attenuated final body weight and body weight gain, whereas adiposity index was increased by HFD. Reduction in weight of epididymal white adipose tissue in obese mice treated with GT is probably attributable to increase in lipolytic genes including Prkacb (PKA), Pnpla2 (Atgl), and Lipe (HSL) mRNA levels observed in western blots as well as SIRT, Ppargc1a, and UCP1 associated with thermogenesis only in the presence of the β3-adrenoceptor. Plasma insulin resistance was improved as treatment with GT reduced fasting glycemia in WT mice but did not appreciably modulate fasting glycemia in β3KO mice. More importantly, GT attenuated plasma leptin levels induced by HFD in both WT and β3KO mice with concomitant rise in adiponectin concentrations in β3KO but not in WT. Plasma levels of total triglycerides, total cholesterol, aspartate amino transferase (AST), and serum alanine aminotransferase (ALT) were raised by HFD treatment. Apparently, GT treatment reduced total triglyceride and cholesterol content in both WT and β3KO; however, these parameters remained higher in β3KO mice compared with WT. RT-qPCR results showed that GT upregulated FGF21 (fibroblast growth factor 21) and FGFr1 (fibroblast growth factor receptor 1) mRNA levels in brown adipose tissue of GT-treated β3KO mice when compared with WT, likely because of the requirement of FGF21 in order to induce thermogenesis and energy expenditure through activation of brown adipose tissue. Increased lipolysis with reduced adipocyte size and increased browning of subcutaneous white adipose tissue were therefore found to be β3-adrenoceptor–dependent. Treatment with GT also increased brown adipose tissue mRNA levels of lipolytic (oxidative) genes ADRB3/UCP1 as well as energy expenditure. Additionally, increase in subcutaneous white adipose tissue expression of SIRT1, Ppargc1a, and UCP1 mRNA levels observed is indicative of the induction of the browning process. Altogether, the data suggest that GT employs β3-adrenoceptor pathway activation to achieve some therapeutic effects [215].
The proposed molecular mechanism by which GT consumption may ameliorate body weight loss is by inhibition of the activity of catechol-O-methyltransferase (COMT) (Figure 7), one of the several enzymes that degrade catecholamines, catechol estrogens, and any substances containing a catechol structure through meta or para-O-methylation of OH− groups of the catechols. The potent inhibitors of COMT present in GT are catechins. Catechins are flavan-3-ols (epicatechin, epicatechin gallate, epigallocatechin, and epigallocatechin-3-gallate) which are plant metabolites that exhibit antioxidant properties in plants. Catechins belong to a group of polyphenols known as flavonoids. Inhibition of COMT by catechins results in increased concentration of catecholamines (norepinephrine and epinephrine) available for stimulation of β-adrenoceptors and may thus lead to prolonged stimulation of the receptors. The resulting downstream events lead to increase in energy expenditure and fat oxidation in adipocytes which strategically contain high β3-adrenoceptors essentially leading to loss of body weight (Figure 7). This mechanism has been demonstrated in vitro [218]; however, more in vivo evidence is required to be conclusive. Nevertheless, this is indicative of therapeutic potential of catechins of which β-adrenoceptor–mediated pathways (particularly of the β3-subtype which are abundant on adipocytes) are targeted to achieve therapeutic benefits [215]. The β3-adrenoceptor is again therapeutically significant in this regard.
Figure 7.

Hypothesized molecular mechanism by which green tea extract ameliorates weight loss. All β-adrenoceptors mediate catecholamine-stimulated lipolysis, beiging, and thermogenesis of adipocytes. Inhibition of COMT which degrades norepinephrine and epinephrine which are the natural ligands of β-adrenoceptors by catechins results in an increased concentration of ligands and therefore prolonged stimulation of the β-adrenoceptors especially of the β3-subtype. The resulting downstream cascade leads to upregulation of lipolytic genes Pnpla2 (Atgl) and Lipe (HSL), and thermogenesis-associated genes such as SIRT, Ppargc1a, and UCP1. The outcome is increased browning, energy expenditure, and fat oxidation of white adipocytes, thermogenesis in brown adipose tissues and loss in body weight. GT, therefore, employs majorly the β3-adrenoceptor pathway activation to achieve therapeutic effects.
5.5. Targeting the β-Adrenoceptor in Vision, Ocular Tumors, and Other Eye Diseases
β-Adrenergic receptors are expressed in ocular structures (cornea, conjunctiva, lacrimal gland, uvea, trabecular meshwork, and retina) where they are presumed to play various pathophysiological functions. β-Adrenoceptors have already been identified as potential targets for the treatment of glaucoma, ocular neoplasms (e.g., hemangioma and uveal melanoma), etc. Though the role of β2-subtype in corneal epithelium regeneration is controversial at this stage [219], researchers have demonstrated that treatment with β2-subtype antagonists and consequential upregulation of kinase activity of ERK enhance corneal cell migration and corneal wound healing and may therefore have therapeutic potential [220, 221].
All 3 β-subtypes are expressed in human conjunctiva [222] and are thought to be involved in the pathogenesis of some conjunctival diseases, e.g., conjunctival inflammation [140]. Liu et al. [223] observed an irregular expression pattern of the β1-adrenoceptor in all epithelial layers of conjunctival biopsy specimens of patients with vernal keratoconjunctivitis. This indicates that the receptor might play a role in pathogenesis and, if so, may have therapeutic potential. Additionally, salbutamol and terbutaline (β2-subtype agonists) seemingly attenuate microvascular permeability as well as exhibit anti-inflammatory effects in allergic conjunctivitis. As such, β-adrenoceptors may therefore have potential for the treatment of allergic conjunctivitis [140, 224, 225]. Studies also indicate that β-adrenoceptors are involved in the regulation of tear secretion in lacrimal gland and may play an important role in the pathophysiology of dry eye disease [226, 227]. As such, manipulation of β-adrenoceptor pathways may be potentially helpful in the treatment of dry eye disease [228].
Researchers have demonstrated abundant expression of β1- and β2-adrenoceptors in both ciliary body and epithelium of the ciliary process in humans and other mammalian species [229]. Agonists of β-adrenoceptors were earlier reported to induce desensitization of the β-adrenoceptor-AC complex indicating that this may have a delayed effect on intraocular pressure reduction following application of topical agonists of β-adrenoceptors. However, both agonists and antagonists lower intraocular pressure [230]. Uveal vasculature vasoconstriction coupled with reduction in aqueous humor production might be another mechanistic explanation of intraocular pressure decreasing property of β-blockers. As some non-subtype selective β-adrenoceptor antagonists (e.g., timolol) and some β2-type selective agonists to other β-receptors in ciliary processes possess potent characteristic lowering of intraocular pressure in various mammalian species, β2-adrenoceptors may be pharmacologically relevant for the treatment of glaucoma [231–233]. Timolol is also reported to increase choroidal vascular tone. This is indicative of the involvement of the sympathetic system in the maintenance of choroidal vasculature [234, 235]. Researchers have further shown that the β3-subtype plays a role in choroidal cell invasion and elongation using BRL 37344 (a β3-type specific agonist) [236]. Additionally, β2-type in turn plays a role in the regulation of VEGF (vascular endothelial growth factor) and interleukin 6 (IL-6) expression in choroidal endothelium. This indicates that blocking these β-adrenoceptors will synergistically attenuate choroidal neovascularization which may leak and cause vision loss [237].
Expression, cellular distribution, functions, and therapeutic implications of the β-adrenoceptors in the retina have been reviewed by several scientists [140, 238–240]. All 3 subtypes of β-adrenoceptors have been isolated in the retina of some mammalian species including man [241, 242]. Based on receptor localization in retinal blood vessels and neural retina, they are perceived to play vital roles in retinal vasculature and possess neuronal function [209, 239]. Catecholamine overstimulation of the cardiovascular system under stress conditions such as hypoxia has been reported in rat models [243, 244]. Catecholamines activate β-receptors which in turn induce overexpression of HIF-1α (hypoxia-inducible factor-1α) and VEGF which together activate proangiogenic cascades leading to the formation of pathogenic blood vessels (abnormal growth of blood vessels) in various retinal diseases including retinopathy of prematurity and diabetic retinopathy [239, 245]. More recently, scientists have demonstrated that treatment with propranolol a β-adrenoceptor blocker sufficiently inhibits hypoxia-induced overexpression of proangiogenic factors, VEGF and HIF-1α, and subsequent pathologic neovascularization in the retina of animal models [246]. This implies that β-adrenoceptor blockade may be protective against retinal angiogenesis [247]. Other studies have also shown that the β3-adrenoceptor antagonists, L-748337 and SR59230A, are both capable of downregulating hypoxia-induced VEGF expression and release in the retina [248]. In addition, the β3-adrenoceptor agonist, CL 316243, has been demonstrated to attenuate retinal damage after N-methyl-D-aspartate (NMDA; a mimic of glutamate) injection [249]. These properties of the β3-subtype are pharmacologically relevant for the treatment of ischemic retinal diseases [239]. Jiang et al. [250] reported a novel β-adrenoceptor agonist (compound 49b) which is capable of attenuating VEGF levels in type I diabetic rat model suggesting that it may be protective against edema. The possible mechanism by which compound 49b achieves reduction of VEGF levels may be by increasing levels of insulin-like growth factor binding protein 3 (IGFBP-3) in diabetic retina via downregulation of eNOS and PKC signaling [250]. As some agonists and antagonists of β-adrenoceptors cause reduction of VEGF and are therefore antiangiogenic, these effects are likely regulated by different mechanisms, and it is important to fully understand these differential mechanisms.
An infantile hemangioma is a benign vascular neoplasm resulting from an abnormal proliferation of endothelial cells and enhanced angiogenesis [251]. Over the years, corticosteroids, vincristine, cyclophosphamide, and interferon alpha have been used in the treatment of infantile hemangiomas. Unfortunately, these drugs have multiple side effects [252–254]. Earlier studies showed that VEGF promotes proliferation of vascular endothelial cell and angiogenesis by binding VEGF receptor-2 on hemangioma derived endothelial cells [255–257]. Later, scientists observed that propranolol and other β-blockers caused regression of infantile hemangiomas which implies that β-adrenoceptors may play an important role in the pathogenesis of the disease [251, 258–261]. Propranolol, an antagonist of β-adrenoceptors, has therefore been targeted to treat infantile hemangiomas by employing its vasoconstriction and antiangiogenic properties and its ability to induce apoptosis since it downregulates intracellular cAMP concentration and consequently prevents PKA activation. Propranolol causes inhibition of eNOS and therefore no NO is released ultimately resulting in vasoconstriction [262, 263]. This implies that β-adrenoceptor antagonists and β-blockers have potential to be developed for treatment of hemangiomas.
Uveal melanoma is the primary intraocular malignant tumor arising largely from neoplastic melanocyte proliferation in the uveal tract. It is a relatively rare cancer and occurs in adult populations causing morbidity (blindness) and increasing risk of mortality. Current treatment includes tumor resection, transpupillary thermo-therapy, local radiation, or ultimately enucleation (surgical removal of the eye); however, ∼50% of patients eventually develop metastasis primarily to the liver which has an impact on patients' prognosis, giving 6 months survival on average [264, 265]. Recently, β-adrenoceptors have emerged as novel targets for inhibition of melanoma growth and dissemination [266]. The contribution of β-adrenoceptors in cancer progression (see Section 5.6) including melanoma has been established [267]. β1- and β2-subtype overexpression in uveal melanoma of all patients has been documented [266]. In addition, the role of the β3-subtype in melanoma growth and vascularization has been demonstrated in mouse models [268, 269]. Catecholamine levels are increased in cancer patients which tend to activate β-adrenoceptor signaling resulting in activation of eNOS and therefore NO release which lead to vasodilation, cellular proliferation and survival, matrix metalloproteinase synthesis, and release of proangiogenic factors, IL-6, IL-8, and VEGF. These factors enhance melanoma development and progression [270]. β-Adrenoceptor antagonists such as propranolol are now known to exhibit antiproliferative properties with good safety profiles. Propranolol reverses the downstream cancer progression effects and stimulates cell apoptosis [271–273]. As such, β-blockers (e.g., propranolol, nebivolol, carvedilol, and labetalol) have therapeutic potential for the treatment of intraocular tumors. Some β-blockers have already gone through trials and have been approved by the Food and Drug Administration [274, 275]. According to Farhoumand et al. [276], nebivolol exclusively showed antitumor activity in uveal melanoma among all β-blockers in their investigation.
5.6. Targeting the β-Adrenoceptor in Cancer Therapy
Cancer is one of the leading causes of patient morbidity and mortality worldwide. Several cancer therapies have been developed, yet without the much needed efficiency, efficacy, and/or safety profiles. Currently, research has geared toward understanding the role of the sympathetic system in cancer progression via β-adrenoceptor activation by catecholamines [277]. This intriguing school of thought has been extensively researched and reviewed in detail by several scientists [278–281]. It had already been established in various in vitro and in vivo studies that the activation of β-adrenoceptors by catecholamines promotes the synthesis of cytokines especially IL-6 [282], cancer cell immunity [283], initiation of tumorigenesis [284], stimulation of tumor-associated macrophage [285], VEGF/FGF2-mediated angiogenesis [286], potentiation of the tumor micro- and macroenvironments [287, 288], and cancer cell proliferation, differentiation, and migration in animal models of various cancers [278, 289–291]. β-Adrenoceptor activation can stimulate activation of mitogen-activated protein kinase (MAPK) family of proteins via ERK. Both MAPK and ERK phosphorylate nuclear transcription factors to regulate expression of multiple genes involved in cell proliferation [292]. These characteristics of β-adrenoceptor activation tend to enhance cancer cell progressivity.
β-Adrenoceptor activation is reported to also regulate cellular metabolism which in turn promotes cancer cell progressivity, cell inflammation, cell apoptosis [293], cell communication and movement, DNA damage repair, etc. [294]. This implies that blocking β-adrenoceptor activation with β-blockers, in principle, will downregulate cancer progressivity. For these reasons, all 3 β-adrenoceptor subtypes are being targeted for treatment of various cancers [295]. β-Blockers (e.g., nebivolol, carvedilol, and propranolol) are inexpensive. They have good safety profiles, prevent tissue exposure to hazardous radiations employed in radiotherapy, and enhance sensitivity to chemotherapy agents such as response to anti-PD-1 and anti-CTLA4 [296–298]. They antagonize the effect of catecholamines yielding potentially beneficial outcomes. Indeed β-adrenoceptor antagonism has already been employed and has shown promising clinical effects in breast cancer. Propranolol is currently the gold standard in infantile hemangioma treatment [259]. Propranolol has also shown antitumor effects on colorectal and breast cancers, glioblastoma, ovarian carcinoma, and pancreatic cancers [299–301].
