1,996
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      Acta Materia Medica now indexed by SCOPUS from May 2024. Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      G protein-coupled receptor-biased signaling: potential drug discovery to facilitate treatment of metabolic diseases

      Published
      review-article
      Bookmark

            Abstract

            G protein-coupled receptors (GPCRs) are important, potential drug targets for the treatment of metabolic disorders, such as obesity. GPCRs crosstalk with several transducers, including heterotrimeric G proteins, GPCR kinases (GRKs), and β-arrestins. GPCR-biased agonism has raised the potential of novel drug development to preferentially activate therapeutic signaling pathways over pathways that lead to unwanted side effects. The obesity epidemic and its metabolic complications continue to be a major global public health threat but effective treatments are limited. The accelerated development of structural techniques, like X-ray crystallography and cryo-electron microscopy, has paved the way to understanding how biased agonism measured at GPCRs results in specific downstream physiologic responses. Herein some well-validated GPCR targets are briefly summarized and several new and promising receptors for obesity treatment are outlined. This review highlights the significance of deciphering the role of GPCRs in obesity pathology and biased signaling for drug development. We anticipate the review will facilitate the development of novel GPCR-targeted anti-obesity drugs that lead to heightened therapeutic efficacy with decreased side effect profiles.

            Main article text

            1. INTRODUCTION

            An obese or overweight body habitus is defined as excess body fat mass due to a chronic accumulation of energy intake over energy expenditure. A body mass index (BMI) > 25 kg/m2 and > 30 kg/m2 is considered overweight and obese, respectively. Obesity increases the risk of diseases, such as cancer, cardiovascular diseases, and type 2 diabetes, and complicates the management of many diseases [14]. Generally, factors accounting for increasing obesity prevalence involve the intake of energy-dense food with a reduction in physical activity [5], as well as chronic stress, circadian desynchronization [6], and sleep deprivation [7].

            The global obesity prevalence is increasing and has nearly tripled since 1975. This phenomenon has caused healthcare providers to focus on obesity management, including lifestyle improvement and medication development [4]. Notably, short-term behavioral interventions are not ideal for long-term weight loss. In addition, lifestyle interventions only provide moderate efficacy against obesity. Drug discovery for the pharmacologic management of obesity has been extremely challenging and failed due to safety concerns [8]. Most of the previously approved anti-obesity drugs have been withdrawn because of adverse cardiovascular effects (sibutramine, dexfenfluramine, and rainbow pills), increased risk of depression (rimonabant), or drug addiction and abuse (methamphetamine) [8]. Among the small molecule drugs, only phentermine is not associated with adverse cardiovascular effects and can be used long-term as an anti-obesity drug [8].

            Since the discovery of leptin, a mechanistic understanding of energy homeostasis has been achieved, but how to translate the leptin studies to clinical use has not been determined. The central nervous system controls appetite and systemic energy metabolism but direct modulation of these signaling pathways needs specific and selective targeting of cellular circuits, which is difficult. Recently, clinical trials involving therapeutic agents targeting the glucagon-like peptide 1 receptor (GLP1R), such as semaglutide, indicated drug-based management of metabolic diseases, including obesity, may be forthcoming [911]. To balance food intake and energy expenditure, novel drugs targeting the neuroendocrine system mediate bidirectional crosstalk between the central nervous system and the periphery. Understanding the molecular mechanism that balances appetite and energy expenditure provides insight into metabolic disease drug discovery [12].

            G protein-coupled receptors (GPCRs) are important membrane proteins for transducing signals involving ions, odorants, hormones, neurotransmitters, and other stimuli from the extracellular matrix into the cell [13]. The classic signal transduction through GPCRs is dependent on activation of heterotrimeric G proteins, which are composed of three subunits (Gα, Gβ, and Gγ). Interestingly, only four G-protein families have been classified (Gs, Gi/o, Gq/11, and G12/13). The G-protein families couple to diverse receptors, which is then translated into diverse physiologic effects. A conserved mechanism exists for receptor-catalyzed G-protein activation that involves alterations in the nucleotide-bound state of the Gα subunit [14]. Arrestins are responsible for desensitizing GPCRs. Compared to G protein diversity, there are only four arrestins widely distributed in a variety of tissues. Generally, phosphorylation of GPCRs by GPCR kinases (GRKs) desensitizes G-protein signaling and promotes arrestin recruitment [15]. Biased agonism is primarily focused on defining if the ligand is biased towards G protein or arrestin. A full agonist that activates multiple pathways and leads to side effects is referred to as a balanced ligand. Likewise, a drug that increases or biases activity transduction of a specific signaling pathway is referred to as a biased ligand [16].

            Biased agonism is a broad concept that attempts to implement agonist use in clinical practice. Structural studies of various GPCRs and molecular dynamics of GPCRs in aqueous solutions indicate how GPCRs mediate biased signaling. Biased agonism fits into functional selectivity, which may be afforded using a single agonist. A classic example is the dopamine receptor discovered by Rashid and co-workers. Specifically, the dopamine receptors, D1 and D2, have been shown to be coupled to Gs and Gi proteins, respectively, according to the International Union of Basic and Clinical Pharmacology (IUPHAR). D1 and D2 can form a heterodimer which do not couple to Gs or Gi proteins. Instead, the dimer couples to Gq, which activates cAMP and calcium signals [17]. In addition to G protein selectivity, signaling bias of GPCRs involves bias on G protein or arrestin triggering.

            The basis for signaling bias involves the ability of ligands to stabilize active conformations of a GPCR, which are distinct from stabilization by other agonists that transduce different signaling pathways, such as GRKs or β-arrestins [18]. Drug development based on signaling bias leads to the discovery of novel and more effective drugs with fewer adverse effects [19]. A classic example of signaling bias is G protein-biased agonism on μ-opioid receptors, which would yield safer analgesic therapeutics because induction of respiratory depression through β-arrestin signaling would be less likely [20]. Thus, GPCR-biased signaling provides hope for drug discovery and development that improves energy expenditure or metabolic homeostasis and has higher potency while avoiding adverse effects.

            Given the possible therapeutic effect of modulating GPCR signaling pathways in obesity, we summarized the roles of food intake-related gut hormones and their receptors involving ghrelin, glucagon-like peptide 1 (GLP1), cholecystokinin (CKK), peptide tyrosine tyrosine (PYY), apelin, α-melanocyte-stimulating hormone (α-MSH), and cannabinoid-1 receptor. The potential drug leads against these receptors are summarized in Table 1 . We speculate that these findings will provide insight into novel drug discovery against GPCRs with potential therapeutic benefits in obesity treatment.

            Table 1 |

            Therapeutic agents with biased signaling approved or in clinical trials.

            LigandLigand typeReceptorSignaling biasDevelopment statusManufacturerreference
            ExenatidepeptideGLP-1Rβ-arrestin-2 recruitmentApproved by FDAAstraZeneca[21]
            Semaglutideacylated peptideGLP-1RFull agonist for both pathwaysApproved by FDANovo Nordisk[22]
            Liraglutideacylated peptideGLP-1Rβ-arrestin-2 recruitmentApproved by FDANovo Nordisk[22]
            AlbiglutideGLP-1-albumin proteinGLP-1RFull agonist for both pathwaysApproved by FDA but withdrawn due to economic reasonsGlaxoSmithKline[23]
            LixisenatidepeptideGLP-1RFull agonist for both pathwaysApprovedSanofi[23]
            DulaglutideGLP-1-Fc conjugateGLP-1RGαiApprovedEli Lilly[24]
            PF-5190457Small moleculeGHSR1aInverse agonistCompleted phase 1 trialsPfizer[25]
            MacrilenModified peptideGHSR1aGαqApprovedAEterna Zentaris[26]
            JMV3002miscellaneous compoundsGHSR1aPartial agonistPreclinical studyAEterna Zentaris[27, 28]
            NNC0165-1875peptideY2RFull agonist for both pathwaysCompleted phase 2 trialsNovo Nordisk[8]
            AMG3054peptideAPJRGαiPreclinical studyAmgen[29]
            SetmelanotideCyclic peptideMC4RGαqApprovedRhythm Pharmaceuticals[30]
            ORG27569Small moleculeCB1β-arrestin-dependent ERK1/2 pathwaysPreclinical studyPfizer[31]
            CP55940Small moleculeCB1Full agonistAs research toolPfizer[32]