5.7. Targeting the β3-Adrenoceptor in Other Therapies
Research is still ongoing to unravel more novel therapeutic potentials of the β3-adrenoceptor. The list is not exhaustive. For example, nonalcoholic fatty liver disease (NAFLD) is a liver condition characterized by lipid accumulation in hepatocytes and hepatocyte steatosis. With rapid development and as economies improve, more and more people are currently consuming less vegetables and more meat. Fatty meat, animal visceral food, fried foods, and sweets are rich in saturated fatty acids and cholesterol. Sugars are normally stored as glycogen but can also be converted to fats for storage. Moreover, people are infatuated with technology rather than performing physical tasks which result in less exercise and high build-up of fat stores ultimately leading to an increase in the prevalence of NAFLD [136]. NAFLD is in turn a risk factor for other disease conditions including type 2 diabetes, dyslipidemia, hypertension, and cardiovascular diseases. Wang et al. [302] recently tested the hypothesis that β3-adrenoceptor expression is upregulated in NAFLD, and that BRL 37344 (a β3-adrenoceptor selective agonist) treatment is protective against liver steatosis and inflammation. Wang and colleagues employed a male Sprague Dawley rat model of a HFD-induced NAFLD. Setting up 4 groups of experiment rats, control (fed with SD), HFD (fed with HFD), HFD + BRL 37344, and HFD + L748337 (a β3-adrenoceptor antagonist), they showed that levels of ALT, AST, triglycerides, total cholesterol, low density lipoprotein cholesterol, and FFA levels relatively decreased among the group that had been treated with the β3-adrenoceptor agonist (BRL37344). Additionally, BRL 37344-treated rats had lower body and liver weights, liver index values, and lipid droplet accumulation compared to controls. Besides, reduction in NAFLD activity scores (NASs) also suggested liver steatosis and inflammation were ameliorated in BRL37344-treated rats. However, these parameters were reversed in L748337-treated rats compared to controls. In addition, while β3-adrenoceptor, PPAR-α (peroxisome proliferator-activated receptor alpha), and mCPT-1 (mouse cell protein 1) expressions were upregulated by treatment with BRL 37344, PPAR-γ (peroxisome proliferator-activated receptor gamma) and FAT/CD36 (fatty acid translocase) expressions were downregulated. Decrease in FAT/CD36 expression reduced entry of fatty acids into liver, whereas increase in mCPT-1 facilitated transport of hepatocellular fatty acids into mitochondria to be metabolized. Increase in PPAR-α (a transcription factor) expression decreased hepatocyte apolipoprotein C-III expression and increased lipoprotein lipase gene expression in order to enhance triglyceride metabolism. PPARγ is a transcription factor that downregulates immune cell function by increasing expression of anti-inflammatory related gene expression and downregulates expression of proinflammatory mediators which essentially reduce inflammation. Their results therefore indicate that upregulation of β3-adrenoceptor overexpression coupled with β3-adrenoceptor-Gs signal overactivity is protective against liver steatosis and inflammation and is suggestive of appreciable potential of BRL 37344 as a therapeutic target for the treatment of NAFLD [302].
Alzheimer's disease is a condition characterized by a decline in cognitive ability, gradual neurodegeneration, and the development of amyloid β (Aβ)-plaques and neurofibrillary tangles constituting hyperphosphorylated tau [303]. To date, there is no curative treatment for the disease. Alzheimer's disease is associated with defects in thermoregulation and cold-induced tau hyperphosphorylation in mammals and so β3-adrenoceptor activation may be beneficial in the treatment of Alzheimer's disease. Tournissac et al. [304] assessed this hypothesis and have reported experimental evidence in a triple transgenic mouse model that administration of a specific β3-adrenoceptor agonist (CL 316243) decreases body weight and improves peripheral glucose metabolism as well as brown adipose tissue thermogenesis in concordance with earlier studies [305]. More importantly, they stated that treated transgenic mice improved in recognition index by 19% while locomotion, anxiety, and tau pathology were unaffected. Besides, insoluble Aβ42/Aβ40 ratio also decreased by 27% in the hippocampus of CL 316243-treated mice. All together, these experimental outcomes suggest that CL 316243 might have significant potential for the synergistic treatment of Alzheimer's disease including some metabolic syndromes such as type 2 diabetes [304].
High level of cAMP is believed to be involved in the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD) which is a condition characterized by progressive expansion of fluid-filled cysts from various nephron segments that disrupt the parenchyma. It is now known that increased intracellular cAMP concentration in the cyst epithelial cells promotes cell proliferation as well as fluid secretion which results in cyst expansion [306]. To date, only vasopressin type 2 receptor blockade with tolvaptan remains the only FDA-approved remedy [307]. Vasopressin type 2 receptor activation results in elevated cellular cAMP levels. Another pathway leading to an increase in cAMP levels is the activated β3-adrenoceptor–induced cAMP synthesis by AC. β3-Adrenoceptor is expressed in human renal cyst-lining epithelial cells. Besides, β3-adrenoceptor expression is upregulated in ADPKD mice. As such, β3-adrenoceptor blockade may be a novel approach to treating ADPKD. Schena et al. [61] demonstrated that administration of SR59230A (a β3-adrenoceptor selective antagonist) to an ADPKD mouse model ameliorates cAMP levels with concomitant reduction in kidney/body weight ratio and a significant improvement in kidney function. SR59230A therefore has potential to be developed for ADPKD therapy.
More recent data unravel the potential of β3-adrenoceptor in the management of preterm birth which is the leading cause of infant morbidity and mortality. β3-Adrenoceptor is expressed in uterine myocytes and microvascular endothelial cells of women. Its expression is upregulated in myometrium of pregnant women suggesting a role of this receptor in pregnancy [278]. Asif and colleagues [278] observed that mirabegron (a selective β3-subtype agonist) in the tissue both relaxed oxytocin-induced contraction of the myometrium with an EC50 of 41.5 μM. Their data taken together give experimental evidence that underscores the potential of mirabegron or as part of a combination with others as a uterine tocolytic. β3-Adrenoceptor is thought to relax myometrium in a cGMP-independent manner. However, more research is required to fully understand the mechanisms underlying the β3-adrenoceptor stimulation of myometrium relaxation.
6. Conclusion and Perspectives
β-Adrenoceptor signaling cascades regulate crucial physiological processes involved in multiple pathogenesis and therefore remain potentially vital for the development of therapeutics. The strategies discussed for possible development of therapeutics here are but a few. More characterization reports unraveling novel modalities, new biochemical pathways, and physiological roles of β3-adrenoceptor are rapidly being revealed which make the β3-adrenoceptor an intriguing target for drug discovery. Most studies are still at the preliminary stages, for instance, the proposed use of β3-adrenoceptor in the treatment of Alzheimer's disease, ADPKD, and preterm birth. This notwithstanding, the role of β-adrenoceptors, generally, in some tissues remains ambiguous. Many questions remain unanswered and clinical trials versus sight effects have not been assessed as most hypotheses have only been tested at the level of disease animal models. As β-adrenoceptor activation or inhibition may couple multiple downstream signal molecules, can a particular disease be targeted without necessarily activating redundant signals? While some disease pathologies cannot be duplicated by animal models, some findings between scientists have been controversial and some clinical trials for the few at advanced stages have been disappointing. However, taken together, the development of β-adrenoceptor agonists and antagonists for novel therapeutic approaches is far more promising than ever as some have been approved for use by the FDA while others have reached advanced stages in clinical trials—Phase II and III. Meanwhile, research is rapidly revealing new pharmacological approaches. Developing new strategies requires understanding of the underlying molecular and biochemical basis of disease pathology. The β3-adrenoceptor in particular has suffered a setback owing to its late discovery, a few controversial, inconsistent, and/or contradictory findings, and the lack of selective detection tools coupled with interspecies differences. To bridge controversial findings, more insights into the structure of the β3-adrenoceptor, its function, regulation, expression patterns, roles in disease pathology, etc. will enhance the drug discovery process. To do so, more appropriate disease animal models and techniques are required in order to fully explore the potential of the β3-adrenoceptor as well as the β1- and β2-subtypes to fast-forward the developmental processes. Should the therapeutic potentials of the β-adrenoceptors be fully explored, the healthcare and lives of many patients especially those with chronic issues will be transformed.
Nomenclature
- β-ARK(s)
β-adrenergic kinase(s)
- AC
Adenylyl cyclase
- ADP
Adenosine diphosphate
- Anti-PD-1
Anti-programmed cell death protein 1
- ATF-2
Activating transcription factor 2
- ATP
Adenosine triphosphate
- CaMKII
Ca2+/calmodulin-dependent protein kinase II
- cAMP
Cyclic adenosine monophosphate
- CBP
CREB-binding Protein
- CEBPα
CCAAT enhancer binding protein alpha
- CGI-58
Comparative gene identification-58
- cGMP
Cyclic guanosine monophosphate
- COP-1
Constitutive photomorphogenesis protein 1
- CRE
cAMP response element
- CREB
CREB-binding element
- DAG
Diacyl glycerol
- EGFR
Epidermal growth factor receptor
- eNOS
Nitric oxide synthase
- EPAC
Exchange protein activated by cAMP
- ERK
Extracellular regulated kinase
- FAT/CD36
Fatty acid translocase/platelet glycoprotein 4
- FFA(s)
Free fatty acid(s)
- GC
Guanylyl cyclase
- GDP
Guanosine diphosphate
- GPCR(s)
G protein-coupled receptor(s)
- G protein
Guanine nucleotide-binding protein
- GRK2
G protein linked receptor kinase 2
- GRKs
G protein linked receptor kinases
- GTP
Guanosine triphosphate
- HFD
High-fat diet
- HSL
Hormone-sensitive lipase
- IL-6
Interleukin 6
- IL-8
Interleukin 8
- IP3
Inositol 1,4,5-triphosphate
- L-TCC
L-type calcium channel
- MAPK
Mitogen-activated protein kinase
- MaxiK
Large-conductance Ca2+-activated K+
- NMDA
N-methyl-D-aspartate
- NO
Nitric oxide
- PPAR-α
Peroxisome proliferator-activated receptor alpha
- PPAR-γ
Peroxisome proliferator-activated receptor gamma
- PIP2
Phosphatidylinositol 4,5-bisphosphate
- PI-PLC
Phosphoinositide-phospholipase C
- PKA
cAMP-dependent protein kinase A
- PKC
Protein kinase C
- PKG
Protein kinase G
- PLB
Phospholamban
- PLC
Phospholipase C
- PLIN
Perilipin
- RAP2A
Ras-related protein Rap-2a
- RyR
Ryanodine receptor
- SD
Standard diet
- SMAD
Suppressor of Mothers against Decapentaplegi
- SNS
Sympathetic nervous system
- TM(s)
Transmembrane(s)
- TNFR
Tumor necrosis factor alpha receptor
- TNF-α
Tumor necrosis factor alpha
- TRIB1
Tribbles pseudokinase 1
- T-TCC
T-type calcium channel
- Ub
Ubiquitin
- UCP1
Uncoupling protein 1
- VEGF
Vascular endothelial growth factor
Data Availability Statement
The data used to support the findings of this study are included within the article and are available upon request from the corresponding author.
Conflicts of Interest
The authors declare no conflicts of interest.
Author Contributions
J.T.D. conceived the idea and authored the introduction, gene and protein structure, and targeting the β-adrenoceptor in eye diseases as well as in other therapies. A.E.E. authored targeting the β-adrenoceptor in arterial hypertension and cancer therapy. H.K. authored the β-adrenoceptor signaling and desensitization while I.A. wrote on targeting the β-adrenoceptor in urinary and cardiovascular disease. S.B. authored the topic on obesity and related metabolic disorders. Diagrams were done by J.T.D. who also compiled and drafted the manuscript. C.A.W. contributed to reviewing the manuscript.
Funding
No funding was received for this study.
References
- 1.Ciccarelli M., Sorriento D., Coscioni E., Iaccarino G., Santulli G. Endocrinology of the Heart in Health and Disease: Integrated, Cellular, and Molecular Endocrinology of the Heart . Elsevier Inc; 2017. Adrenergic Receptors; pp. 285–315. [DOI] [Google Scholar]
- 2.Sriram K., Insel P. A. G Protein-Coupled Receptors as Targets for Approved Drugs: How Many Targets and How Many Drugs? Molecular Pharmacology . 2018;93(4):251–258. doi: 10.1124/mol.117.111062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ahles A., Engelhardt S. Polymorphic Variants of Adrenoceptors: Pharmacology, Physiology, and Role in Disease. Pharmacological Reviews . 2014;66(3):598–637. doi: 10.1124/pr.113.008219. [DOI] [PubMed] [Google Scholar]
- 4.Farzam K., Ariel Kidron A., Lakhkar A. D. Adrenergic DRUGS. 2023. https://www.ncbi.nlm.nih.gov/books/NBK534230/ [PubMed]
- 5.Giovannitti J. A., Thoms S. M., Crawford J. J. Alpha-2 Adrenergic Receptor Agonists: A Review of Current Clinical Applications. Anesthesia Progress . 2015;62(1):31–38. doi: 10.2344/0003-3006-62.1.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ahlquist R. P. A Study of the Adrenotropic Receptors. American Journal of Physiology-Legacy Content . 1948;153(3):586–600. doi: 10.1152/ajplegacy.1948.153.3.586. [DOI] [PubMed] [Google Scholar]
- 7.Perez D. M. The Adrenergic Receptors in the 21st Century . Totowa, New Jersey: Humana Press; 2006. [Google Scholar]
- 8.Bylund D. B. Sub Types of α2-adrenoceptors: Pharmacological and Molecular Biological Evidence Converg. Trends in Pharmacological Sciences . 1988;9(10):356–361. doi: 10.1016/0165-6147(88)90254-4. [DOI] [PubMed] [Google Scholar]
- 9.Bylund D. B., Eikenberg D. C., Hieble J. P., et al. International Union of Pharmacology Nomenclature of Adrenoceptors. Pharmacological Reviews . 1994;46(2):121–136. [PubMed] [Google Scholar]
- 10.Chhajlani V., Rangel N., Uhlén S., Wikberg J. E. Identification of an Additional Gene Belonging to the α2adrenergic Receptor Family in the Human Genome by PCR. FEBS Letters . 1991;280(2):241–244. doi: 10.1016/0014-5793(91)80301-i. [DOI] [PubMed] [Google Scholar]
- 11.Hoehe M. R., Otterud B., Hsieh W. T., et al. Genetic Mapping of Adrenergic Receptor Genes in Humans. Journal of Molecular Medicine . 1995;73(6):299–306. doi: 10.1007/BF00231616. [DOI] [PubMed] [Google Scholar]
- 12.Rockman H. A., Koch W. J., Lefkowitz R. J. Seven-Transmembrane-Spanning Receptors and Heart Function. Nature . 2002;415(6868):206–212. doi: 10.1038/415206a. [DOI] [PubMed] [Google Scholar]
- 13.Baker J. G., Balaji P., Bond R. A., et al. Adrenoceptors in GtoPdb v.2023.1. IUPHAR/BPS Guide to Pharmacology CITE . 2023;2023(1) doi: 10.2218/gtopdb/F4/2023.1. [DOI] [Google Scholar]
- 14.Brodde O. β-1 and β-2 Adrenoceptor Polymorphisms: Functional Importance, Impact on Cardiovascular Diseases and Drug Responses. Pharmacology & Therapeutics . 2008;117:1–29. doi: 10.1016/j.pharmthera.2007.07.002. [DOI] [PubMed] [Google Scholar]
- 15.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. Pharmacological Reviews . 2013;66(1):308–333. doi: 10.1124/pr.112.007203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Scanzano A., Cosentino M. Adrenergic Regulation of Innate Immunity: A Review. Frontiers in Pharmacology . 2015;6:p. 171. doi: 10.3389/fphar.2015.00171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Chhatar S., Lal G. Role of Adrenergic Receptor Signalling in Neuroimmune Communication. Current Research in Immunology . 2021;2:202–217. doi: 10.1016/j.crimmu.2021.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yoo B., Lemaire A., Mangmool S., et al. β1-Adrenergic Receptors Stimulate Cardiac Contractility and CaMKII Activation In Vivo and Enhance Cardiac Dysfunction Following Myocardial Infarction. American Journal of Physiology - Heart and Circulatory Physiology . 2009;297(4):1377–1386. doi: 10.1152/ajpheart.00504.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Snyder E. M., Johnson B. D., Joyner M. J. Genetics of β2-Adrenergic Receptors and the Cardiopulmonary Response to Exercise. Exercise and Sport Sciences Reviews . 2008;36(2):98–105. doi: 10.1097/JES.0b013e318168f276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Schena G., Caplan M. J. Everything You Always Wanted to Know About β3-AR (But Were Afraid to Ask) Cells . 2019;8(4):p. 357. doi: 10.3390/cells8040357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Velmurugan B. K., Baskaran R., Huang C. Y. Detailed Insight on β-Adrenoceptors as Therapeutic Targets. Biomedicine & Pharmacotherapy . 2019;117:p. 109039. doi: 10.1016/j.biopha.2019.109039. [DOI] [PubMed] [Google Scholar]
- 22.Coman O. A., Păunescu H., Ghiţă I., Coman L., Bădărăru A., Fulga I. Beta 3 Adrenergic Receptors: Molecular, Histological, Functional and Pharmacological Approaches. Romanian Journal of Morphology and Embryology . 2009;50(2):169–179. [PubMed] [Google Scholar]
- 23.Nagiri C., Kobayashi K., Tomita A., et al. Cryo-EM Structure of the β3-Adrenergic Receptor Reveals the Molecular Basis of Subtype Selectivity. Molecular Cell . 2021;81(15):3205–3215.e5. doi: 10.1016/j.molcel.2021.06.024. [DOI] [PubMed] [Google Scholar]
- 24.Skeberdis V. A. Structure and Function of Beta3-Adrenergic Receptors. Medicina . 2004;40(5):407–413. [PubMed] [Google Scholar]
- 25.Baker J. G. The Selectivity of β-Adrenoceptor Antagonists at the Human β1, β2 and β3 Adrenoceptors. British Journal of Pharmacology . 2005;144(3):317–322. doi: 10.1038/sj.bjp.0706048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Michel M. C., Gravas S. Safety and Tolerability of β3-Adrenoceptor Agonists in the Treatment of Overactive Bladder Syndrome—Insight From Transcriptosome and Experimental Studies. Expert Opinion on Drug Safety . 2016;15(5):647–657. doi: 10.1517/14740338.2016.1160055. [DOI] [PubMed] [Google Scholar]
- 27.Cernecka H., Sand C., Michel M. C. The Odd Sibling: Features of β3-Adrenoceptor Pharmacology. Molecular Pharmacology . 2014;86(5):479–484. doi: 10.1124/mol.114.092817. [DOI] [PubMed] [Google Scholar]
- 28.Okeke K., Angers S., Bouvier M., Michel M. C. Agonist-Induced Desensitisation of β3-Adrenoceptors: Where, When, and How? British Journal of Pharmacology . 2019;176(14):2539–2558. doi: 10.1111/bph.14633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wachter S. B., Gilbert E. M. Beta-Adrenergic Receptors, From Their Discovery and Characterization Through Their Manipulation to Beneficial Clinical Application. Cardiology . 2012;122(2):104–112. doi: 10.1159/000339271. [DOI] [PubMed] [Google Scholar]
- 30.Langley J. N. Preliminary Notice of Experiments on the Physiological Action of Jaborandi. British Medical Journal . 1875;1(738):241–242. doi: 10.1136/bmj.1.738.241-a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Maehle A. H. ‘Receptive Substances’: John Newport Langley (1852–1925) and His Path to a Receptor Theory of Drug Action. Medical History . 2004;48(2):153–174. doi: 10.1017/s0025727300000090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Limbird L. E. The Receptor Concept: A Continuing Evolution. Molecular Interventions . 2004;4(6):326–336. doi: 10.1124/mi.4.6.6. [DOI] [PubMed] [Google Scholar]
- 33.Abel J. J. Ueber Den Blutdruckerregenden Bestandtheil der Nebenniere, das Epinephrin. Hoppe-Seyler’s Zeitschrift für Physiologische Chemie . 1899;28(3-4):318–362. doi: 10.1515/bchm2.1899.28.3-4.318. [DOI] [Google Scholar]
- 34.Davenport H. W. Epinephrin(e) The Physiologist . 1982;25(2):76–82. [PubMed] [Google Scholar]
- 35.Lands A. M., Arnold A., McAuliff J. P., Luduena F. P., Brown T. G. Differentiation of Receptor Systems Activated by Sympathomimetic Amines. Nature . 1967;214(5088):597–598. doi: 10.1038/214597a0. [DOI] [PubMed] [Google Scholar]
- 36.Furchgott R. F. The Classification of Adrenoceptors (Adrenergic Receptors). An Evaluation from the Standpoint of Receptor Theory. In: Blaschko H., Muecholl F., editors. Catecholamines . New York: Springer Verlag; 1972. pp. 283–335. [Google Scholar]
- 37.Harms H. H., Zaagsma J., van der Wal B. β-Adrenoceptor Studies. III. On the β-Adrenoceptors in Rat Adipose Tissue. European Journal of Pharmacology . 1974;25:87–97. doi: 10.1016/0014-2999(74)90098-3. [DOI] [PubMed] [Google Scholar]
- 38.Bianchetti A., Manara L. In Vitro Inhibition of Intestinal Motility by Phenylethanolaminotetralines: Evidence of Atypical β‐Adrenoceptors in Rat Colon. British Journal of Pharmacology . 1990;100(4):831–839. doi: 10.1111/j.1476-5381.1990.tb14100.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Nergardh A., Boréus L. O., Naglo A. S. Characterization of the Adrenergic Beta-Receptor in the Urinary Bladder of Man and Cat. Acta Pharmacologica et Toxicologica . 1977;40(1):14–21. doi: 10.1111/j.1600-0773.1977.tb02049.x. [DOI] [PubMed] [Google Scholar]
- 40.Arch J. R. S. The Brown Adipocyte β-Adrenoceptor. Proceedings of the Nutrition Society . 1989;48(2):215–223. doi: 10.1079/pns19890032. [DOI] [PubMed] [Google Scholar]
- 41.Emorine L. J., Marullo S., Briend-Sutren M. M., et al. Molecular Characterization of the Human β3-Adrenergic Receptor. Science . 1989;245(4922):1118–1121. doi: 10.1126/science.2570461. [DOI] [PubMed] [Google Scholar]
- 42.Banfi S., Gusarova V., Gromada J., Cohen J. C., Hobbs H. H. Increased Thermogenesis by a Noncanonical Pathway in Angptl3/8-Deficient Mice. Proceedings of the National Academy of Sciences . 2018;115(6):E1249–E1258. doi: 10.1073/pnas.1717420115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Decara J., Rivera P., Arrabal S., et al. Cooperative Role of the Glucagon‐Like Peptide‐1 Receptor and β3‐Adrenergic‐Mediated Signalling on Fat Mass Reduction Through the Downregulation of PKA/AKT/AMPK Signalling in the Adipose Tissue and Muscle of Rats. Acta Physiologica . 2018;222(4):p. e13008. doi: 10.1111/apha.13008. [DOI] [PubMed] [Google Scholar]
- 44.Procino G., Carmosino M., Milano S., et al. β3 Adrenergic Receptor in the Kidney May be a New Player in Sympathetic Regulation of Renal Function. Kidney International . 2016;90(3):555–567. doi: 10.1016/j.kint.2016.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhang X., Hartung J. E., Bortsov A. V., et al. Sustained Stimulation of β2- and β3-Adrenergic Receptors Leads to Persistent Functional Pain and Neuroinflammation. Brain, Behavior, and Immunity . 2018;73:520–532. doi: 10.1016/j.bbi.2018.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gauthier C., Tavernier G., Charpentier F., Langin D., Le Marec H. Functional Beta3-Adrenoceptor in the Human Heart. Journal of Clinical Investigation . 1996;98(2):556–562. doi: 10.1172/JCI118823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhu W., Woo A. Y., Zhang Y., Cao C. M., Xiao R. P. β-Adrenergic Receptor Subtype Signaling in the Heart: From Bench to the Bedside. Current Topics in Membranes . 2011;67:191–204. doi: 10.1016/B978-0-12-384921-2.00009-4. [DOI] [PubMed] [Google Scholar]
- 48.Granneman J. G., Lahners K. N., Chaudhry A. Molecular Cloning and Expression of the Rat Beta 3-Adrenergic Receptor. Molecular Pharmacology . 1991;40(6):895–899. [PubMed] [Google Scholar]
- 49.Nahmias C., Blin N., Elalouf J. M., Mattei M. G., Strosberg A. D., Emorine L. J. Molecular Characterization of the Mouse Beta 3-Adrenergic Receptor: Relationship With the Atypical Receptor of Adipocytes. The EMBO Journal . 1991;10(12):3721–3727. doi: 10.1002/j.1460-2075.1991.tb04940.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Forrest R. H., Hickford J. G. Rapid Communication: Nucleotide Sequences of the Bovine, Caprine, and Ovine Beta3-Adrenergic Receptor Genes. Journal of Animal Science . 2000;78(5):1397–1398. doi: 10.2527/2000.7851397x. [DOI] [PubMed] [Google Scholar]
- 51.Sasaki N., Uchida E., Niiyama M., Yoshida T., Saito M. Anti-Obesity Effects of Selective Agonists to the BETA.3-Adrenergic Receptor in Dogs. I. The Presence of Canine BETA.3-Adrenergic Receptor and In Vivo Lipomobilization by its Agonists. Journal of Veterinary Medical Science . 1998;60(4):459–463. doi: 10.1292/jvms.60.459. [DOI] [PubMed] [Google Scholar]
- 52.van Spronsen A., Nahmias C., Krief S., Briend-Sutren M. M., Strosberg A. D., Emorine L. J. The Promoter and Intron/Exon Structure of the Human and Mouse β3‐Adrenergic‐Receptor Genes. European Journal of Biochemistry . 1993;213(3):1117–1124. doi: 10.1111/j.1432-1033.1993.tb17861.x. [DOI] [PubMed] [Google Scholar]
- 53.Granneman J. G., Lahners K. N., Rao D. D. Rodent and Human Beta 3-Adrenergic Receptor Genes Contain an Intron Within the Protein-Coding Block. Molecular Pharmacology . 1992;42(6):964–970. [PubMed] [Google Scholar]
- 54.Boldridge M., Shimabukuro J., Nakamatsu K., et al. Characterization of the C-Terminal Tail of the Arc Protein. PLoS One . 2020;15(9):p. e0239870. doi: 10.1371/journal.pone.0239870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Strosberg A. D. Structure and Function of the β3-Adrenergic Receptor. Annual Review of Pharmacology and Toxicology . 1997;37(1):421–450. doi: 10.1146/annurev.pharmtox.37.1.421. [DOI] [PubMed] [Google Scholar]
- 56.Strosberg A. D., Pietri-Rouxel F. Function and Regulation of the β3-Adrenoceptor. Trends in Pharmacological Sciences . 1996;17(10):373–381. doi: 10.1016/s0165-6147(96)80011-3. [DOI] [PubMed] [Google Scholar]
- 57.Adachi N., Hess D. T., Kaku M., Ueda C., Numa C., Saito N. Differential S-Palmitoylation of the Human and Rodent β3-Adrenergic Receptors. Journal of Biological Chemistry . 2019;294(7):2569–2578. doi: 10.1074/jbc.ra118.004978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.O’Dowd B. F., Hnatowich M., Caron M. G., Lefkowitz R. J., Bouvier M. Palmitoylation of the Human β2-Adrenergic Receptor. Journal of Biological Chemistry . 1989;264(13):7564–7569. doi: 10.1016/s0021-9258(18)83271-9. [DOI] [PubMed] [Google Scholar]