            2. ROLES OF ENDOGENOUS GHRELIN ON ENERGY HOMEOSTASIS

            Ghrelin, also called the hunger hormone, is a gastric peptide hormone related to food intake, body weight, and taste sensation. Ghrelin is an orexigenic peptide hormone secreted from the stomach in response to hunger, which in turn stimulates the ghrelin receptor in the brain to initiate appetite [33, 34]. The ghrelin receptor, growth hormone secretagogue receptor (GSHR), is a class A GPCR that couples to Gαq/11, Gαi/o, and Gα11/13, as well as β-arrestin-based scaffolds [35]. Thus, ghrelin receptor activation transduces biased signals for diverse physiologic responses. Ghrelin receptor coupling to Gi and Gq has been elucidated by cryo-EM technology [36, 37]. Compared to the ghrelin receptor conformations coupled to Gq and Gi, only minor conformational differences were demonstrated ( Figure 1 ).

            Figure 1 |

            Structural basis of ghrelin receptor activation.

            (A) Complex of ghrelin receptor coupled to Gi protein bound with ghrelin (PDB ID: 7NA7). The ghrelin receptors (Gαi, Gβ, and Gγ) are displayed as ribbon colored in the sky (orchid, light green, and pink, respectively). The ligand ghrelin is shown in green stick. (B) Complex of ghrelin receptor-Gq bound to ghrelin (PDB ID: 7F9Y). The ghrelin receptors (Gαq, Gβ, and Gγ) are displayed as ribbon colored in the sky (forest green, light green, and pink, respectively). Ghrelin is shown in brown stick. (C) Structures of GHSR bound with agonist ghrelin and the inverse agonist, PF-519045. Superimposed of active (PDB ID: 7W2Z) and inactive (PDB ID: 7F83) conformation of GHSR (left panel). The blue and orange ribbon indicate active and inactive states of GHSR, respectively. The TM6 of GHSR displayed an outward movement (red arrow) in active conformation. The middle panel shows the key binding sites between ghrelin and GHSR. The polar interaction network, which is critical in activation, is composed of S217, R283, and N305. The right panel represents the binding of PF-5190457 in GHSR. D99 and S308 are important in recognition of the inverse agonist.

            In addition to the orthosteric binding pocket composed of the ghrelin receptor transmembrane helices (TM) 3, 4, 5, 6, and 7, acyl-ghrelin occupies an extended pocket involved in the second extracellular loop (ECL2). A mouse line in which the ghsr gene was deleted in all tissues was established for understanding the roles of GHSR, which exhibited improved insulin sensitivity, elevated energy expenditure, and reduced body fat mass [38]. Furthermore, neuronal ghsr knock-out mice were shown to have increased energy expenditure and thermogenesis without decreasing food intake, which indicated that suppressing central ghrelin signaling may shed light on a novel anti-obesity strategy that simultaneously boosts fat combustion and physical activity [39]. Notably, the mouse Gq/11 protein was shown to inversely correlate with UCP1 expression in brown adipose tissue (BAT), further reducing whole-body energy expenditure [40]. The result indicated that inhibition of Gq signaling may be a novel therapeutic strategy for obesity.

            As mentioned above, GHSR activation transduces Gq or Gi signaling, which depends on ligand selectivity. Because the ghrelin receptor displays a high basal activity representing 50% of its maximal activity [41], reducing the basal activity of the ghrelin receptor by an inverse agonist may be essential in developing anti-obesity agents. Previous studies have mined GHSR ligands and showed inverse agonism, such as [D-Arg1,D-Phe5,D-Trp7,9,Leu11]-substance P [42], PF-5190457 [43], and N-terminal fragment of liver-expressed antimicrobial peptide 2 (LEAP2) [44]. Among these ligands, PF-5190457 has progressed to a phase I clinical trial for type 2 diabetes treatment (NCT01522807 and NCT01247896). The crystal structure of GHSR bound to PF-5190457 and the cryo-EM structure of the ghrelin-GHSR-Go complex was determined, which revealed the GHSR inverse agonism binding mode [45]. As shown in the GHSR complex bound to PF-5190457 (PDB ID: 7F83), TM6 displays an outward shift, which enlarges the orthosteric pocket of GHSR and shows that the inactive conformation differs from the conformation of ghrelin-GHSR ( Figure 1 ). In the GHSR conformation activated by ghrelin, a polar interaction network formed by E1243.33, R2836.55, S2175.43, and N3057.35 is necessary for receptor activation, as shown by a mutagenesis study and functional assay ( Figure 1 ). Notably, S2175.43 and R2836.55 were observed in the agonist-bound conformation but not in the antagonist-bound structure. A bifurcated ligand-binding pocket was separated by a salt bridge between E1243.33 and R2836.55 in the interaction of PF-5190457 and GHSR. In addition, F1193.28 and Q1203.29 are essential for inverse agonism. Hydrogen bonds formed by PF-5190457 and D992.60 and S3087.38 of the GHSR are key facets for inverse agonist recognition by the GHSR. The bottom of the binding pocket is composed of F2796.51, W2766.48, and F3127.42.

            The hydrophobic clusters are also important for antagonism of GHSR by compound 21 [46]. LEAP2 is an endogenous antagonist of GHSR that is secreted from the liver and intestines that has been shown to fully inhibit GHSR activation by ghrelin and block the physiologic effects, such as food intake, growth hormone release, and blood glucose elevation [47]. LEAP2 and its N-terminal portion exhibit GHSR1a inverse agonism, which competitively antagonizes ghrelin-induced calcium mobilization and inositol-1-phosphate (IP1) production [44], revealing activity on Gi and Gq signaling. A randomized controlled trial moved one step forward to the clinical use of LEAP2 by showing that LEAP2 intervention suppresses ad libitum food intake and meal duration but not a liquid-mixed meal without additional hunger [48]. Even though further pharmaceutical and clinical exploration is needed, another step towards the clinical pharmacotherapy targeting the ghrelin-GHSR pathway for obesity, type 2 diabetes, and other related metabolic diseases was achieved [49].

            Based on the GHSR structural information and biological studies, novel development of non-peptide small molecules targeting GHSR-biased signaling has been carried out [25], such as JMV1843, AZ-GHS-38, and JMV3002.

            As mentioned above, ghrelin is a hunger hormone but ghrelin mediates diverse physiologic actions, including growth hormone release, gastric motility, reward behavior, and emotion. Developing these non-peptide agents is necessary for avoiding the on-target side effects. The discovery of small molecules with biased signaling can distinguish between ghrelin responses.

            3. GLUCAGON-LIKE PEPTIDE 1 (GLP-1) RECEPTOR AGONISTS FOR GLUCOSE AND ENERGY METABOLISM

            GLP-1 receptor is an established drug target against metabolic diseases. GLP-1 receptor belongs to class B GPCRs and is coupled to Gs protein. GLP-1 receptor is characterized by having long, extracellular N-terminal domains [50]. GLP-1 receptor is important in the secretion of insulin. Thus, GLP-1 agonists are widely considered for drug development in the treatment of diabetes and obesity [51]. Despite clinical success, GLP-1 receptor peptide agonists are suboptimal due to administration and side effect profiles, including nausea and vomiting [52]. For peptide GLP-1, mutagenesis has been carried out to clarify the signaling profiles, aiming to avoid the adverse effects in future studies [53]. Mutating any single residue of GLP-1 (7-36) to alanine results in a loss of ability to lower blood glucose. With T11A or S14A, the mutant reduces β-arrestin recruitment approximately 10-fold. With S17A or S18A, ERK1/2 phosphorylation increases β-arrestin recruitment approximately 5-fold. Mutations of three serines silence ERK1/2 phosphorylation while mutations of all four residues abolish β-arrestin 2 recruitment, ERK1/2 phosphorylation, and calcium mobilization but still stimulate cAMP and insulin secretion in mice and cells. These findings suggest that hydrogen bonding controls cell signaling, as well as an important regulatory hydroxyl patch in class B GPCR hormones.