- 59.Cannavo A., Koch W. J. Targeting β3-Adrenergic Receptors in the Heart: Selective Agonism and β-Blockade. Journal of Cardiovascular Pharmacology . 2017;69(2):71–78. doi: 10.1097/FJC.0000000000000444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hoffmann C., Leitz M. R., Oberdorf-Maass S., Lohse M. J., Klotz K. N. Comparative Pharmacology of Human Beta-Adrenergic Receptor Subtypes--Characterization of Stably Transfected Receptors in CHO Cells. Naunyn-Schmiedeberg’s Archives of Pharmacology . 2004;369(2):151–159. doi: 10.1007/s00210-003-0860-y. [DOI] [PubMed] [Google Scholar]
- 61.Schena G., Carmosino M., Chiurlia S., et al. β3 Adrenergic Receptor as Potential Therapeutic Target in ADPKD. Physics Reports . 2021;9(20):p. e15058. doi: 10.14814/phy2.15058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Woo A. Y., Xiao R. P. β-Adrenergic Receptor Subtype Signaling in Heart: From Bench to Bedside. Acta Pharmacologica Sinica . 2012;33(3):335–341. doi: 10.1038/aps.2011.201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Jockers R., Da Silva A., Strosberg A. D., Bouvier M., Marullo S. New Molecular and Structural Determinants Involved in β2-Adrenergic Receptor Desensitization and Sequestration. Journal of Biological Chemistry . 1996;271(16):9355–9362. doi: 10.1074/jbc.271.16.9355. [DOI] [PubMed] [Google Scholar]
- 64.Nantel F., Bonin H., Emorine L. J., et al. The Human Beta 3-Adrenergic Receptor is Resistant to Short Term Agonist-Promoted Desensitization. Molecular Pharmacology . 1993;43(4):548–555. [PubMed] [Google Scholar]
- 65.Ursino M. G., Vasina V., Raschi E., Crema F., De Ponti F. The β3-Adrenoceptor as a Therapeutic Target: Current Perspectives. Pharmacological Research . 2009;59(4):221–234. doi: 10.1016/j.phrs.2009.01.002. [DOI] [PubMed] [Google Scholar]
- 66.Liggett S. B., Freedman N. J., Schwinn D. A., Lefkowitz R. J. Structural Basis for Receptor Subtype-Specific Regulation Revealed by a Chimeric Beta 3/Beta 2-Adrenergic Receptor. Proceedings of the National Academy of Sciences of the United States of America . 1993;90(8):3665–3669. doi: 10.1073/pnas.90.8.3665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Milano S., Gerbino A., Schena G., Carmosino M., Svelto M., Procino G. Human β3-Adrenoreceptor is Resistant to Agonist-Induced Desensitization in Renal Epithelial Cells. Cellular Physiology and Biochemistry . 2018;48(2):847–862. doi: 10.1159/000491916. [DOI] [PubMed] [Google Scholar]
- 68.Pasha A., Tondo A., Favre C., Calvani M. Inside the Biology of the β3-Adrenoceptor. Biomolecules . 2024;14(2):p. 159. doi: 10.3390/biom14020159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Walston J., Lowe A. L., Silver K., et al. The β3-Adrenergic Receptor in the Obesity and Diabetes Prone Rhesus Monkey is Very Similar to Human and Contains Arginine at Codon 64. Gene . 1997;188(2):207–213. doi: 10.1016/s0378-1119(96)00796-2. [DOI] [PubMed] [Google Scholar]
- 70.Lymperopoulos A., Rengo G., Koch W. J. Adrenergic Nervous System in Heart Failure: Pathophysiology and Therapy. Circulation Research . 2013;113(6):739–753. doi: 10.1161/CIRCRESAHA.113.300308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Vargas Pelaez A. F., Gao Z., Ahmad T. A., et al. Effect of Adrenergic Agonists on Coronary Blood Flow: A Laboratory Study in Healthy Volunteers. Physics Reports . 2016;4(10):p. e12806. doi: 10.14814/phy2.12806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Chen I. C., Kuo J., Ko W. J., Shih H. C., Kuo C. D. Increased Flow Resistance and Decreased Flow Rate in Patients With Acute Respiratory Distress Syndrome: The Role of Autonomic Nervous Modulation. Journal of the Chinese Medical Association . 2016;79(1):17–24. doi: 10.1016/j.jcma.2015.10.001. [DOI] [PubMed] [Google Scholar]
- 73.de Lucia C., Femminella G. D., Gambino G., et al. Adrenal Adrenoceptors in Heart Failure. Frontiers in Physiology . 2014;5:p. 246. doi: 10.3389/fphys.2014.00246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Karemaker J. M. An Introduction into Autonomic Nervous Function. Physiological Measurement . 2017;38(5):R89–R118. doi: 10.1088/1361-6579/aa6782. [DOI] [PubMed] [Google Scholar]
- 75.Haji E., Al Mahri S., Aloraij Y., Malik S. S., Mohammad S. Functional Characterization of the Obesity-Linked Variant of the β3-Adrenergic Receptor. International Journal of Molecular Sciences . 2021;22(11):p. 5721. doi: 10.3390/ijms22115721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kamato D., Thach L., Bernard R., et al. Structure, Function, Pharmacology, and Therapeutic Potential of the G Protein, Gα/q,11. Frontiers in Cardiovascular Medicine . 2015;2:p. 14. doi: 10.3389/fcvm.2015.00014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Syrovatkina V., Alegre K. O., Dey R., Huang X. Y. Regulation, Signaling, and Physiological Functions of G-Proteins. Journal of Molecular Biology . 2016;428(19):3850–3868. doi: 10.1016/j.jmb.2016.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Benovic J. L. Novel β2-Adrenergic Receptor Signaling Pathways. Journal of Allergy and Clinical Immunology . 2002;110(6):S229–S235. doi: 10.1067/mai.2002.129370. [DOI] [PubMed] [Google Scholar]
- 79.Grogan A., Lucero E. Y., Jiang H., Rockman H. A. Pathophysiology and Pharmacology of G Protein-Coupled Receptors in the Heart. Cardiovascular Research . 2023;119(5):1117–1129. doi: 10.1093/cvr/cvac171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hanania N. A., Mario Cazzola M. Chapter 27 - Bronchodilators: Beta2-Agonists and Anticholinergics. In: Castro M., Monica Kraft M., editors. Clinical Asthma . Mosby; 2008. pp. 241–250. [DOI] [Google Scholar]
- 81.Penn R. B., Panettieri R. A., Benovic J. L. Mechanisms of Acute Desensitization of the β2AR–Adenylyl Cyclase Pathway in Human Airway Smooth Muscle. American Journal of Respiratory Cell and Molecular Biology . 1998;19(2):338–348. doi: 10.1165/ajrcmb.19.2.3025. [DOI] [PubMed] [Google Scholar]
- 82.Reiter E., Lefkowitz R. J. GRKs and β-Arrestins: Roles in Receptor Silencing, Trafficking and Signaling. Trends in Endocrinology and Metabolism . 2006;17(4):159–165. doi: 10.1016/j.tem.2006.03.008. [DOI] [PubMed] [Google Scholar]
- 83.Lymperopoulos A., Rengo G., Zincarelli C., Kim J., Soltys S., Koch W. J. An Adrenal β-Arrestin 1-Mediated Signaling Pathway Underlies Angiotensin II-Induced Aldosterone Production In Vitro and In Vivo. Proceedings of the National Academy of Sciences of the United States of America . 2009;106(14):5825–5830. doi: 10.1073/pnas.0811706106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lymperopoulos A., Rengo G., Zincarelli C., Kim J., Koch W. J. Adrenal Beta-Arrestin 1 Inhibition In Vivo Attenuates Post-Myocardial Infarction Progression to Heart Failure and Adverse Remodeling via Reduction of Circulating Aldosterone Levels. Journal of the American College of Cardiology . 2011;57(3):356–365. doi: 10.1016/j.jacc.2010.08.635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Valentine J. M., Ahmadian M., Keinan O., et al. β3-Adrenergic Receptor Downregulation Leads to Adipocyte Catecholamine Resistance in Obesity. The Journal of Clinical Investigation . 2022;132(2):p. e153357. doi: 10.1172/JCI153357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Nantel F., Marullo S., Krief S., Strosberg A. D., Bouvier M. Cell-Specific Down-Regulation of the Beta 3-Adrenergic Receptor. Journal of Biological Chemistry . 1994;269(18):13148–13155. doi: 10.1016/s0021-9258(17)36811-4. [DOI] [PubMed] [Google Scholar]
- 87.Hadri K. E., Courtalon A., Gauthereau X., Chambaut-Guérin A. M., Pairault J., Fève B. Differential Regulation by Tumor Necrosis Factor-α of β1-β2-and β3-Adrenoreceptor Gene Expression in 3T3-F442a Adipocytes. Journal of Biological Chemistry . 1997;272(39):24514–24521. doi: 10.1074/jbc.272.39.24514. [DOI] [PubMed] [Google Scholar]
- 88.Liu P., Huang S., Ling S., et al. Foxp1 Controls Brown/Beige Adipocyte Differentiation and Thermogenesis Through Regulating β3-AR Desensitization. Nature Communications . 2019;10(1):p. 5070. doi: 10.1038/s41467-019-12988-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Granneman J. G., Li P., Zhu Z., Lu Y. Metabolic and Cellular Plasticity in White Adipose Tissue I: Effects of β3-adrenergic Receptor Activation. American Journal of Physiology, Endocrinology and Metabolism . 2005;289(4):E608–E616. doi: 10.1152/ajpendo.00009.2005. [DOI] [PubMed] [Google Scholar]
- 90.Bengtsson T., Redegren K., Strosberg A. D., Nedergaard J., Cannon B. Down-Regulation of β3 Adrenoreceptor Gene Expression in Brown Fat Cells is Transient and Recovery is Dependent Upon a Short-Lived Protein Factor. Journal of Biological Chemistry . 1996;271(52):33366–33375. doi: 10.1074/jbc.271.52.33366. [DOI] [PubMed] [Google Scholar]
- 91.Davel A. P., Victorio J. A., Delbin M. A., Fukuda L. E., Rossoni L. V. Enhanced Endothelium-Dependent Relaxation of Rat Pulmonary Artery Following β-Adrenergic Overstimulation: Involvement of the NO/cGMP/VASP Pathway. Life Sciences . 2015;125:49–56. doi: 10.1016/j.lfs.2015.01.018. [DOI] [PubMed] [Google Scholar]
- 92.Hutchinson D. S., Evans B. A., Summers R. J. β3-Adrenoceptor Regulation and Relaxation Responses in Mouse Ileum. British Journal of Pharmacology . 2000;129(6):1251–1259. doi: 10.1038/sj.bjp.0703160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Michel‐Reher M. B., Michel M. C. Agonist-Induced Desensitization of Human β3-Adrenoceptors Expressed in Human Embryonic Kidney Cells. Naunyn-Schmiedeberg’s Archives of Pharmacology . 2013;386(10):843–851. doi: 10.1007/s00210-013-0891-y. [DOI] [PubMed] [Google Scholar]
- 94.Okeke K., Michel‐Reher M. B., Michel M. C. β3‐Adrenoceptors Desensitisation in HEK Cells: A Role for Biased Agonism? 2017.
- 95.Carmen G. Y., Víctor S. M. Signalling Mechanisms Regulating Lipolysis. Cellular Signalling . 2006;18(4):401–408. doi: 10.1016/j.cellsig.2005.08.009. [DOI] [PubMed] [Google Scholar]
- 96.Mowers J., Uhm M., Reilly S. M., et al. Inflammation Produces Catecholamine Resistance in Obesity via Activation of PDE3B by the Protein Kinases IKKε and TBK1. Elife . 2013;2(2):p. e01119. doi: 10.7554/eLife.01119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Cao W., Medvedev A. V., Daniel K. W., Collins S. Beta-Adrenergic Activation of P38 MAP Kinase in Adipocytes: cAMP Induction of the Uncoupling Protein 1 (UCP1) Gene Requires P38 MAP Kinase. Journal of Biological Chemistry . 2001;276(29):27077–27082. doi: 10.1074/jbc.M101049200. [DOI] [PubMed] [Google Scholar]
- 98.Robidoux J., Cao W., Quan H., et al. Selective Activation of Mitogen-Activated Protein (MAP) Kinase Kinase3 and P38α MAP Kinase is Essential for Cyclic AMP-Dependent UCP1 Expression in Adipocytes. Molecular and Cellular Biology . 2005;25(13):5466–5479. doi: 10.1128/MCB.25.13.5466-5479.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Bauer R. C., Sasaki M., Cohen D. M., et al. Tribbles-1 Regulates Hepatic Lipogenesis Through Posttranscriptional Regulation of C/EBPα. Journal of Clinical Investigation . 2015;125(10):3809–3818. doi: 10.1172/JCI77095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hankir M. K., Cowley M. A., Fenske W. K. A BAT-Centric Approach to the Treatment of Diabetes: Turn on the Brain. Cell Metabolism . 2016;24(1):31–40. doi: 10.1016/j.cmet.2016.05.003. [DOI] [PubMed] [Google Scholar]
- 101.Michel M. C., Vrydag W. α 1‐, α2‐ and β‐Adrenoceptors in the Urinary Bladder, Urethra and Prostate. British Journal of Pharmacology . 2006;147(2):S88–S119. doi: 10.1038/sj.bjp.0706619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Larsen J. J. α- and β-Adrenoceptors in the Detrusor Muscle and Bladder Base of the Pig and β-Adrenoceptors in the Detrusor Muscle of Man. British Journal of Pharmacology . 1979;65(2):215–222. doi: 10.1111/j.1476-5381.1979.tb07821.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Fujimura T., Tamura K., Tsutsumi T., et al. Expression and Possible Functional Role of the Beta 3-Adrenoceptor in Human and Rat Detrusor Muscle. The Journal of Urology . 1999;161(2):680–685. doi: 10.1016/s0022-5347(01)61994-3. [DOI] [PubMed] [Google Scholar]
- 104.Bannowsky A., Juenemann K. P. Innervation and Function of the Female Urinary Bladder and Urethra. EAU Update Series . 2003;1(3):120–127. doi: 10.1016/s1570-9124(03)00035-7. [DOI] [Google Scholar]
- 105.Michel M. C., Oelke M., Peters S. L. M. The Neuro–Urological Connection. European Urology Supplements . 2005;4(1):18–28. doi: 10.1016/j.eursup.2004.10.004. [DOI] [Google Scholar]
- 106.Igawa Y., Yamazaki Y., Takeda H., et al. Functional and Molecular Biological Evidence for a Possible β3-adrenoceptor in the Human Detrusor Muscle. British Journal of Pharmacology . 1999;126(3):819–825. doi: 10.1038/sj.bjp.0702358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Igawa Y., Yamazaki Y., Takeda H., et al. Relaxant Effects of Isoproterenol and Selective β3-Adrenoceptor Agonists on Normal, Low Compliant and Hyperreflexic Human Bladders. The Journal of Urology . 2001;165(1):240–244. doi: 10.1097/00005392-200101000-00071. [DOI] [PubMed] [Google Scholar]
- 108.Yamaguchi O. β3-Adrenoceptors in Human Detrusor Muscle. Urology . 2002;59(5):25–29. doi: 10.1016/s0090-4295(01)01635-1. [DOI] [PubMed] [Google Scholar]