            Several non-peptide GLP-1 receptor agonists have been identified, including PF-0688296, glutazumab, semaglutide, and TT-OAD2 [5456]. As shown in the structural comparison ( Figure 2 ), TT-OAD2 binding has very limited overlap with full length GLP-1 and the biased agonist, exendin-P5 (ExP5). Therefore, this compound series may modulate peptide activity in the physiologic response [57, 58]. It has been shown that TT-OAD2 inhibits cAMP, calcium mobilization, ERK1/2 phosphorylation, and β-arrestin responses meditated by GLP-1 and oxyntomodulin [56]. This phenomenon indicates that the presence of TT-OAD2 probably inhibits all endogenous peptide effects at higher concentrations, biasing receptor responses primarily to cAMP production mediated by the compound, and the signaling transduced by endogenous peptide may still occur when the compound is at a lower concentration. Similarly, TTP273 has been reported to display greater efficacy at lower levels, revealing the basal physiologic responses induced by the endogenous peptide ligands may be important for clinical effects [59]. ERK1/2 phosphorylation is not the only measurement for β-arrestin recruitment, although β-arrestin is required for ERK1/2 and CREB activation at pharmacologic doses of GLP-1 [60]. During GLP-1R activation, β-arrestin-2 (ARRB2) has a role in uncoupling G-proteins and recruits additional pathways, including phosphatidylinositol 3-kinase (PI3K), focal adhesion kinase (FAK), and ERK1/2 phosphorylation [61]. ARRB2 knockout mice were shown to have a minor role in GLP-1R internalization. In addition, ARRB2 has been shown to contribute to partial uncoupling of cAMP/PKA signaling in the physiologic concentration range of GLP1, which leads to a reduction in insulin secretion [60].

            Figure 2 |

            Structural models of GLP-1 receptor bind to different ligands.

            (A) The GLP-1R complex bound to the non-peptide ligand, TT-OAD2 (blue stick), viewed from transmembrane bundles (PDB ID: 6ORV). TT-OAD2 binds high up in the helical bundle and the pocket is composed of TM1-3 and ECL1-2. (B) Active state of GLP-1R bound to its endogenous peptide ligand, GLP-1 (blue ribbon and sticks), viewed from helical bundles (PDB ID: 5VAI). GLP-1 is clasped between the N-terminus and the transmembrane core of the receptor and further stabilized by ECLs. (C) The model of GLP-1R activated by the G-protein biased ligand ExP5 (blue ribbon and sticks) viewed from the cytoplasm (PDB ID: 6B3J). The receptor is shown in ribbons with carbons in light green.

            Biased agonism at the GLP-1R stimulates cAMP over β-arrestin recruitment, resulting in less receptor internalization. However, GLP-1R agonism or glucose-dependent insulinotropic polypeptide receptor (GIPR) infusion causes a heart rate increase [62, 63]. Combined GLP-1R/GIPR agonism appears to induce fewer side effects. Tirzepatide is a synthetic, linear peptide composed of 39 amino acids and has a half-life of approximately 5 days, which is suitable for weekly dosing [64]. A phase II clinical trial involving tirzepatide in type 2 diabetes patients reported a significant reduction in body weight (5%-10%) alongside substantial reductions in waist circumference following a 12-week administration [65]. GCGR agonism has a positive chronotropic and inotropic action on the heart [66]. To maximize the drug efficacy and mitigate safety, tri-agonists (GLP-1R/GCGR/GIPR) have been developed. A phase II trial involving the tri-agonist, retatrutide, showed a dose-dependent increase in heart rate that peaked at 24 weeks and declined thereafter; treatment for 48 weeks resulted in a substantial reduction in body weight [67].

            4. CHOLECYSTOKININ A RECEPTOR (CCKAR): AN ATTRACTIVE DRUG TARGET FOR METABOLIC DISEASES

            The CCKAR belongs to class A GPCRs that are recognized by CCK, which regulates nutrient homeostasis. The CCKAR is distributed in the pylorus, pancreas, and gallbladder, contributing to micelle formation, stimulating lipolytic and proteolytic enzymes delivery, and modulating the delivery rate of nutrients to regulate optimal food absorption [68, 69]. The CCK peptide with a sulfated tyrosine possesses high-affinity binding and full agonist activity to CCK1R. In the last three decades, enormous effort has been put forth to develop drugs to treat obesity and diabetes through the effects of CCK peptides and CCKAR [70]. However, none of the drugs have been approved for clinical use because some highly potent CCKAR agonists with a long duration of action, such as GW-5823, CE-326597, and Glaxo-11p, display adverse effects (diarrhea, nausea, and abdominal cramping) [70].

            The octapeptide, CCK-8 (DYMGWMDF), which is derived from the C-terminal of CCK, is responsible for CCKAR activation, manipulating the appetite, and satiety [68]. The sulfated CCK-8 (DYSO3HMGMWDF-NH2)-bound CCKAR structures complexed with Gi, Gs, or Gq heterotrimers, which were determined by cryo-EM technology, provide a structural basis for understanding the molecular details of ligand recognition and biased agonism during CCKAR activation [71]. By inspecting the orthosteric binding pocket of CCKAR occupied by sulfated CCK-8, the binding pocket was shown to be composed of TM3, 4, 5, 6, and 7 and ECL1, 2, and 3 ( Figure 3 ). As shown in the binding pocket, the conformations of ECLs favor recognition of the N-terminal of CCK-8. The sulfate group of Y2 in CCK-8 forms a polar contact with R197ECL2, prompting the aromatic ring of Y2 in the CCK-8 hydrophobic bound with F185ECL2, M195ECL2, and K105ECL1. The CCK-8 residues, including M3, G4, and W5, interact with the interior surface of ECL3 via A343ECL3, E344ECL3, L347ECL3, and S348ECL3. In addition, W5 and M6 in CCK-8 forms bifurcated hydrophobic cavities with the ECLs of CCKAR. For W5 in CCK-8, the side chain lies in the middle of the side chains of I3527.53 and R3366.58, making its indole nitrogen atom hydrogen bond with N3336.55 and the carbonyl group of the main chain interacts with R3366.58. M6 in CCK-8 occupies a shallow hydrophobic pocket, including F107ECL1, C196ECL2, T1183.29, and M1213.32. At the bottom of the CCKAR binding pocket, the conformation of D7 in CCK-8 is stabilized by a polar interaction network with H2105.39, N3336.55, R3366.58, and Y3607.43. The aromatic ring of F8 in CCK-8 contacts Y1764.60 through a polar hydrogen-π interaction.

            Figure 3 |

            Cryo-EM structures of sulfated CCK-8 (DYSO3HMGMWDF-NH2)-bound CCKAR in complex with G proteins.

            (A) Complex of CCKAR bound to sulfated CCK-8 coupled to Gi protein (PDB ID: 7EZH). (B) Complex of CCKAR bound to sulfated CCK-8 coupled to Gs protein (PDB ID: 7EZK). (C) Complex of CCKAR bound to sulfated CCK-8 coupled to Gq protein (PDB ID: 7EZM). The receptors, Gαi, Gαs, Gαq, Gβ, and Gγ, are shown as ribbons colored in light blue, yellow, light green, light pink, marine blue, and magentas. The peptide ligand sulfated CCK-8 is displayed in orange stick. The detailed residues of the binding pocket of each complex coupled to the indicated G protein are shown in the right side of each panel.