- 109.Igawa Y., Michel M. C. Pharmacological Profile of β3-Adrenoceptor Agonists in Clinical Development for the Treatment of Overactive Bladder Syndrome. Naunyn-Schmiedeberg’s Archives of Pharmacology . 2013;386(3):177–183. doi: 10.1007/s00210-012-0824-1. [DOI] [PubMed] [Google Scholar]
- 110.Yoshimura N., Chancellor M. B. Neurophysiology of Lower Urinary Tract Function and Dysfunction. Reviews in Urology . 2003;5(8):S3–S10. [PMC free article] [PubMed] [Google Scholar]
- 111.Calmasini F. B., Candido T. Z., Alexandre E. C., et al. The Beta-3 Adrenoceptor Agonist, Mirabegron Relaxes Isolated Prostate From Human and Rabbit: New Therapeutic Indication? The Prostate . 2015;75(4):440–447. doi: 10.1002/pros.22930. [DOI] [PubMed] [Google Scholar]
- 112.Masunaga K., Chapple C. R., McKay N. G., Yoshida M., Sellers D. J. The β3-Adrenoceptor Mediates the Inhibitory Effects of β-Adrenoceptor Agonists via the Urothelium in Pig Bladder Dome. Neurourology and Urodynamics . 2010;29(7):1320–1325. doi: 10.1002/nau.20838. [DOI] [PubMed] [Google Scholar]
- 113.Vij M., Drake M. J. Clinical Use of the β3 Adrenoceptor Agonist Mirabegron in Patients With Overactive Bladder Syndrome. Therapeutic Advances in Urology . 2015;7(5):241–248. doi: 10.1177/1756287215591763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Leon L. A., Hoffman B. E., Gardner S. D., et al. Effects of the β3-Adrenergic Receptor Agonist Disodium 5-[(2r)-2-[[(2r)-2-(3-Chlorophenyl)-2-Hydroxyethyl]amino]propyl]-1,3-Benzodioxole-2,2-Dicarboxylate (CL-316243) on Bladder Micturition Reflex in Spontaneously Hypertensive Rats. Journal of Pharmacology and Experimental Therapeutics . 2008;326(1):178–185. doi: 10.1124/jpet.108.138651. [DOI] [PubMed] [Google Scholar]
- 115.Kullmann F. A., Limberg B. J., Artim D. E., et al. Effects of β3-Adrenergic Receptor Activation on Rat Urinary Bladder Hyperactivity Induced by Ovariectomy. Journal of Pharmacology and Experimental Therapeutics . 2009;330(3):704–717. doi: 10.1124/jpet.109.155010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Takasu T., Ukai M., Sato S, et al. Effect of (R)-2-(2-Aminothiazol-4-yl)-4′-{2-[(2-hydroxy-2-phenylethyl)amino]ethyl} Acetanilide (YM178), a Novel Selective β3-Adrenoceptor Agonist, on Bladder Function. Journal of Pharmacology and Experimental Therapeutics . 2007;321(2):642–647. doi: 10.1124/jpet.106.115840. [DOI] [PubMed] [Google Scholar]
- 117.Baka-Ostrowska M., Bolong D. T., Persu C., et al. Efficacy and Safety of Mirabegron in Children and Adolescents With Neurogenic Detrusor Overactivity: An Open-Label, Phase 3, Dose-Titration Study. Neurourology and Urodynamics . 2021;40(6):1490–1499. doi: 10.1002/nau.24657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Chapple C. R., Cardozo L., Nitti V. W., Siddiqui E., Michel M. C. Mirabegron in Overactive Bladder: A Review of Efficacy, Safety, and Tolerability. Neurourology and Urodynamics . 2014;33(1):17–30. doi: 10.1002/nau.22505. [DOI] [PubMed] [Google Scholar]
- 119.Tyagi P., Tyagi V., Chancellor M. Mirabegron: A Safety Review. Expert Opinion on Drug Safety . 2011;10(2):287–294. doi: 10.1517/14740338.2011.542146. [DOI] [PubMed] [Google Scholar]
- 120.Kanie S., Otsuka A., Yoshikawa S., et al. Pharmacological Effect of TRK-380, a Novel Selective Human B3-Adrenoceptor Agonist, on Mammalian Detrusor Strips. Urology . 2012;79:744.e1–744.e7. doi: 10.1016/j.urology.2011.08.080. [DOI] [PubMed] [Google Scholar]
- 121.Otsuka A., Shinbo H., Matsumoto R., Kurita Y., Ozono S. Expression and Functional Role of β-Adrenoceptors in the Human Urinary Bladder Urothelium. Naunyn-Schmiedeberg’s Archives of Pharmacology . 2008;377(4-6):473–481. doi: 10.1007/s00210-008-0274-y. [DOI] [PubMed] [Google Scholar]
- 122.Tanaka Y., Horinouchi T., Koike K. New Insights into β‐Adrenoceptors in Smooth Muscle: Distribution of Receptor Subtypes and Molecular Mechanisms Triggering Muscle Relaxation. Clinical and Experimental Pharmacology and Physiology . 2005;32(7):503–514. doi: 10.1111/j.1440-1681.2005.04222.x. [DOI] [PubMed] [Google Scholar]
- 123.Djurup R. Adrenoceptors: Molecular Nature and Role in Atopic Diseases. Allergy . 1981;36(5):289–307. doi: 10.1111/j.1398-9995.1981.tb01581.x. [DOI] [PubMed] [Google Scholar]
- 124.Gambardella J., Fiordelisi A., Avvisato R., et al. Adrenergic Receptors in Endothelial and Vascular Smooth Muscle Cells. Current Opinion in Physiology . 2023;36:p. 100721. doi: 10.1016/j.cophys.2023.100721. [DOI] [Google Scholar]
- 125.Brozovich F. V., Nicholson C. J., Degen C. V., Gao Y. Z., Aggarwal M., Morgan K. G. Mechanisms of Vascular Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle Disorders. Pharmacological Reviews . 2016;68(2):476–532. doi: 10.1124/pr.115.010652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Horman S., Morel N., Vertommen D., et al. AMP Activated Protein Kinase Phosphorylates and Desensitizes Smooth Muscle Myosin Light Chain Kinase. Journal of Biological Chemistry . 2008;283(27):18505–18512. doi: 10.1074/jbc.M802053200. [DOI] [PubMed] [Google Scholar]
- 127.Ito M., Hartshorne D. J. Phosphorylation of Myosin as a Regulatory Mechanism in Smooth Muscle. Progress in Clinical and Biological Research . 1990;327:57–72. [PubMed] [Google Scholar]
- 128.Mouat M. A., Coleman J. L. J., Smith N. J. GPCRs in Context: Sexual Dimorphism in the Cardiovascular System. British Journal of Pharmacology . 2018;175(21):4047–4059. doi: 10.1111/bph.14160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Virani S. S., Alonso A., Benjamin E. J., et al. American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. Heart Disease and Stroke Statistics-2020 Update: A Report From the American Heart Association. Circulation . 2020;3(9):e139–e596. doi: 10.1161/CIR.0000000000000757. [DOI] [PubMed] [Google Scholar]
- 130.Zhou P., Pu W. T. Recounting Cardiac Cellular Composition. Circulation Research . 2016;118(3):368–370. doi: 10.1161/CIRCRESAHA.116.308139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Wang J., Gareri C., Rockman H. A. G-Protein-Coupled Receptors in Heart Disease. Circulation Research . 2018;123(6):716–735. doi: 10.1161/CIRCRESAHA.118.311403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Salazar N. C., Chen J., Rockman H. A. Cardiac GPCRs: GPCR Signaling in Healthy and Failing Hearts. Biochimica et Biophysica Acta (BBA) - Biomembranes . 2007;1768(4):1006–1018. doi: 10.1016/j.bbamem.2007.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Bernstein D., Fajardo G., Zhao M. The Role of β-Adrenergic Receptors in Heart Failure: Differential Regulation of Cardiotoxicity and Cardioprotection. Progress in Pediatric Cardiology . 2011;31(1):35–38. doi: 10.1016/j.ppedcard.2010.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Ferguson S. S. G., Feldman R. D. β-Adrenoceptors as Molecular Targets in the Treatment of Hypertension. Canadian Journal of Cardiology . 2014;30(5):S3–S8. doi: 10.1016/j.cjca.2014.01.017. [DOI] [PubMed] [Google Scholar]
- 135.Tilley D. G., Zhu W., Myers V. D., et al. β-Adrenergic Receptor–Mediated Cardiac Contractility is Inhibited via Vasopressin Type 1A-receptor–Dependent Signaling. Circulation . 2014;130(20):1800–1811. doi: 10.1161/circulationaha.114.010434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Wang Q., Liu Y., Fu Q., et al. Inhibiting Insulin-Mediated β2-Adrenergic Receptor Activation Prevents Diabetes-Associated Cardiac Dysfunction. Circulation . 2017;135(1):73–88. doi: 10.1161/CIRCULATIONAHA.116.022281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Vasudevan N. T., Mohan M. L., Gupta M. K., et al. Gβγ-Independent Recruitment of G-Protein Coupled Receptor Kinase 2 Drives Tumor Necrosis Factor α-Induced Cardiac β-Adrenergic Receptor Dysfunction. Circulation . 2013;128(4):377–387. doi: 10.1161/CIRCULATIONAHA.113.003183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Liu S., Li Y., Kim S., et al. Phosphodiesterases Coordinate Camp Propagation Induced by Two Stimulatory G Protein-Coupled Receptors in Hearts. Proceedings of the National Academy of Sciences of the United States of America . 2012;109(17):6578–6583. doi: 10.1073/pnas.1117862109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Song Y., Woo A., Zhang Y., Xiao R. Cardiac β-Adrenoceptor Signaling: The New Insight on an Old Target in the Therapy of Cardiovascular Disease. International Journal of Drug Discovery and Pharmacology . 2022;1(1):p. 3. doi: 10.53941/ijddp.v1i1.177. [DOI] [Google Scholar]
- 140.Böhm E. W., Stoffelns B., Gericke A. β-Adrenoreceptors as Therapeutic Targets for Ocular Tumors and Other Eye Diseases—Historical Aspects and Nowadays Understanding. International Journal of Molecular Sciences . 2023;24(5):p. 4698. doi: 10.3390/ijms24054698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Zhu W. Z., Chakir K., Zhang S., et al. Heterodimerization of Beta1- and Beta2-Adrenergic Receptor Subtypes Optimizes Beta-Adrenergic Modulation of Cardiac Contractility. Circulation Research . 2005;97(3):244–451. doi: 10.1161/01.RES.0000176764.38934.86. [DOI] [PubMed] [Google Scholar]
- 142.Communal C., Singh K., Sawyer D. B., Colucci W. S. Opposing Effects of Beta(1)- and Beta(2)-Adrenergic Receptors on Cardiac Myocyte Apoptosis: Role of a Pertussis Toxin-Sensitive G Protein. Circulation . 1999;100(22):2210–2212. doi: 10.1161/01.cir.100.22.2210. [DOI] [PubMed] [Google Scholar]
- 143.Chesley A., Lundberg M. S., Asai T., et al. The β2 -Adrenergic Receptor Delivers an Antiapoptotic Signal to Cardiac Myocytes through GI -Dependent Coupling to Phosphatidylinositol 3′-Kinase. Circulation Research . 2000;87(12):1172–1179. doi: 10.1161/01.res.87.12.1172. [DOI] [PubMed] [Google Scholar]
- 144.Packer M., Bristow M. R., Cohn J. N., et al. The Effect of Carvedilol on Morbidity and Mortality in Patients With Chronic Heart Failure. New England Journal of Medicine . 1996;334(21):1349–1355. doi: 10.1056/NEJM199605233342101. [DOI] [PubMed] [Google Scholar]
- 145.Simon T., Mary-Krause M., Funck-Brentano C., Lechat P., Jaillon P. Bisoprolol Dose-Response Relationship in Patients With Congestive Heart Failure: A Subgroup Analysis in the Cardiac Insufficiency Bisoprolol Study (CIBIS II) European Heart Journal . 2003;24(6):552–559. doi: 10.1016/s0195-668x(02)00743-1. [DOI] [PubMed] [Google Scholar]
- 146.Kukin M. L., Kalman J., Charney R. H., et al. Prospective, Randomized Comparison of Effect of Long-Term Treatment With Metoprolol or Carvedilol on Symptoms, Exercise, Ejection Fraction, and Oxidative Stress in Heart Failure. Circulation . 1999;99(20):2645–2651. doi: 10.1161/01.cir.99.20.2645. [DOI] [PubMed] [Google Scholar]
- 147.Lewis R. V., Lofthouse C. Adverse Reactions With Beta-Adrenoceptor Blocking Drugs. An Update. Drug Safety . 1993;9(4):272–279. doi: 10.2165/00002018-199309040-00005. [DOI] [PubMed] [Google Scholar]
- 148.Sweatt S. K., Gower B. A., Chieh A. Y., Liu Y., Li L. HHS Public Access. Physiology and Behavior . 2016;176(1):139–148. doi: 10.1161/CIRCRESAHA.118.311403. [DOI] [Google Scholar]
- 149.Noma T., Lemaire A., Naga Prasad S. V., et al. β-Arrestin–Mediated β1-Adrenergic Receptor Transactivation of the EGFR Confers Cardioprotection. Journal of Clinical Investigation . 2007;117(9):2445–2458. doi: 10.1172/JCI31901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Zhang Z. S., Cheng H. J., Onishi K., Ohte N., Wannenburg T., Cheng C. P. Enhanced Inhibition of L-Type Ca2+ Current by β3-Adrenergic Stimulation in Failing Rat Heart. Journal of Pharmacology and Experimental Therapeutics . 2005;315(3):1203–1211. doi: 10.1124/jpet.105.089672. [DOI] [PubMed] [Google Scholar]
- 151.Rozec B., Gauthier C. β3-Adrenoceptors in the Cardiovascular System: Putative Roles in Human Pathologies. Pharmacology & Therapeutics . 2006;111(3):652–673. doi: 10.1016/j.pharmthera.2005.12.002. [DOI] [PubMed] [Google Scholar]
- 152.Skeberdis V. A., Gendvilienë V., Zablockaitë D., et al. Beta3-adrenergic Receptor Activation Increases Human Atrial Tissue Contractility and Stimulates the L-Type Ca2+ Current. Journal of Clinical Investigation . 2008;118(9):3219–3227. doi: 10.1172/JCI32519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ferreira P. M., Tielemans B., Wagenaar A., et al. Beta 3 Adrenoceptor: A Potential Therapeutic Target for Pulmonary Arterial Hypertension. Basic Research in Cardiology . 2020;56(64):p. 1490. doi: 10.1183/13993003.congress-2020.1490. [DOI] [Google Scholar]
- 154.Kamiya M., Asai K., Maejima Y., et al. β3-Adrenergic Receptor Agonist Prevents Diastolic Dysfunction in an Angiotensin II–Induced Cardiomyopathy Mouse Model. Journal of Pharmacology and Experimental Therapeutics . 2021;376(3):473–481. doi: 10.1124/jpet.120.000140. [DOI] [PubMed] [Google Scholar]
- 155.Moniotte S., Kobzik L., Feron O., Trochu J. N., Gauthier C., Balligand J. L. Upregulation of β3 -Adrenoceptors and Altered Contractile Response to Inotropic Amines in Human Failing Myocardium. Circulation . 2001;103(12):1649–1655. doi: 10.1161/01.cir.103.12.1649. [DOI] [PubMed] [Google Scholar]
- 156.Treskatsch S., Feldheiser A., Rosin A. T., et al. A Modified Approach to Induce Predictable Congestive Heart Failure by Volume Overload in Rats. PLoS One . 2014;9(1):p. e87531. doi: 10.1371/journal.pone.0087531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Fisker F. Y., Grimm D., Wehland M. Third-Generation Beta-Adrenoceptor Antagonists in the Treatment of Hypertension and Heart Failure. Basic and Clinical Pharmacology and Toxicology . 2015;117(1):5–14. doi: 10.1111/bcpt.12396. [DOI] [PubMed] [Google Scholar]