            The structural basis of the CCKAR binding pocket provides insight for anti-obesity drug discovery. One of the potential powerful tools is the use of a positive allosteric modulator (PAM), which displays no intrinsic agonist activity. The fully bioactive forms of CCK share the C-terminal amide and a sulfated tyrosine (sTyr) residue. Almost any reported mutagenesis or replacement of Gly-29, such as N-methylglycine or a fluorescent artificial amino acid, lead to major loss of CCKAR potency [72, 73]. Based on a systematic investigation of synthetic CCK-8 analogues with N-terminal linkage to fatty acids, peptide-based, long-acting, and stable highly selective CCKAR agonists have been identified and characterized [74]. By replacing Asp at the penultimate position of CCK-8 by DMeAsp, novel agonists with high CCKAR selectivity have been achieved. In addition, inhibition of food intake in a pig model for up to 48 h after subcutaneous injection, as well as increasing plasma half-lives, were observed with an sTyr modification. Furthermore, compound NN9056, an N-terminal C18 fatty acid derivative with a free carboxyl group of CCK8, has potential for further development as an injectable anti-obesity drug [75, 76]. Moreover, when replacing the unstable methionine with two CCK-8 norleucine residues, the modified peptide displays maximum CCKAR selectivity, efficacy, and potency [74].

            Individual CCKARs recognize and couple to divergent G-protein subtypes. Comparison of the three complexes formed by CCKAR and G-proteins suggest similar configurations. The most notable difference is at the tilts of the α5 helix of Gα subtypes (coupling ranking, Gq > Gs > Gi). The selectivity coupling between CCKAR and G proteins is important for the physiologic response and development of therapeutic strategies for metabolic disorders.

            5. PYY IN REGULATION OF SATIETY SIGNALS AND APPETITE EFFECTS

            PYY belongs to the neuropeptide Y (NPY) family. PYY is secreted as PYY1-36 along with GLP1 from the intestinal L cells [77]. PYY1-36 is rapidly cleaved by dipeptidyl peptidase 4 (DPP4) to PYY3-36, which activates NPY receptor 2 (Y2R) and decreases food intake [78]. PYY3-36 signaling in food intake inhibition has provided insight for anti-obesity drug discovery, like developing PYY3-36 analogs [79]. In agreement with this notion, phase I trials have been carried out for two PYY3-36 analogs (NN9748 and NNC0165-1875) for long-acting activity in obesity therapy. Phase II trials are on-going using NNC0165-1875 combined with semaglutide for its treatment against obesity [8].

            Structural understanding of ligand selectivity and biased signaling of Y2R is useful in the design of efficient and stabilized anti-obesity agents. Recently, the active state of Y2R bound to its endogenous ligands, including PYY3-36 and NPY, was resolved [80, 81]. In the structure of Y2R coupled with Gi protein, the agonist, NPY, adopts a hairpin shape with the unstructured N terminus and the α-helix in the C-terminal region running antiparallel and occupying a large binding pocket involving ECLs and helices II–VII of Y2R ( Figure 4 ). In appetite regulation, Y1R and Y2R display different responses and NPY-activated Y1R stimulates appetite, whereas NPY-activated Y2R suppresses food intake [82]. After cleavage of the two N-terminal amino acids, PPY3-36 preferentially activates Y2R but not Y1R [83]. Two Y2R cryo-EM structures bound to its endogenous ligands, NPY and PYY3-36, have been resolved and analyzed combined with molecular dynamics (MD) simulations and functional assays, elucidating Y2R activation and the molecular mechanism underlying exclusive binding of PYY3-36 to Y2R [81]. At the 6.58 position, the amino acid varies between Y1R and Y2R, which is F6.58 and V6.58, respectively. Another distinct feature of NPY/PYY3-36 recognition by Y2R occurs in the isoleucine-rich region of ECL2. Based on MD simulation analysis [81], the ECL2 of Y2R forms a relatively rigid hydrophobic cluster with PPY helix, but it is very flexible when simulated on Y1R. This finding may help explain the mechanism underlying the opposite physiologic response on appetite transduced by Y1R and Y2R activation.

            Figure 4 |

            Structural models of neuropeptide Y receptor viewed from the cytoplasm side.

            (A) The conformation of Y2R (pale green ribbons) activated by PYY3-36 (PDB ID: 7YON). N-terminal of PYY3-36 does not directly interact with Y2R. PYY3-36 is drawn in sticks and ribbons with carbons in orange. V6.58 is shown in spheres and sticks. (B) The Y2R model (pale green ribbons) bound to NPY (PDB ID: 7X9B). NPY (light blue) adopted a hairpin-like shape with the unstructured N-terminus and the α-helix in the C-terminus running antiparallel. The N-terminal of NPY stacked on top of ECL2. (C) The Y1R structure (teal ribbons) binding with NPY (PDB ID: 7X9A). In addition to the hairpin-like fold of NPY, the N-terminus of NPY shifted towards ECL3 and inserted deeply to the TM regions of Y1R. F6.58 is shown in spheres and sticks.

            6. APELIN ACTION IN ENERGY METABOLISM

            Apelin is a peptide that is involved in glucose and lipid metabolism [84]. Adipose tissue is a possible source of plasma apelin because apelin was shown to be expressed and released by cultured adipocytes [85]. Apelin is an endogenous ligand of the apelin receptor (angiotensin receptor like-1, the angiotensin II protein J receptor, or APJR). APJR is a member of the class A γ-group of GPCRs which has diverse physiologic roles in angiogenesis, vasoconstriction, and energy metabolism [86]. To develop a novel agent beneficial in balancing energy metabolism, it is important to understand the fundamental biology of ligand recognition and receptor signal transduction. Fortunately, both crystal [86] and cryo-EM [29] structures of APJR have been resolved in recent years.

            In the crystal structure of APJR bound to AMG3054, which is an apelin-17 mimetic peptide, a lactam constraining the curved two-site ligand binding mode was demonstrated. The C-terminal five amino acids of AMG3054 are inserted deeply into the orthosteric binding pocket nearly perpendicular to the membrane plane, which is termed “site 1.” In addition, the lactam ring formed by the side chain of K13 and E10 of the peptide made the main chain bend by approximately 90° from the membrane-perpendicular direction, termed “site 2” ( Figure 5 ). In agreement with AMG3054 binding, mutations, including D172ECL2A, E174ECL2A, D184ECL2A, and E1945.31A, showed no effect on apelin-13 binding affinity [86]. In healthy humans, pyr-apelin-13 displayed a rapid dose-dependent increase in blood flow and reproducible effect after a saline washout. In contrast, apelin-36 showed a more durable response [87]. This observation suggested that additional interactions of longer peptides may display a prolonged duration of action.

            Figure 5 |

            Crystal structure of APJR-AMG3054. The bound apelin-17 mimetic peptide, AMG3054, adopted a curved binding conformation through a two-site binding mode. The C-terminal five amino acids of the peptide inserted into the binding cavity, which is approximately perpendicular to the membrane plane (termed “site 1”). The N-terminal half of the peptide bent by approximately 90 degrees binding to the surface grooves of APJR (termed “site 2”). The PDB ID of the analyzed structure is 5VBL.

            7. MELANOCORTIN-4 RECEPTOR (MC4R) IN REGULATING ENERGY HOMEOSTASIS

            MC4R is expressed in the paraventricular nucleus (PVN), which is a key component of the leptin-melanocortin pathway and responsible for the maintenance of long-term energy balance in humans, which provides a blueprint for the energetic state controlled by the central nervous system [88]. Two decades ago it was shown that MC4R-null mice exhibit late-onset obesity [89]. MC4R is activated by pro-opiomelanocortin (POMC)-derived neuropeptides α- and β-melanocyte-stimulating hormone (MSH). In parallel, the Agouti-related peptide (AgRP), which is inhibited by leptin, has been proposed as an inverse agonist of MC4R [90]. Greater than 80 pathogenic mutations naturally occurring in the leptin-melanocortin pathway have been shown to result in severe obesity in human and rodent models [91].