- 158.Burger C. D., Long P. K., Shah M. R., et al. Characterization of First-Time Hospitalizations in Patients With Newly Diagnosed Pulmonary Arterial Hypertension in the REVEAL Registry. Chest . 2014;146(5):1263–1273. doi: 10.1378/chest.14-0193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.DeLalio L. J., Sved A. F., Stocker S. D. Sympathetic Nervous System Contributions to Hypertension: Updates and Therapeutic Relevance. Canadian Journal of Cardiology . 2020;36(5):712–720. doi: 10.1016/j.cjca.2020.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Mathar I., Vennekens R., Meissner M., et al. Increased Catecholamine Secretion Contributes to Hypertension in TRPM4-Deficient Mice. Journal of Clinical Investigation . 2010;120(9):3267–3279. doi: 10.1172/JCI41348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Chapman J., Schutzer W. E., Watts V. J., Mader S. L. Impaired Cholera Toxin Relaxation With Age in Rat Aorta. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences . 1999;54(4):B154–B159. doi: 10.1093/gerona/54.4.b154. [DOI] [PubMed] [Google Scholar]
- 162.Schutzer W. E., Mader S. L. Biochemical and Molecular Aspects of Vascular Adrenergic Regulation of Blood Pressure in the Elderly. International Journal of Hypertension . 2012;2012:1–10. doi: 10.1155/2012/915057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Gros R., Benovic J. L., Tan C. M., Feldman R. D. G-Protein-Coupled Receptor Kinase Activity is Increased in Hypertension. Journal of Clinical Investigation . 1997;99(9):2087–2093. doi: 10.1172/jci119381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Banik R. K., Berger J. S. Peripheral Vasodilators. In: Kaye A., Kaye A., Urman R., editors. Essentials of Pharmacology for Anesthesia, Pain Medicine, and Critical Care . New York: Springer; 2015. [DOI] [Google Scholar]
- 165.Iaccarino G., Ciccarelli M., Sorriento D., et al. AKT Participates in Endothelial Dysfunction in Hypertension. Circulation . 2004;109(21):2587–2593. doi: 10.1161/01.cir.0000129768.35536.fa. [DOI] [PubMed] [Google Scholar]
- 166.Ozaki M., Kawashima S., Yamashita T., et al. Overexpression of Endothelial Nitric Oxide Synthase Accelerates Atherosclerotic Lesion Formation in apoE-Deficient Mice. Journal of Clinical Investigation . 2002;110(3):331–340. doi: 10.1172/JCI15215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Gielis J. F., Quirynen L., Briedé J. J., Roelant E., Cos P., Van Schil P. E. Y. Pathogenetic Role of Endothelial Nitric Oxide Synthase Uncoupling During Lung Ischaemia-Reperfusion Injury. European Journal of Cardio-Thoracic Surgery . 2017;52(2):256–263. doi: 10.1093/ejcts/ezx125. [DOI] [PubMed] [Google Scholar]
- 168.Janaszak-Jasiecka A., Płoska A., Wierońska J. M., Dobrucki L. W., Kalinowski L. Endothelial Dysfunction Due to eNOS Uncoupling: Molecular Mechanisms as Potential Therapeutic Targets. Cellular and Molecular Biology Letters . 2023;28(1):p. 21. doi: 10.1186/s11658-023-00423-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Cervantes Gracia K., Llanas-Cornejo D., Husi H. CVD and Oxidative Stress. Journal of Clinical Medicine . 2017;6(2):p. 22. doi: 10.3390/jcm6020022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Fleg J. L., Aronow W. S., Frishman W. H. Cardiovascular Drug Therapy in the Elderly: Benefits and Challenges. Nature Reviews Cardiology . 2011;8(1):13–28. doi: 10.1038/nrcardio.2010.162. [DOI] [PubMed] [Google Scholar]
- 171.Kobilka B. K. Structural Insights into Adrenergic Receptor Function and Pharmacology. Trends in Pharmacological Sciences . 2011;32(4):213–218. doi: 10.1016/j.tips.2011.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Perros F., Ranchoux B., Izikki M., et al. Nebivolol for Improving Endothelial Dysfunction, Pulmonary Vascular Remodeling, and Right Heart Function in Pulmonary Hypertension. Journal of the American College of Cardiology . 2015;65(7):668–680. doi: 10.1016/j.jacc.2014.11.050. [DOI] [PubMed] [Google Scholar]
- 173.Ignarro L. J. Different Pharmacological Properties of Two Enantiomers in a Unique β‐Blocker, Nebivolol. Cardiovascular Therapeutics . 2008;26(2):115–134. doi: 10.1111/j.1527-3466.2008.00044.x. [DOI] [PubMed] [Google Scholar]
- 174.Mason R. P., Kalinowski L., Jacob R. F., Jacoby A. M., Malinski T. Nebivolol Reduces Nitroxidative Stress and Restores Nitric Oxide Bioavailability in Endothelium of Black Americans. Circulation . 2005;112(24):3795–3801. doi: 10.1161/circulationaha.105.556233. [DOI] [PubMed] [Google Scholar]
- 175.Rubin L. J. The Beta-Adrenergic Receptor in Pulmonary Arterial Hypertension. Journal of the American College of Cardiology . 2015;65(7):681–683. doi: 10.1016/j.jacc.2014.11.049. [DOI] [PubMed] [Google Scholar]
- 176.Esler M., Lambert G., Jennings G. Increased Regional Sympathetic Nervous Activity in Human Hypertension: Causes and Consequences. Journal of Hypertension - Supplement::Official Journal of the International Society of Hypertension . 1990;8(7):S53–S57. [PubMed] [Google Scholar]
- 177.Feldman R. D., Gros R. Defective Vasodilatory Mechanisms in Hypertension: A G-Protein-Coupled Receptor Perspective. Current Opinion in Nephrology and Hypertension . 2006;15(2):135–140. doi: 10.1097/01.mnh.0000214772.96361.ef. [DOI] [PubMed] [Google Scholar]
- 178.Wisler J. W., DeWire S. M., Whalen E. J., et al. A Unique Mechanism of β-blocker Action: Carvedilol Stimulates β-Arrestin Signaling. Proceedings of the National Academy of Sciences of the USA . 2007;104(42):16657–16662. doi: 10.1073/pnas.0707936104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Kenakin T. Functional Selectivity Through Protean and Biased Agonism: Who Steers the Ship? Molecular Pharmacology . 2007;72(6):1393–1401. doi: 10.1124/mol.107.040352. [DOI] [PubMed] [Google Scholar]
- 180.Toda N. Vasodilating β-Adrenoceptor Blockers as Cardiovascular Therapeutics. Pharmacology & Therapeutics . 2003;100(3):215–234. doi: 10.1016/j.pharmthera.2003.09.001. [DOI] [PubMed] [Google Scholar]
- 181.Blüher M. Obesity: Global Epidemiology and Pathogenesis. Nature Reviews Endocrinology . 2019;15(5):288–298. doi: 10.1038/s41574-019-0176-8. [DOI] [PubMed] [Google Scholar]
- 182.Zhan S., Ho S. C. Meta‐Analysis of the Association of the Trp64Arg Polymorphism in the β3 Adrenergic Receptor With Insulin Resistance. Obesity Research . 2005;13(10):1709–1719. doi: 10.1038/oby.2005.209. [DOI] [PubMed] [Google Scholar]
- 183.Abramson E. A., Arky R. A. Role of Beta-Adrenergic Receptors in Counterregulation to Insulin-Induced Hypoglycemia. Diabetes . 1968;17(3):141–146. doi: 10.2337/diab.17.3.141. [DOI] [PubMed] [Google Scholar]
- 184.Dietz M. R., Chiasson J. L., Soderling T. R., Exton J. H. Epinephrine Regulation of Skeletal Muscle Glycogen Metabolism. Studies Utilizing the Perfused Rat Hindlimb Preparation. Journal of Biological Chemistry . 1980;255(6):2301–2307. doi: 10.1016/s0021-9258(19)85890-8. [DOI] [PubMed] [Google Scholar]
- 185.Watt M. J., Howlett K. F., Febbraio M. A., Spriet L. L., Hargreaves M. Adrenaline Increases Skeletal Muscle Glycogenolysis, Pyruvate Dehydrogenase Activation and Carbohydrate Oxidation During Moderate Exercise in Humans. The Journal of Physiology . 2001;534(1):269–278. doi: 10.1111/j.1469-7793.2001.t01-1-00269.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Kasiske B. L., Ma J. Z., Kalil R. S., Louis T. A. Effects of Antihypertensive Therapy on Serum Lipids. Annals of Internal Medicine . 1995;122(2):133–141. doi: 10.7326/0003-4819-122-2-199501150-00010. [DOI] [PubMed] [Google Scholar]
- 187.Bartelt A., Bruns O. T., Reimer R., et al. Brown Adipose Tissue Activity Controls Triglyceride Clearance. Nature Medicine . 2011;17(2):200–205. doi: 10.1038/nm.2297. [DOI] [PubMed] [Google Scholar]
- 188.Cannon B. J., Nedergaard J. Brown Adipose Tissue: Function and Physiological Significance. Physiological Reviews . 2004;84(1):277–359. doi: 10.1152/physrev.00015.2003. [DOI] [PubMed] [Google Scholar]
- 189.Wang W., Seale P. Control of Brown and Beige Fat Development. Nature Reviews Molecular Cell Biology . 2016;17(11):691–702. doi: 10.1038/nrm.2016.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Dwaib H. S., Michel M. C. Is the β3-Adrenoceptor a Valid Target for the Treatment of Obesity and/or Type 2 Diabetes? Biomolecules . 2023;13(12):p. 1714. doi: 10.3390/biom13121714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Revelli J. P., Preitner F., Samec S., et al. Targeted Gene Disruption Reveals a Leptin-Independent Role for the Mouse Beta3-Adrenoceptor in the Regulation of Body Composition. Journal of Clinical Investigation . 1997;100(5):1098–1106. doi: 10.1172/JCI119620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Baskin A. S., Linderman J. D., Brychta R. J., et al. Regulation of Human Adipose Tissue Activation, Gallbladder Size, and Bile Acid Metabolism by a β3-Adrenergic Receptor Agonist. Diabetes . 2018;67(10):2113–2125. doi: 10.2337/db18-0462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Cypess A. M., Weiner L. S., Roberts-Toler C., et al. Activation of Human Brown Adipose Tissue by a β3-Adrenergic Receptor Agonist. Cell Metabolism . 2015;21(1):33–38. doi: 10.1016/j.cmet.2014.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Blondin D. P., Nielsen S., Kuipers E. N., et al. Human Brown Adipocyte Thermogenesis is Driven by β2-AR Stimulation. Cell Metabolism . 2020;32(2):287–300. doi: 10.1016/j.cmet.2020.07.005. [DOI] [PubMed] [Google Scholar]
- 195.Wang C. H., Wei Y. H. Therapeutic Perspectives of Thermogenic Adipocytes in Obesity and Related Complications. International Journal of Molecular Sciences . 2021;22(13):p. 7177. doi: 10.3390/ijms22137177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Clément K., Vaisse C., Manning B. S., et al. Genetic Variation in the β3-Adrenergic Receptor and an Increased Capacity to Gain Weight in Patients With Morbid Obesity. New England Journal of Medicine . 1995;333(6):352–354. doi: 10.1056/NEJM199508103330605. [DOI] [PubMed] [Google Scholar]
- 197.Walston J., Silver K., Bogardus C., et al. Time of Onset of Non-Insulin-Dependent Diabetes Mellitus and Genetic Variation in the β3-Adrenergic–Receptor Gene. New England Journal of Medicine . 1995;333(6):343–347. doi: 10.1056/NEJM199508103330603. [DOI] [PubMed] [Google Scholar]
- 198.Allison D., Heo M., Faith M. S., Pietrobelli A. Meta-Analysis of the Association of the Trp64Arg Polymorphism in the β3 Adrenergic Receptor With Body Mass Index. International Journal of Obesity . 1998;22(6):559–566. doi: 10.1038/sj.ijo.0800625. [DOI] [PubMed] [Google Scholar]
- 199.Fujisawa T., Ikegami H., Kawaguchi Y., Ogihara T. Meta-Analysis of the Association of Trp64Arg Polymorphism of β3-Adrenergic Receptor Gene With Body Mass Index. Journal of Clinical Endocrinology and Metabolism . 1998;83(7):2441–2444. doi: 10.1210/jcem.83.7.4922. [DOI] [PubMed] [Google Scholar]
- 200.Kurokawa N., Nakai K., Kameo S., Liu Z. M., Satoh H. Association of BMI With the β3‐Adrenergic Receptor Gene Polymorphism in Japanese: Meta‐Analysis. Obesity Research . 2001;9(12):741–745. doi: 10.1038/oby.2001.102. [DOI] [PubMed] [Google Scholar]
- 201.de Luis D. A., Aller R., Izaola O., Gonzalez Sagrado M., Conde R. Relation of Trp64Arg Polymorphism of Beta 3-adrenergic Receptor Gene to Adipocytokines and Fat Distribution in Obese Patients. Annals of Nutrition & Metabolism . 2008;52(4):267–271. doi: 10.1159/000144047. [DOI] [PubMed] [Google Scholar]
- 202.De Luis Román D. A., Primo D., Izaola O., Aller R. Relation of Trp64Arg Polymorphism of Beta 3 Adrenoreceptor Gene With Metabolic Syndrome and Insulin Resistance in Obese Women. Nutricion Hospitalaria . 2017;34(2):383–388. doi: 10.20960/nh.384. [DOI] [PubMed] [Google Scholar]
- 203.Højlund K., Christiansen C., Bjørnsbo K. S., et al. Energy Expenditure, Body Composition and Insulin Response to Glucose in Male Twins Discordant for the Trp64Arg Polymorphism of the β3‐Adrenergic Receptor Gene. Diabetes, Obesity and Metabolism . 2006;8(3):322–330. doi: 10.1111/j.1463-1326.2005.00509.x. [DOI] [PubMed] [Google Scholar]
- 204.Wang H. D., Zhang C. S., Li M. W., et al. The Association of Trp64Arg Polymorphism in the Beta-Adrenergic Receptor With Insulin Resistance: Meta-Analysis. Frontiers in Endocrinology . 2021;12:p. 708139. doi: 10.3389/fendo.2021.708139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Adeva-Andany M. M., González-Lucán M., Fernandez-Fernandez C., CarneiroFreire N., Seco-Filgueira M., Pedre-Pineiro A. M. Effect of Diet Composition on Insulin Sensitivity in Humans. Clinical Nutrition ESPEN . 2019;33:29–38. doi: 10.1016/j.clnesp.2019.05.014. [DOI] [PubMed] [Google Scholar]
- 206.Chen C. C., Kuo C. H., Leu Y. L., Wang S. H. Corylin Reduces Obesity and Insulin Resistance and Promotes Adipose Tissue Browning Through SIRT-1 and β3-AR Activation. Pharmacological Research . 2021;164:p. 105291. doi: 10.1016/j.phrs.2020.105291. [DOI] [PubMed] [Google Scholar]
- 207.Pfluger P. T., Herranz D., Velasco-Miguel S., Serrano M., Tschöp M. H. Sirt1 Protects Against High-Fat Diet-Induced Metabolic Damage. Proceedings of the National Academy of Sciences of the United States of America . 2008;105(28):9793–9798. doi: 10.1073/pnas.0802917105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Qiang L., Wang L., Kon N., et al. Brown Remodeling of White Adipose Tissue by SirT1-Dependent Deacetylation of Pparγ. Cell . 2012;150(3):620–632. doi: 10.1016/j.cell.2012.06.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Oliveira F. C. B., Bauer E. J., Ribeiro C. M., et al. Liraglutide Activates Type 2 Deiodinase and Enhances β3-Adrenergic-Induced Thermogenesis in Mouse Adipose Tissue. Frontiers in Endocrinology . 2021;12:p. 803363. doi: 10.3389/fendo.2021.803363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Fda. Saxenda (Liraglutide 3.0 Mg) Prescribing Information . Springer Field, Maryland: Food and Drug Administration; 2014. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/206321Orig1s000lbl.pdf . [Google Scholar]
- 211.Beiroa D., Imbernon M., Gallego R., et al. GLP-1 Agonism Stimulates Brown Adipose Tissue Thermogenesis and Browning Through Hypothalamic AMPK. Diabetes . 2014;63(10):3346–3358. doi: 10.2337/db14-0302. [DOI] [PubMed] [Google Scholar]
- 212.Ejarque M., Guerrero-Pérez F., de la Morena N., et al. Role of Adipose Tissue GLP-1R Expression in Metabolic Improvement After Bariatric Surgery in Patients With Type 2 Diabetes. Scientific Reports . 2019;9(1):p. 6274. doi: 10.1038/s41598-019-42770-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Pelletier P., Gauthier K., Sideleva O., Samarut J., Silva J. E. Mice Lacking the Thyroid Hormone Receptor-α Gene Spend More Energy in Thermogenesis, Burn More Fat, and are Less Sensitive to High-Fat Diet-Induced Obesity. Endocrinology . 2008;149(12):6471–6486. doi: 10.1210/en.2008-0718. [DOI] [PubMed] [Google Scholar]
- 214.Ribeiro M. O., Carvalho S. D., Schultz J. J., et al. Thyroid Hormone--Sympathetic Interaction and Adaptive Thermogenesis are Thyroid Hormone Receptor Isoform--Specific. Journal of Clinical Investigation . 2001;108(1):97–105. doi: 10.1172/JCI12584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Sousa-Filho C. P. B., Faria H. O. F., Esposito J. C., Melo A., Ribeiro M. O., Otton R. Green Tea Improves the Metabolism of Peripheral Tissues in β3-Adrenergic Receptor-Knockout Mice. Pharmacological Research . 2020;159:p. 104956. doi: 10.1016/j.phrs.2020.104956. [DOI] [PubMed] [Google Scholar]
- 216.Otton R., Bolin A. P., Ferreira L. T., Marinovic M. P., Rocha A. L. S., Mori M. A. Polyphenol-Rich Green Tea Extract Improves Adipose Tissue Metabolism by Down-Regulating miR-335 Expression and Mitigating Insulin Resistance and Inflammation. The Journal of Nutritional Biochemistry . 2018;57:170–179. doi: 10.1016/j.jnutbio.2018.03.024. [DOI] [PubMed] [Google Scholar]
- 217.Rocha A., Bolin A. P., Cardoso C. A., Otton R. Green Tea Extract Activates AMPK and Ameliorates White Adipose Tissue Metabolic Dysfunction Induced by Obesity. European Journal of Nutrition . 2016;55(7):2231–2244. doi: 10.1007/s00394-015-1033-8. [DOI] [PubMed] [Google Scholar]
- 218.Dostal A. M., Samavat H., Espejo L., Arikawa A. Y., Stendell-Hollis N. R., Kurzer M. S. Green Tea Extract and Catechol-O-Methyltransferase Genotype Modify Fasting Serum Insulin and Plasma Adiponectin Concentrations in a Randomized Controlled Trial of Overweight and Obese Postmenopausal Women. The Journal of Nutrition . 2016;146(1):38–45. doi: 10.3945/jn.115.222414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Yuan X., Ma X., Yang L., Zhou Q., Li Y. β-Blocker Eye Drops Affect Ocular Surface Through β2 Adrenoceptor of Corneal Limbal Stem Cells. BMC Ophthalmology . 2021;21(1):p. 419. doi: 10.1186/s12886-021-02186-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Ghoghawala S. Y., Mannis M. J., Pullar C. E., Rosenblatt M. I., Isseroff R. R. β2-Adrenergic Receptor Signaling Mediates Corneal Epithelial Wound Repair. Investigative Opthalmology & Visual Science . 2008;49(5):1857–1863. doi: 10.1167/iovs.07-0925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Pullar C. E., Zhao M., Song B., et al. SS‐Adrenergic Receptor Agonists Delay While Antagonists Accelerate Epithelial Wound Healing: Evidence of an Endogenous Adrenergic Network Within the Corneal Epithelium. Journal of Cellular Physiology . 2007;211(1):261–272. doi: 10.1002/jcp.20934. [DOI] [PubMed] [Google Scholar]
- 222.Diebold Y., Ríos J. D., Hodges R. R., Rawe I., Dartt D. A. Presence of Nerves and Their Receptors in Mouse and Human Conjunctival Goblet Cells. Investigative Ophthalmology & Visual Science . 2001;42(10):2270–2282. [PubMed] [Google Scholar]
- 223.Liu J., Huang S., Li F., et al. Sympathetic Nerves Positively Regulate Eosinophil-Driven Allergic Conjunctivitis via α1-Adrenergic Receptor Signaling. American Journal Of Pathology . 2020;190(6):1298–1308. doi: 10.1016/j.ajpath.2020.02.004. [DOI] [PubMed] [Google Scholar]
- 224.Gautheron P., Sugrue M. F. The Ability of Salbutamol and Theophylline to Suppress Immediate Allergic Conjunctivitis in the guinea Pig. Graefes Archive for Clinical and Experimental Ophthalmology . 1987;225(5):331–334. doi: 10.1007/bf02153399. [DOI] [PubMed] [Google Scholar]
- 225.Woodward D. F., Nieves A. L. Topical Anti-Inflammatory Activity of β2-Adrenoceptor Agonists in the Conjunctiva. Journal of Ocular Pharmacology and Therapeutics . 1985;1(4):391–396. doi: 10.1089/jop.1985.1.391. [DOI] [PubMed] [Google Scholar]
- 226.Esmaeli-Gutstein B., Hewlett B. R., Harvey J. T. Characterization of Adrenergic Receptors in the Accessory Lacrimal Glands of the Upper Eyelid. Ophthalmic Plastic and Reconstructive Surgery . 1999;15(4):245–251. doi: 10.1097/00002341-199907000-00005. [DOI] [PubMed] [Google Scholar]
- 227.Jun I., Choi Y. J., Kim B. R., Seo K. Y., Kim T. I. Activation of ADRB2/PKA Signaling Pathway Facilitates Lipid Synthesis in Meibocytes, and Beta-Blocker Glaucoma Drug Impedes PKA-Induced Lipid Synthesis by Inhibiting ADRB2. International Journal of Molecular Sciences . 2022;23(16):p. 9478. doi: 10.3390/ijms23169478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.García-Posadas L., Hodges R. R., Utheim T. P., et al. Lacrimal Gland Myoepithelial Cells are Altered in a Mouse Model of Dry Eye Disease. American Journal of Pathology . 2020;190(10):2067–2079. doi: 10.1016/j.ajpath.2020.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Bromberg B. B., Gregory D. S., Sears M. L. Beta-Adrenergic Receptors in Ciliary Processes of the Rabbit. Investigative Ophthalmology & Visual Science . 1980;19(2):203–207. [PubMed] [Google Scholar]
- 230.Mittag T., Tormay A. Desensitization of the β-Adrenergic Receptor-Adenylate Cyclase Complex in Rabbit Iris-Ciliary Body Induced by Topical Epinephrine. Experimental Eye Research . 1981;33(5):497–503. doi: 10.1016/s0014-4835(81)80124-8. [DOI] [PubMed] [Google Scholar]
- 231.Nathanson J. A. ICI 118,551: An Effective Ocular Hypotensive Agent With Selectivity for the Ciliary Process β2-adrenoceptor and With Minimal Cardiac Side Effects. British Journal of Pharmacology . 1984;83(3):821–829. doi: 10.1111/j.1476-5381.1984.tb16238.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Potter D. E. Adrenergic Pharmacology of Aqueous Humor Dynamics. Pharmacological Reviews . 1981;33(3):133–153. [PubMed] [Google Scholar]
- 233.Trope G. E., Clark B. Binding Potencies of 3 New Beta 2 Specific Blockers to Beta Receptors in the Ciliary Processes and the Possible Relevance of These Drugs to Intraocular Pressure Control. British Journal of Ophthalmology . 1984;68(4):245–247. doi: 10.1136/bjo.68.4.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Grajewski A. L., Ferrari-Dileo G., Feuer W. J., Anderson D. R. Beta-adrenergic Responsiveness of Choroidal Vasculature. Ophthalmology . 1991;98(6):989–995. doi: 10.1016/s0161-6420(91)32216-4. [DOI] [PubMed] [Google Scholar]
- 235.Steinle J. J., Smith P. G. Role of Adrenergic Receptors in Vascular Remodelling of the Rat Choroid. British Journal of Pharmacology . 2002;136(5):730–734. doi: 10.1038/sj.bjp.0704771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Steinle J. J., Zamora D. O., Rosenbaum J. T., Granger H. J. β3-Adrenergic Receptors Mediate Choroidal Endothelial Cell Invasion, Proliferation, and Cell Elongation. Experimental Eye Research . 2005;80(1):83–91. doi: 10.1016/j.exer.2004.08.015. [DOI] [PubMed] [Google Scholar]
- 237.Lavine J. A., Farnoodian M., Wang S., et al. β2–Adrenergic Receptor Antagonism Attenuates CNV Through Inhibition of VEGF and IL-6 Expression. Investigative Opthalmology & Visual Science . 2017;58(1):299–308. doi: 10.1167/iovs.16-20204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Cammalleri M., Amato R., Dal Monte M., Filippi L., Bagnoli P. The β3 Adrenoceptor in Proliferative Retinopathies: “Cinderella” Steps Out of its Family Shadow. Pharmacological Research . 2023;190:p. 106713. doi: 10.1016/j.phrs.2023.106713. [DOI] [PubMed] [Google Scholar]
- 239.Ruan Y., Böhmer T., Jiang S., Gericke A. The Role of Adrenoceptors in the Retina. Cells . 2020;9(12):p. 2594. doi: 10.3390/cells9122594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Tsang J. K. W., Liu J., Lo A. C. Y. Vascular and Neuronal Protection in the Developing Retina: Potential Therapeutic Targets for Retinopathy of Prematurity. International Journal of Molecular Sciences . 2019;20(17):p. 4321. doi: 10.3390/ijms20174321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Granneman J. G. The Putative β4-Adrenergic Receptor is a Novel State of the β1-Adrenergic Receptor. American Journal of Physiology. Endocrinology and Metabolism . 2001;280(2):E199–E202. doi: 10.1152/ajpendo.2001.280.2.E199. [DOI] [PubMed] [Google Scholar]
- 242.Steinle J. J., Booz G. W., Meininger C. J., Day J. N., Granger H. J. β3-Adrenergic Receptors Regulate Retinal Endothelial Cell Migration and Proliferation. Journal of Biological Chemistry . 2003;278(23):20681–20686. doi: 10.1074/jbc.m300368200. [DOI] [PubMed] [Google Scholar]
- 243.Dal Monte M., Martini D., Latina V., Pavan B., Filippi L., Bagnoli P. Beta-Adrenoreceptor Agonism Influences Retinal Responses to Hypoxia in a Model of Retinopathy of Prematurity. Investigative Opthalmology & Visual Science . 2012;53(4):2181–2192. doi: 10.1167/iovs.11-9408. [DOI] [PubMed] [Google Scholar]
- 244.Lindgren I., Altimiras J. Chronic Prenatal Hypoxia Sensitizes β-Adrenoceptors in the Embryonic Heart but Causes Postnatal Desensitization. American Journal of Physiology - Regulatory, Integrative and Comparative Physiology . 2009;297(2):R258–R264. doi: 10.1152/ajpregu.00167.2009. [DOI] [PubMed] [Google Scholar]
- 245.Chen J., Smith L. E. H. Retinopathy of Prematurity. Angiogenesis . 2007;10(2):133–140. doi: 10.1007/s10456-007-9066-0. [DOI] [PubMed] [Google Scholar]
- 246.Casini G., Dal Monte M., Fornaciari I., Filippi L., Bagnoli P. The β-Adrenergic System as a Possible New Target for Pharmacologic Treatment of Neovascular Retinal Diseases. Progress in Retinal and Eye Research . 2014;42:103–129. doi: 10.1016/j.preteyeres.2014.06.001. [DOI] [PubMed] [Google Scholar]
- 247.Ristori C., Filippi L., Dal Monte M., et al. Role of the Adrenergic System in a Mouse Model of Oxygen-Induced Retinopathy: Antiangiogenic Effects of β-Adrenoreceptor Blockade. Investigative Opthalmology & Visual Science . 2011;52(1):155–170. doi: 10.1167/iovs.10-5536. [DOI] [PubMed] [Google Scholar]
- 248.Dal Monte M., Filippi L., Bagnoli P. Beta3-Adrenergic Receptors Modulate Vascular Endothelial Growth Factor Release in Response to Hypoxia Through the Nitric Oxide Pathway in Mouse Retinal Explants. Naunyn-Schmiedeberg’s Archives of Pharmacology . 2013;386(4):269–278. doi: 10.1007/s00210-012-0828-x. [DOI] [PubMed] [Google Scholar]
- 249.Oikawa F., Nakahara T., Akanuma K., et al. Protective Effects of the β3-Adrenoceptor Agonist CL316243 Against N-Methyl-D-Aspartate-Induced Retinal Neurotoxicity. Naunyn-Schmiedeberg’s Archives of Pharmacology . 2012;385(11):1077–1081. doi: 10.1007/s00210-012-0796-1. [DOI] [PubMed] [Google Scholar]
- 250.Jiang Y., Zhang Q., Steinle J. J. Beta-Adrenergic Receptor Agonist Decreases VEGF Levels Through Altered eNOS and PKC Signaling in Diabetic Retina. Growth Factors . 2015;33(3):192–199. doi: 10.3109/08977194.2015.1054990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Ji Y., Chen S., Li K., Xiao X., Zheng S., Xu T. The Role of β-Adrenergic Receptor Signaling in the Proliferation of Hemangioma-Derived Endothelial Cells. Cell Division . 2013;8(1):p. 1. doi: 10.1186/1747-1028-8-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Barlow C. F., Priebe C. J., Mulliken J. B., et al. Spastic Diplegia as a Complication of Interferon Alfa-2a Treatment of Hemangiomas of Infancy. The Journal of Pediatrics . 1998;132(3):527–530. doi: 10.1016/s0022-3476(98)70034-4. [DOI] [PubMed] [Google Scholar]
- 253.Boon L. M., MacDonald D. M., Mulliken J. B. Complications of Systemic Corticosteroid Therapy for Problematic Hemangioma. Plastic and Reconstructive Surgery . 1999;104(6):1616–1623. doi: 10.1097/00006534-199911000-00002. [DOI] [PubMed] [Google Scholar]
- 254.Dubois J., Hershon L., Carmant L., Bélanger S., Leclerc J. M., David M. Toxicity Profile of Interferon Alfa-2b in Children: A Prospective Evaluation. The Journal of Pediatrics . 1999;135(6):782–785. doi: 10.1016/s0022-3476(99)70104-6. [DOI] [PubMed] [Google Scholar]
- 255.Boye E., Yu Y., Paranya G., Mulliken J. B., Olsen B. R., Bischoff J. Clonality and Altered Behavior of Endothelial Cells From Hemangiomas. Journal of Clinical Investigation . 2001;107(6):745–752. doi: 10.1172/jci11432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Greenberger S., Boscolo E., Adini I., Mulliken J. B., Bischoff J. Corticosteroid Suppression of VEGF-A in Infantile Hemangioma-Derived Stem Cells. New England Journal of Medicine . 2010;362(11):1005–1013. doi: 10.1056/nejmoa0903036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Takahashi K., Mulliken J. B., Kozakewich H. P., Rogers R. A., Folkman J., Ezekowitz R. A. Cellular Markers That Distinguish the Phases of Hemangioma during Infancy and Childhood. Journal of Clinical Investigation . 1994;93(6):2357–2364. doi: 10.1172/jci117241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Buckmiller L. M., Munson P. D., Dyamenahalli U., Dai Y., Richter G. T. Propranolol for Infantile Hemangiomas: Early Experience at a Tertiary Vascular Anomalies Center. The Laryngoscope . 2010;120(4):676–681. doi: 10.1002/lary.20807. [DOI] [PubMed] [Google Scholar]