            Unlike the direct interaction between ligands and their transmembrane bundles, the N-terminal domain of MC4R is responsible for ligand recognition and activation. The N-terminus of MC4R can act as a diffusible agonist; the minimal activation sequence is HLWNRS [90]. The first FDA-approved medication for obesity due to suspected leptin receptor (LEPR), POMC, or proprotein subtilisin/kexin type 1 (PCSK1) deficiency is a cyclic peptide (setmelanotide) that displays high affinity towards MC4R compared to natural α-MSH [92]. The cryo-EM structure of MC4R-Gs-protein bound to setmelanotide has been resolved [93, 94]. Combined with the structural data, a further functional assay suggested that the allosteric interaction between the orthosteric binding pocket and G-protein binding sites is mediated by TM3 and facilitated by TM6.

            Another study involving structural information of MC4R reported the complex with Gαs protein stimulated by α-MSH, selective ligand THIQ, and FDA-approved drugs (afamelanotide and bremelanotide), revealing the details of ligand recognition and receptor subtype selectivity [95]. Setmelanotide was shown to engage MC4R via hydrophobic interactions involving π-π stacking with F184, Y268, F284, and hydrogen bonding with S188 within the transmembrane regions ( Figure 6A ). Notably, one characteristic of setmelanotide binding to MC4R is a calcium ion coordinated by both the agonist and the receptor. The calcium ion was also observed in a similar position, while the antagonist SHU9119 ( Figure 6B ) binds to MC4R [94]. These structural studies provide insight into understanding the mechanism underlying MC4R activation and inhibition, setting the stage for development of new therapeutic agents to fight against obesity and other metabolic diseases.

            Figure 6 |

            Structural models of MC4R bind to the agonist, setmelanotide, and the antagonist, SHU9119.

            (A) Active conformation of MC4R binds to setmelanotide (PDB ID: 7PIU). Setmelanotide (yellow stick) engages MC4R (ribbon in light green) via hydrophobic interactions involving π-π stacking with F184, Y268, F284, and hydrogen bonding with S188. Ca2+ (green ball) is required for agonist binding. (B) Inactive state of MC4R induced by SHU9119 (PDB ID: 6W25). MC4R and SHU9119 are shown in blue ribbon and orange stick, respectively. The calcium ion (green ball) was also observed in a similar position as the antagonist, SHU9119.

            In addition to stimulation of Gαs signaling, inducing anorexogenic signaling in the hypothalamus to result in negative energy balance, MC4R activation triggers signaling pathways involved in Gαi, Gαq, and β-arrestin recruitment [96, 97]. Both T1503.53 and H158ICL2 regulation of the MC4R signaling profile are likely related to G-protein selectivity [93]. To fully elucidate G-protein selectivity of MC4R, especially to differentiate Gq and Gs coupling, the structure of an agonist bound with MC4R-Gq complex will be necessary. The structural findings will help improve the development of novel anti-obesity drugs targeting the MC4R.

            8. SIGNALING MECHANISM UNDERLYING CANNABINOID-1 RECEPTOR (CB1R) IN ANTI-OBESITY TREATMENT

            Obesity is related to an overactive endocannabinoid system. The human cannabinoid GPCRs, CB1 and CB2, have been the targets of intensive drug discovery efforts [98]. Progress in structure determination of cannabinoid receptors had identified the active, intermediate, and inactive conformations, providing valuable clues into signaling of cannabinoid receptors [99]. Diverse ligands can bind to the membrane-embedded orthosteric pocket or allosteric sites to modulate CB1R signaling via Gi/o or arrestin pathways. Understanding the bias signaling of CB1R may help decrease side effects in the therapeutic applications [100]. As the endocannabinoid system is involved in the regulation of energy metabolism, inverse agonists, such as rimonabant and tarabant, have been shown to be effective in the clinical treatment of obesity but failed to receive approval due to adverse CNS side effects [101]. Similarly, the CB1R inverse agonist, GFB-024, has been shown to mitigate diabetes-induced inflammation, displaying well-tolerated and no dose-limiting adverse effects [102]. Therefore, peripheral blockade of CB1R by inverse agonists avoiding CB1R binding in CNS might be an alternative strategy for obesity treatment.

            Binding of an allosteric modulator is believed to induce a conformational change in the receptor that affects the potency or efficacy of an orthosteric agonist to generate receptor conformational states with unique structural and functional phenotypes. From the high-resolution (2.6 Å) crystal structure of the human CB1R ( Figure 7A ), the membrane-proximal N-terminal region of CB1R has been shown to form a critical part of the ligand-binding pocket, which is distinct from other lipid-activated GPCRs [98]. One of the first reported allosteric modulators is ORG27569, which is an indole-containing small molecule that increases binding of the full agonist, CP55940, in a concentration-dependent mode favoring an overall inactive state for G protein coupling, as revealed by spectroscopic studies [103, 104]. The crystal structure of CB1R with ORG27569 and the agonist, CP55940, elucidated ORG27569, a negative allosteric modulator (NAM) of CB1R, binds to the extrahelical site within the inner leaflet of the membrane overlapping with a conserved site of cholesterol interaction in many GPCRs ( Figure 7B ).

            Figure 7 |

            Different conformations of CB1R.

            (A) Crystal structure of CB1R recognized by its antagonist, taranabant (PDB ID: 5U09). The inactive state of CB1R is shown in yellow ribbon. The antagonist, taranabant, is displayed in sticks and spheres with carbon in blue. (B) Ternary structure of CB1R determined by X-ray crystallography (PDB ID: 6KQI). The active conformation of CB1R is represented as a pink ribbon. The agonist, CP55940, and the negative allosteric modulator, ORG27569, is shown in sticks and spheres with carbon in grey and orange, respectively.

            Binding of ORG27569-CP55940 captures an intermediate state of CB1R, in which aromatic residues at the bottom of the orthosteric pocket adopt an inactive conformation, despite the large contraction of the binding pocket [100]. The reported crystallographic and cryo-EM structures of CB1R bound to the positive allosteric modulator (PAM), ZCZ011, showed that the PAM binds to an extrahelical site in TM2-TM3-TM4 surface [105]. Recently, the complex of CB1R-Gi bound with an analog of endocannabinoid, AMG315, found “toggle switch” residues (F2003.36 and W3566.48) and the intracellular side of TM2 are a determinant of efficacy in Gi signaling [106]. These structural studies showed binding to TM2-TM3-TM4 and regulation of TM2 rearrangement is critical in allosteric modulations of CB1R, filling a gap in understanding CB1R allostery and rational discovery of CB1R allosteric modulators.

            While blockade of CB1R has been shown to halt diet-induced obesity [107], the solved structures of CB1R have advanced drug leads or rational design for obesity treatment. A series of 3,4-diarylpyrazolines bearing rational pharmacophoric pendants designed to limit brain blood barrier penetration were synthesized and displayed high binding affinity and potent CB1R antagonistic activities, acting as AMPK activators [108].

            9. FUTURE PERSPECTIVES

            The development of X-ray crystallography and cryo-electron microscopy techniques have accelerated the understanding of the structural biology of GPCRs. GPCRs consist of seven transmembrane α-helices linked by three intracellular and three extracellular loops. The structure is essential for extracellular stimulus binding to extracellular loops or transmembrane bundles, then transduction through intracellular proteins involves G proteins, arrestins, and G protein kinases (GRKs). When GPCRs are activated, their conformations alter to a state that favors binding to those intracellular proteins for signal transmission. The allosteric ligands positively or negatively modulate the binding of orthosteric ligands. The allosteric sites are less conserved than orthosteric sites, leading to higher selectivity for a given receptor and consequently causing fewer adverse effects [109]. Other therapeutically promising ligands are biased agonists and biased allosteric modulators. The biased signaling of these drug leads can be measured or detected by receptor-G protein resonance energy transfer-based assays with fluorescence (FRET) and bioluminescence (BRET) biosensors, as well as cAMP, MAPK phosphorylation, and β-arrestin recruitment [110].

            The dynamics of GPCRs enable diversity across ligand binding pockets and intracellular binding sites that increase the engaged pathways to balance efficacy and adverse drug reactions [111, 112]. As an important target in obesity and type 2 diabetes treatment, drugs that target GLP-1R, like semaglutide and liraglutide, are successful in regulating blood glucose levels but with adverse drug reactions ranging from nausea and diarrhea to pancreatitis [113]. It has been reported that GLP-1R activation engage 15 different pathways, including Gs, Gq, G11, G14, G15, G12, G13, Gi1, Gi2, Gi3, GoA, GoB, GZ, GRK/Gβγ, and β-arrestin [114]. The agonists, GLP1(7-36), liraglutide, and exenatide, strongly activate Gi/o, while semaglutide and lixisenatide engage Gs, GRK/Gβγ, and β-arrestin pathways, which has been reported to have side effects, such as anxiety, nausea, and loss of appetite [115]. In addition, a small molecule agonist, danuglipron, displays biased engagement away from β-arrestin towards Gs and Gi/o [114], with the side effects, including nausea, vomiting, and diarrhea [116]. Given the pleiotropic signaling of cardiovascular angiotensin II type 1 receptor (AT1R), it is a target for drug development for cardiovascular homeostasis. In the case of A1TR activation, both Gq and β-arrestin pathways need to be inactive to suppress aldosterone production to prevent chronic heart failure and hypertension [117]. Instead of focusing solely on Gq or β-arrestin-dependent signaling, other G protein types, like Gi/o and G12/13, are also important in biased AT1R signal transduction [118].

            The current molecular biology toolkit for GPCR has powered drug discovery towards the generation of several potent molecules for treatment of metabolic diseases [119, 120]. One of the major medical needs is the prevention or therapeutics for metabolic diseases, like obesity. It has been predicted that > 500 million individuals will be obese by 2035 [121]. To date > 30 GPCRs have been implicated in the development and progression of β-cell dysfunction, insulin resistance, T2DM, and obesity but only GLP1R has been successfully targeted in therapeutic strategies [122].

            A lack of understanding of basic receptor pharmacology makes development of therapeutic drug leads immensely difficult [122]. Early stage target validation is required. Nonetheless, with the constant advances in technological platforms, including improved compound screening techniques, deorphanization strategies, novel targeting mechanisms, signaling bias, allostery, and the explosion in GPCR structural biology, combined with clinical data, we anticipate the medications will be aligned with the treatment paradigm in 10 years. Statistical significance is the historical benchmark to get a medication approved. In the modern era, drugs need to show substantial benefit to the healthcare system to engender payment. Exploitation in bias signaling of GPCR structural biology provides the possibility and successfully targeting these GPCRs is even increasing. The current review highlights the mechanism on agonism or antagonism of these receptors, paving the way for basic pharmacologic probes which will enable these targets to fulfill the promise for metabolic discase therapeutics.

            CONFLICTS OF INTEREST

            No conflicts of interest, financial or otherwise, are declared by the authors.

            REFERENCES

            1. Yoon YS, Kwon AR, Lee YK, Oh SW. Circulating Adipokines and Risk of Obesity Related Cancers: A Systematic Review and Meta-Analysis. Obesity Research & Clinical Practice. 2019. Vol. 13:329–339

            2. Powell-Wiley TM, Poirier P, Burke LE, Després JP, Gordon-Larsen P, Lavie CJ, et al.. Obesity and Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation. 2021. Vol. 143:e984–e1010

            3. DiStefano JK, Gerhard GS. Metabolic Dysfunction and Nonalcoholic Fatty Liver Disease Risk in Individuals with a Normal Body Mass Index. Current Opinion in Gastroenterology. 2023. Vol. 39:156–162

            4. WHO. Health service delivery framework for prevention and management of obesity. 2023

            5. Biological BMI Uncovers Hidden Health Risks and Is More Responsive to Lifestyle Shifts. Nature Medicine. 2023. Vol. 29:801–802

            6. Altaha B, Heddes M, Pilorz V, Niu Y, Gorbunova E, Gigl M, et al.. Genetic and Environmental Circadian Disruption Induce Weight Gain Through Changes in the Gut Microbiome. Molecular Metabolism. 2022. Vol. 66:101628

            7. Noh J. The Effect of Circadian and Sleep Disruptions on Obesity Risk. Journal of Obesity & Metabolic Syndrome. 2018. Vol. 27:78–83

            8. Müller TD, Blüher M, Tschöp MH, DiMarchi RD. Anti-Obesity Drug Discovery: Advances and Challenges. Nature Reviews Drug Discovery. 2022. Vol. 21:201–223

            9. Baggio LL, Drucker DJ. Glucagon-like peptide-1 receptor co-agonists for treating metabolic disease. Molecular Metabolism. 2021. Vol. 46:101090

            10. Campbell JE, Müller TD, Finan B, DiMarchi RD, Tschöp MH, D’Alessio DA. GIPR/GLP-1R dual agonist therapies for diabetes and weight loss-chemistry, physiology, and clinical applications. Cell Metabolism. 2023. Vol. 35:1519–1529

            11. Hanssen R, Rigoux L, Kuzmanovic B, Iglesias S, Kretschmer AC, Schlamann M, et al.. Liraglutide Restores Impaired Associative Learning in Individuals with Obesity. Nature Metabolism. 2023. Vol. 5:1352–1363

            12. Zhang C, Chen G, Tang G, Xu X, Feng Z, Lu Y, et al.. Multi-component Chinese medicine formulas for drug discovery: State of the art and future perspectives. Acta Materia Medica. 2023. Vol. 2:106–125

            13. Hilger D, Masureel M, Kobilka BK. Structure and dynamics of GPCR signaling complexes. Nature Structural & Molecular Biology. 2018. Vol. 25:4–12

            14. Rasmussen SG, DeVree BT, Zou Y, Kruse AC, Chung KY, Kobilka TS, et al.. Crystal Structure of the beta2 Adrenergic Receptor-Gs Protein Complex. Nature. 2011. Vol. 477:549–555

            15. Duan J, Liu H, Zhao F, Yuan Q, Ji Y, Cai X, et al.. GPCR Activation and GRK2 Assembly by a Biased Intracellular Agonist. Nature. 2023. Vol. 620:676–681

            16. Wang X, McFarland A, Madsen JJ, Aalo E, Ye L. The Potential of (19)F NMR Application in GPCR Biased Drug Discovery. Trends in Pharmacological Sciences. 2021. Vol. 42:19–30

            17. Rashid AJ, So CH, Kong MM, Furtak T, El-Ghundi M, Cheng R, et al.. D1-D2 Dopamine Receptor Heterooligomers with Unique Pharmacology are Coupled to Rapid Activation of Gq/11 in the Striatum. Proceedings of the National Academy of Sciences of the United States of America. 2007. Vol. 104:654–659

            18. Wingler LM, Lefkowitz RJ. Conformational Basis of G Protein-Coupled Receptor Signaling Versatility. Trends in Cell Biology. 2020. Vol. 30:736–747

            19. Li H, Wei WY, Xu HX. Drug discovery is an eternal challenge for the biomedical sciences. Acta Materia Medica. 2022. Vol. 1:1–3

            20. Kelly E, Conibear A, Henderson G. Biased Agonism: Lessons from Studies of Opioid Receptor Agonists. Annual Review of Pharmacology and Toxicology. 2023. Vol. 63:491–515

            21. Weston C, Poyner D, Patel V, Dowell S, Ladds G. Investigating G Protein Signalling Bias at the Glucagon-Like Peptide-1 Receptor in Yeast. British Journal of Pharmacology. 2014. Vol. 171:3651–3665

            22. Jones B. The Therapeutic Potential of GLP-1 Receptor Biased Agonism. British Journal of Pharmacology. 2022. Vol. 179:492–510

            23. Drucker DJ. Mechanisms of Action and Therapeutic Application of Glucagon-Like Peptide-1. Cell Metabolism. 2018. Vol. 27:740–756