- 259.Léauté-Labrèze C., de la Roque E. D., Hubiche T., Boralevi F., Thambo J. B., Taieb A. Propranolol for Severe Hemangiomas of Infancy. New England Journal of Medicine . 2008;358(24):2649–2651. doi: 10.1056/nejmc0708819. [DOI] [PubMed] [Google Scholar]
- 260.Sans V., de la Roque E. D., Berge J., et al. Propranolol for Severe Infantile Hemangiomas: Follow-Up Report. Pediatrics . 2009;124(3):e423–e431. doi: 10.1542/peds.2008-3458. [DOI] [PubMed] [Google Scholar]
- 261.Truong M. T., Chang K. W., Berk D. R., Heerema-McKenney A., Bruckner A. L. Propranolol for the Treatment of a Life-Threatening Subglottic and Mediastinal Infantile Hemangioma. The Journal of Pediatrics . 2010;156(2):335–338. doi: 10.1016/j.jpeds.2009.10.010. [DOI] [PubMed] [Google Scholar]
- 262.Chisholm K. M., Chang K. W., Truong M. T., Kwok S., West R. B., Heerema-McKenney A. E. β-Adrenergic Receptor Expression in Vascular Tumors. Modern Pathology . 2012;25(11):1446–1451. doi: 10.1038/modpathol.2012.108. [DOI] [PubMed] [Google Scholar]
- 263.Storch C. H., Hoeger P. H. Propranolol for Infantile Haemangiomas: Insights into the Molecular Mechanisms of Action. British Journal of Dermatology . 2010;163(2):269–274. doi: 10.1111/j.1365-2133.2010.09848.x. [DOI] [PubMed] [Google Scholar]
- 264.Eldh M., Olofsson Bagge R., Lässer C., et al. MicroRNA in Exosomes Isolated Directly From the Liver Circulation in Patients With Metastatic Uveal Melanoma. BMC Cancer . 2014;14(1):p. 962. doi: 10.1186/1471-2407-14-962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Tsai K. K., Bollin K. B., Patel S. P. Obstacles to Improving Outcomes in the Treatment of Uveal Melanoma. Cancer . 2018;124(13):2693–2703. doi: 10.1002/cncr.31284. [DOI] [PubMed] [Google Scholar]
- 266.Bustamante P., Miyamoto D., Goyeneche A., et al. Beta-Blockers Exert Potent Anti-Tumor Effects in Cutaneous and Uveal Melanoma. Cancer Medicine . 2019;8(17):7265–7277. doi: 10.1002/cam4.2594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Yang E. V., Kim S. J., Donovan E. L., et al. Norepinephrine Upregulates VEGF, IL-8, and IL-6 Expression in Human Melanoma Tumor Cell Lines: Implications for Stress-Related Enhancement of Tumor Progression. Brain, Behavior, and Immunity . 2009;23(2):267–275. doi: 10.1016/j.bbi.2008.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Dal Monte M., Calvani M., Cammalleri M., Favre C., Filippi L., Bagnoli P. β-Adrenoceptors as Drug Targets in Melanoma: Novel Preclinical Evidence for a Role of β3-Adrenoceptors. British Journal of Pharmacology . 2019;176(14):2496–2508. doi: 10.1111/bph.14552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Filippi L., Bruno G., Domazetovic V., Favre C., Calvani M. Current Therapies and New Targets to Fight Melanoma: A Promising Role for the β3-Adrenoreceptor. Cancers . 2020;12(6):p. 1415. doi: 10.3390/cancers12061415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Armaiz-Pena G. N., Lutgendorf S. K., Cole S. W., Sood A. K. Neuroendocrine Modulation of Cancer Progression. Brain, Behavior, and Immunity . 2009;23(1):10–15. doi: 10.1016/j.bbi.2008.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.De Giorgi V., Grazzini M., Gandini S., et al. Treatment With β-Blockers and Reduced Disease Progression in Patients With Thick Melanoma. Archives of Internal Medicine . 2011;171(8):779–781. doi: 10.1001/archinternmed.2011.131. [DOI] [PubMed] [Google Scholar]
- 272.De Giorgi V., Grazzini M., Benemei S., et al. Propranolol for Off-Label Treatment of Patients With Melanoma: Results From a Cohort Study. JAMA Oncology . 2018;4(2):p. e172908. doi: 10.1001/jamaoncol.2017.2908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Moretti S., Massi D., Farini V., et al. β-Adrenoceptors are Upregulated in Human Melanoma and Their Activation Releases Pro-Tumorigenic Cytokines and Metalloproteases in Melanoma Cell Lines. Laboratory Investigation . 2013;93(3):279–290. doi: 10.1038/labinvest.2012.175. [DOI] [PubMed] [Google Scholar]
- 274.Abdulkareem N. M., Bhat R., Powell R. T., et al. Screening of GPCR Drugs for Repurposing in Breast Cancer. Frontiers in Pharmacology . 2022;13:p. 1049640. doi: 10.3389/fphar.2022.1049640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Pushpakom S., Iorio F., Eyers P. A., et al. Drug Repurposing: Progress, Challenges and Recommendations. Nature Reviews Drug Discovery . 2019;18(1):41–58. doi: 10.1038/nrd.2018.168. [DOI] [PubMed] [Google Scholar]
- 276.Farhoumand L. S., Liu H., Tsimpaki T., et al. Blockade of SS-Adrenergic Receptors by Nebivolol Enables Tumor Control Potential for Uveal Melanoma in 3D Tumor Spheroids and 2D Cultures. International Journal of Molecular Sciences . 2023;24(6):p. 5894. doi: 10.3390/ijms24065894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Coelho M., Soares-Silva C., Brandão D., Marino F., Cosentino M., Ribeiro L. β-Adrenergic Modulation of Cancer Cell Proliferation: Available Evidence and Clinical Perspectives. Journal of Cancer Research and Clinical Oncology . 2017;143(2):275–291. doi: 10.1007/s00432-016-2278-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Asif H., Barnett S. D., Buxton I. L. O. β3 Adrenergic Receptor Signaling in the Human Myometrium. Reproductive Sciences . 2023;30(1):124–134. doi: 10.1007/s43032-022-00917-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Carnet Le Provost K., Kepp O., Kroemer G., Bezu L. Trial Watch: Beta-Blockers in Cancer Therapy. OncoImmunology . 2023;12(1):p. 2284486. doi: 10.1080/2162402X.2023.2284486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Puzderova B., Belvoncikova P., Grossmannova K., et al. Propranolol, Promising Chemosensitizer and Candidate for the Combined Therapy Through Disruption of Tumor Microenvironment Homeostasis by Decreasing the Level of Carbonic Anhydrase IX. International Journal of Molecular Sciences . 2023;24(13):p. 11094. doi: 10.3390/ijms241311094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Switzer B., Puzanov I., Gandhi S., Repasky E. A. Targeting Beta-Adrenergic Receptor Pathways in Melanoma: How Stress Modulates Oncogenic Immunity. Melanoma Research . 2024;34(2):89–95. doi: 10.1097/CMR.0000000000000943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Burger A., Benicke M., Deten A., Zimmer H. G. Catecholamines Stimulate Interleukin-6 Synthesis in Rat Cardiac Fibroblasts. American Journal of Physiology - Heart and Circulatory Physiology . 2001;281(1):H14–H21. doi: 10.1152/ajpheart.2001.281.1.H14. [DOI] [PubMed] [Google Scholar]
- 283.Shakhar G., Ben-eliyahu S. In Vivo β-Adrenergic Stimulation Suppresses Natural Killer Activity and Compromises Resistance to Tumor Metastasis in Rats. The Journal of Immunology . 1998;160(7):3251–3258. doi: 10.4049/jimmunol.160.7.3251. [DOI] [PubMed] [Google Scholar]
- 284.Krizanova O., Babula P., Pacak K. Stress, Catecholaminergic System and Cancer. Stress: The International Journal on the Biology of Stress . 2016;19(4):419–428. doi: 10.1080/10253890.2016.1203415. [DOI] [PubMed] [Google Scholar]
- 285.Qin J. F., Jin F. J., Li N., et al. Adrenergic Receptor β2 Activation by Stress Promotes Breast Cancer Progression Through Macrophages M2 Polarization in Tumor Microenvironment. BMB Reports . 2015;48(5):295–300. doi: 10.5483/bmbrep.2015.48.5.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Zhou J., Liu Z., Zhang L., et al. Activation of β2-Adrenergic Receptor Promotes Growth and Angiogenesis in Breast Cancer by Down-Regulating PPARγ. Cancer Res Treat. . 2020;52(3):830–847. doi: 10.4143/crt.2019.510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Mravec B., Horvathova L., Hunakova L. Neurobiology of Cancer: The Role of β-Adrenergic Receptor Signaling in Various Tumor Environments. International Journal of Molecular Sciences . 2020;21(21):p. 7958. doi: 10.3390/ijms21217958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Parker J. A., Mattos C. The K-Ras, N-Ras, and H-Ras Isoforms: Unique Conformational Preferences and Implications for Targeting Oncogenic Mutants. Cold Spring Harb Perspect Med. . 2018;8(8):p. a031427. doi: 10.1101/cshperspect.a031427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Anitole-Misleh K. G., Brown K. M. Developmental Regulation of Catecholamine Levels During Sea Urchin Embryo Morphogenesis. Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology . 2004;137(1):39–50. doi: 10.1016/j.cbpb.2003.09.001. [DOI] [PubMed] [Google Scholar]
- 290.Kim M. O., Na S. I., Lee M. Y., Heo J. S., Han H. J. Epinephrine Increases DNA Synthesis via ERK1/2 S Through cAMP, Ca(2+)/PKC, and PI3K/Akt Signaling Pathways in Mouse Embryonic Stem Cells. Journal of Cellular Biochemistry . 2008;104(4):1407–1420. doi: 10.1002/jcb.21716. [DOI] [PubMed] [Google Scholar]
- 291.Rains S. L., Amaya C. N., Bryan B. A. Beta-Adrenergic Receptors are Expressed Across Diverse Cancers. Oncoscience . 2017;4(7-8):95–105. doi: 10.18632/oncoscience.357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Schuller H. M. Beta-Adrenergic Signaling, a Novel Target for Cancer Therapy? Oncotarget . 2010;1(7):466–469. doi: 10.18632/oncotarget.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Zhang D., Ma Q., Wang Z., et al. β2-Adrenoceptor Blockage Induces G1/S Phase Arrest and Apoptosis in Pancreatic Cancer Cells via Ras/Akt/NFκB Pathway. Molecular Cancer . 2011;10(1):p. 146. doi: 10.1186/1476-4598-10-146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Cole S. W., Sood A. K. Molecular Pathways: Beta-Adrenergic Signaling in Cancer. Clinical Cancer Research . 2012;18(5):1201–1206. doi: 10.1158/1078-0432.ccr-11-0641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Peixoto R., Pereira Md., Oliveira M. Beta-Blockers and Cancer: Where are We? Pharmaceuticals . 2020;13(6):p. 105. doi: 10.3390/ph13060105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Chen H., Zhang W., Cheng X., et al. β2‐AR Activation Induces Chemoresistance by Modulating P53 Acetylation Through Upregulating Sirt1 in Cervical Cancer Cells. Cancer Science . 2017;108(7):1310–1317. doi: 10.1111/cas.13275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Gujral D. M., Lloyd G., Bhattacharyya S. Effect of Prophylactic Betablocker or ACE Inhibitor on Cardiac Dysfunction & Heart Failure During Anthracycline Chemotherapy ± Trastuzumab. The Breast . 2018;37:64–71. doi: 10.1016/j.breast.2017.10.010. [DOI] [PubMed] [Google Scholar]
- 298.Seidel J. A., Otsuka A., Kabashima K. Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations. Frontiers Oncology . 2018;8:p. 86. doi: 10.3389/fonc.2018.00086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Kim H. S., Park Y. H., Lee H. S., Kwon M. J., Song J. H., Chang I. B. Propranolol Inhibits the Proliferation of Human Glioblastoma Cell Lines Through Notch1 and Hes1 Signaling System. J. Korean Neurosurg. Soc. . 2021;64(5):716–725. doi: 10.3340/jkns.2021.0068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Liao X., Che X., Zhao W., Zhang D., Bi T., Wang G. The β-Adrenoceptor Antagonist, Propranolol, Induces Human Gastric Cancer Cell Apoptosis and Cell Cycle Arrest via Inhibiting Nuclear Factor κB Signaling. Oncology Reports . 2010;24(6):1669–1676. doi: 10.3892/or_00001032. [DOI] [PubMed] [Google Scholar]
- 301.Pasquier E., Ciccolini J., Carre M., et al. Propranolol Potentiates the Antiangiogenic Effects and Anti-Tumor Efficacy of Chemotherapy Agents: Implication in Breast Cancer Treatment. Oncotarget . 2011;2(10):797–809. doi: 10.18632/oncotarget.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Wang Z., Li S., Wang R., et al. The Protective Effects of the β3 Adrenergic Receptor Agonist BRL37344 Against Liver Steatosis and Inflammation in a Rat Model of High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease (NAFLD) Molecular Medicine . 2020;26(1):p. 54. doi: 10.1186/s10020-020-00164-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Ratan Y., Rajput A., Maleysm S., et al. An Insight into Cellular and Molecular Mechanisms Underlying the Pathogenesis of Neurodegeneration in Alzheimer’s Disease. Biomedicines . 2023;11(5):p. 1398. doi: 10.3390/biomedicines11051398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Tournissac M., Vu T. M., Vrabic N., et al. Repurposing Beta-3 Adrenergic Receptor Agonists for Alzheimer’s Disease: Beneficial Effects in a Mouse Model. Alzheimer’s Research & Therapy . 2021;13(1):p. 103. doi: 10.1186/s13195-021-00842-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Hanssen M. J., Hoeks J., Brans B., et al. Short-Term Cold Acclimation Improves Insulin Sensitivity in Patients With Type 2 Diabetes Mellitus. Nature Medicine . 2015;21(8):863–865. doi: 10.1038/nm.3891. [DOI] [PubMed] [Google Scholar]
- 306.Belibi F. A., Reif G., Wallace D. P., et al. Cyclic AMP Promotes Growth and Secretion in Human Polycystic Kidney Epithelial Cells. Kidney International . 2004;66(3):964–973. doi: 10.1111/j.1523-1755.2004.00843.x. [DOI] [PubMed] [Google Scholar]
- 307.Gattone V. H., Wang X., Harris P. C., Torres V. E. Inhibition of Renal Cystic Disease Development and Progression by a Vasopressin V2 Receptor Antagonist. Nature Medicine . 2003;9(10):1323–1326. doi: 10.1038/nm935. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data used to support the findings of this study are included within the article and are available upon request from the corresponding author.