            24. Pratley RE, Aroda VR, Lingvay I, Ludemann J, Andreassen C, Navarria A, et al.. Semaglutide Versus Dulaglutide Once Weekly in Patients with Type 2 Diabetes (SUSTAIN 7): A Randomised, Open-Label, Phase 3b Trial. The Lancet Diabetes & Endocrinology. 2018. Vol. 6:275–286

            25. Giorgioni G, Del Bello F, Quaglia W, Botticelli L, Cifani C, Micioni Di Bonaventura E, et al.. Advances in the Development of Nonpeptide Small Molecules Targeting Ghrelin Receptor. Journal of Medicinal Chemistry. 2022. Vol. 65:3098–3118

            26. Herodes M, Anderson LJ, Shober S, Schur EA, Graf SA, Ammer N, et al.. Pilot Clinical Trial of Macimorelin to Assess Safety and Efficacy in Patients with Cancer Cachexia. Journal of Cachexia, Sarcopenia And Muscle. 2023. Vol. 14:835–846

            27. Holubova M, Nagelova V, Lacinova Z, Haluzik M, Sykora D, Moulin A, et al.. Triazole GHS-R1a Antagonists JMV4208 and JMV3002 Attenuate Food Intake, Body Weight, and Adipose Tissue Mass in Mice. Molecular and Cellular Endocrinology. 2014. Vol. 393:120–128

            28. Mende F, Hundahl C, Plouffe B, Skov LJ, Sivertsen B, Madsen AN, et al.. Translating Biased Signaling in the Ghrelin Receptor System into Differential In Vivo Functions. Proceedings of the National Academy of Sciences of the United States of America. 2018. Vol. 115:E10255–E10264

            29. Yue Y, Liu L, Wu LJ, Wu Y, Wang L, Li F, et al.. Structural Insight Into Apelin Receptor-G Protein Stoichiometry. Nature Structural & Molecular Biology. 2022. Vol. 29:688–697

            30. Yang L, Zhu X, Finlay DB, Green H, Glass M, Duffull SB. A Kinetic Model for Positive Allosteric Modulator (PAM)-Antagonists for the Type 1 Cannabinoid (CB(1)) Receptor. British Journal of Pharmacology. 2023. Vol. 180:2661–2676

            31. Van Roy N, Heerwegh S, Husein D, Ruys J, Coremans P. A Diagnostic Conundrum in Bardet-Biedl Syndrome: When Genetic Diagnosis Precedes Clinical Diagnosis. Endocrinology, Diabetes & Metabolism Case Reports. 2023. Vol. 2023:

            32. AlKhelb D, Burke EL, Zvonok A, Iliopoulos-Tsoutsouvas C, Georgiadis MO, Jiang S, et al.. Effects of Cannabinoid Agonists and Antagonists in Male Rats Discriminating the Synthetic Cannabinoid AM2201. European Journal of Pharmacology. 2023. Vol. 960:176168

            33. Kojima M, Hosoda H, Matsuo H, Kangawa K. Ghrelin: Discovery of the Natural Endogenous Ligand for the Growth Hormone Secretagogue Receptor. Trends in Endocrinology and Metabolism. 2001. Vol. 12:118–122

            34. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a Growth-Hormone-Releasing Acylated Peptide from Stomach. Nature. 1999. Vol. 402:656–660

            35. Evron T, Peterson SM, Urs NM, Bai Y, Rochelle LK, Caron MG, et al.. G Protein and β-Arrestin Signaling Bias at the Ghrelin Receptor. Journal of Biological Chemistry. 2014. Vol. 289:33442–33455

            36. Wang Y, Guo S, Zhuang Y, Yun Y, Xu P, He X, et al.. Molecular Recognition of an Acyl-Peptide Hormone and Activation of Ghrelin Receptor. Nature Communications. 2021. Vol. 12:5064

            37. Liu H, Sun D, Myasnikov A, Damian M, Baneres JL, Sun J, et al.. Structural Basis of Human Ghrelin Receptor Signaling by Ghrelin and the Synthetic Agonist Ibutamoren. Nature Communications. 2021. Vol. 12:6410

            38. Lin L, Saha PK, Ma X, Henshaw IO, Shao L, Chang BH, et al.. Ablation of Ghrelin Receptor Reduces Adiposity and Improves Insulin Sensitivity During Aging by Regulating Fat Metabolism in White and Brown Adipose Tissues. Aging Cell. 2011. Vol. 10:996–1010

            39. Lee JH, Lin L, Xu P, Saito K, Wei Q, Meadows AG, et al.. Neuronal Deletion of Ghrelin Receptor Almost Completely Prevents Diet-Induced Obesity. Diabetes. 2016. Vol. 65:2169–2178

            40. Klepac K, Kilić A, Gnad T, Brown LM, Herrmann B, Wilderman A, et al.. The Gq Signalling Pathway Inhibits Brown and Beige Adipose Tissue. Nature Communications. 2016. Vol. 7:10895

            41. Els S, Beck-Sickinger AG, Chollet C. Ghrelin Receptor: High Constitutive Activity and Methods for Developing Inverse Agonists. Methods in Enzymology. 2010. Vol. 485:103–121

            42. Holst B, Cygankiewicz A, Jensen TH, Ankersen M, Schwartz TW. High Constitutive Signaling of the Ghrelin Receptor--Identification of a Potent Inverse Agonist. Journal of Molecular Endocrinology. 2003. Vol. 17:2201–2210

            43. Bhattacharya SK, Andrews K, Beveridge R, Cameron KO, Chen C, Dunn M, et al.. Discovery of PF-5190457, a Potent, Selective, and Orally Bioavailable Ghrelin Receptor Inverse Agonist Clinical Candidate. ACS Medicinal Chemistry Letters. 2014. Vol. 5:474–479

            44. M’Kadmi C, Cabral A, Barrile F, Giribaldi J, Cantel S, Damian M, et al.. N-terminal liver-expressed antimicrobial peptide 2 (LEAP2) region exhibits inverse agonist activity toward the ghrelin receptor. Journal of Medicinal Chemistry. 2019. Vol. 62:965–973

            45. Qin J, Cai Y, Xu Z, Ming Q, Ji SY, Wu C, et al.. Molecular Mechanism of Agonism and Inverse Agonism in Ghrelin Receptor. Nature Communications. 2022. Vol. 13:300

            46. Shiimura Y, Horita S, Hamamoto A, Asada H, Hirata K, Tanaka M, et al.. Structure of an Antagonist-Bound Ghrelin Receptor Reveals Possible Ghrelin Recognition Mode. Nature Communications. 2020. Vol. 11:4160

            47. Ge X, Yang H, Bednarek MA, Galon-Tilleman H, Chen P, Chen M, et al.. LEAP2 is an Endogenous Antagonist of the Ghrelin Receptor. Cell Metabolism. 2018. Vol. 27:461–469.e6

            48. Hagemann CA, Jensen MS, Holm S, Gasbjerg LS, Byberg S, Skov-Jeppesen K, et al.. LEAP2 Reduces Postprandial Glucose Excursions and ad Libitum Food Intake in Healthy Men. Cell Reports Medicine. 2022. Vol. 3:100582

            49. Liu D, Li S. LEAP2: Next Game-Changer of Pharmacotherapy for Overweight and Obesity? Cell Reports Medicine. 2022. Vol. 3:100612

            50. Zhao F, Zhou Q, Cong Z, Hang K, Zou X, Zhang C, et al.. Structural Insights into Multiplexed Pharmacological Actions of Tirzepatide and Peptide 20 at the GIP, GLP-1 or Glucagon Receptors. Nature Communications. 2022. Vol. 13:1057

            51. Graaf C, Donnelly D, Wootten D, Lau J, Sexton PM, Miller LJ, et al.. Glucagon-Like Peptide-1 and Its Class B G Protein-Coupled Receptors: A Long March to Therapeutic Successes. Pharmacological Reviews. 2016. Vol. 68:954–1013

            52. Aroda VR, Rosenstock J, Terauchi Y, Altuntas Y, Lalic NM, Morales Villegas EC, et al.. PIONEER 1: Randomized Clinical Trial of the Efficacy and Safety of Oral Semaglutide Monotherapy in Comparison With Placebo in Patients With Type 2 Diabetes. Diabetes Care. 2019. Vol. 42:1724–1732

            53. Wang P, Hill TA, Mitchell J, Fitzsimmons RL, Xu W, Loh Z, et al.. Modifying a Hydroxyl Patch in Glucagon-like Peptide 1 Produces Biased Agonists with Unique Signaling Profiles. Journal of Medicinal Chemistry. 2022. Vol. 65:11759–11775

            54. Malik F, Li Z. Non-Peptide Agonists and Positive Allosteric Modulators of Glucagon-Like Peptide-1 Receptors: Alternative Approaches for Treatment of Type 2 Diabetes. British Journal of Pharmacology. 2022. Vol. 179:511–525

            55. Jepsen MM, Christensen MB. Emerging Glucagon-Like Peptide 1 Receptor Agonists for the Treatment of Obesity. Expert Opinion on Emerging Drugs. 2021. Vol. 26:231–243

            56. Zhao P, Liang YL, Belousoff MJ, Deganutti G, Fletcher MM, Willard FS, et al.. Activation of the GLP-1 Receptor by a Non-Peptidic Agonist. Nature. 2020. Vol. 577:432–436

            57. Liang YL, Khoshouei M, Glukhova A, Furness SGB, Zhao P, Clydesdale L, et al.. Phase-Plate Cryo-EM Structure of a Biased Agonist-Bound Human GLP-1 Receptor-Gs Complex. Nature. 2018. Vol. 555:121–125

            58. Zhang Y, Sun B, Feng D, Hu H, Chu M, Qu Q, et al.. Cryo-EM Structure of the Activated GLP-1 Receptor in Complex with a G Protein. Nature. 2017. Vol. 546:248–253

            59. Lam S. American Diabetes Association - 77th Scientific Sessions (June 9-13, 2017 - San Diego, California, USA). Drugs of today (Barcelona, Spain: 1998). 2017. Vol. 53:405–413

            60. Zaimia N, Obeid J, Varrault A, Sabatier J, Broca C, Gilon P, et al.. GLP-1 and GIP Receptors Signal Through Distinct Beta-Arrestin 2-Dependent Pathways to Regulate Pancreatic Beta Cell Function. Cell Reports. 2023. Vol. 42:113326

            61. Alexander RA, Lot I, Saha K, Abadie G, Lambert M, Decosta E, et al.. Beta-Arrestins Operate an On/Off Control Switch for Focal Adhesion Kinase Activity. Cellular and Molecular Life Sciences. 2020. Vol. 77:5259–5279

            62. Heimburger SM, Bergmann NC, Augustin R, Gasbjerg LS, Christensen MB, Knop FK. Glucose-Dependent Insulinotropic Polypeptide (GIP) and Cardiovascular Disease. Peptides. 2020. Vol. 125:170174

            63. Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular Actions and Clinical Outcomes With Glucagon-Like Peptide-1 Receptor Agonists and Dipeptidyl Peptidase-4 Inhibitors. Circulation. 2017. Vol. 136:849–870

            64. Lafferty RA, Flatt PR, Irwin N. GLP-1/GIP Analogs: Potential Impact in the Landscape of Obesity Pharmacotherapy. Expert Opinion on Pharmacotherapy. 2023. Vol. 24:587–597

            65. Frias JP, Nauck MA, Van J, Benson C, Bray R, Cui X, et al.. Efficacy and Tolerability of Tirzepatide, a Dual Glucose-Dependent Insulinotropic Peptide and Glucagon-Like Peptide-1 Receptor Agonist in Patients with Type 2 Diabetes: A 12-Week, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate Different Dose-Escalation Regimens. Diabetes, Obesity and Metabolism. 2020. Vol. 22:938–946

            66. Petersen KM, Bogevig S, Holst JJ, Knop FK, Christensen MB. Hemodynamic Effects of Glucagon: A Literature Review. The Journal of Clinical Endocrinology and Metabolism. 2018. Vol. 103:1804–1812

            67. Jastreboff AM, Kaplan LM, Frias JP, Wu Q, Du Y, Gurbuz S, et al.. Triple-Hormone-Receptor Agonist Retatrutide for Obesity - A Phase 2 Trial. The New England Journal of Medicine. 2023. Vol. 389:514–526

            68. Moran TH, Smith GP, Hostetler AM, McHugh PR. Transport of Cholecystokinin (CCK) Binding Sites in Subdiaphragmatic Vagal Branches. Brain Research. 1987. Vol. 415:149–152

            69. Weatherford SC, Chiruzzo FY, Laughton WB. Satiety Induced by Endogenous and Exogenous Cholecystokinin is Mediated by CCK-A Receptors in Mice. American Journal of Physiology. 1992. Vol. 262:R574–R578

            70. Miller LJ, Desai AJ. Metabolic Actions of the Type 1 Cholecystokinin Receptor: Its Potential as a Therapeutic Target. Trends in Endocrinology and Metabolism. 2016. Vol. 27:609–619

            71. Liu Q, Yang D, Zhuang Y, Croll TI, Cai X, Dai A, et al.. Ligand Recognition and G-Protein Coupling Selectivity of Cholecystokinin A Receptor. Nature Chemical Biology. 2021. Vol. 17:1238–1244

            72. Takeda Y, Hoshino M, Yanaihara N, Yanaihara C, Isobe J, Sugiura N, et al.. Comparison of CCK-8 Receptors in the Pancreas and Brain of Rats using CCK-8 Analogues. The Japanese Journal of Pharmacology. 1989. Vol. 49:471–481

            73. von Schrenck T, Müller K, Schulze C, Mirau S, Raedler A, Greten H. N Alpha-Carboxyacyl Analogues of CCK with a Substituted Gly: Interaction with Pancreatic and Gallbladder CCK Receptors. Peptides. 1993. Vol. 14:1309–1315

            74. Sensfuss U, Kruse T, Skyggebjerg RB, Uldam HK, Vestergaard B, Huus K, et al.. Structure-Activity Relationships and Characterization of Highly Selective, Long-Acting, Peptide-Based Cholecystokinin 1 Receptor Agonists. Journal of Medicinal Chemistry. 2019. Vol. 62:1407–1419

            75. Christoffersen B, Skyggebjerg RB, Bugge A, Kirk RK, Vestergaard B, Uldam HK, et al.. Long-Acting CCK Analogue NN9056 Lowers Food Intake and Body Weight in Obese Göttingen Minipigs. International Journal of Obesity (Lond). 2020. Vol. 44:447–456

            76. Cheng L, Shao Z. Lighting Up Cholecystokinin. Nature Chemical Biology. 2021. Vol. 17:1213–1214

            77. Dumoulin V, Dakka T, Plaisancie P, Chayvialle JA, Cuber JC. Regulation of Glucagon-Like Peptide-1-(7-36) Amide, Peptide YY, and Neurotensin Secretion by Neurotransmitters and Gut Hormones in the Isolated Vascularly Perfused Rat Ileum. Endocrinology. 1995. Vol. 136:5182–5188

            78. Stadlbauer U, Woods SC, Langhans W, Meyer U. PYY3-36: Beyond Food Intake. Frontiers in Neuroendocrinology. 2015. Vol. 38:1–11

            79. Poulsen C, Pedersen M, Wahlund PO, Sjölander A, Thomsen JK, Conde-Frieboes KW, et al.. Rational Development of Stable PYY(3-36) Peptide Y(2) Receptor Agonists. Pharmaceutical Research. 2021. Vol. 38:1369–1385

            80. Tang T, Tan Q, Han S, Diemar A, Löbner K, Wang H, et al.. Receptor-Specific Recognition of NPY Peptides Revealed by Structures of NPY Receptors. Science Advances. 2022. Vol. 8:eabm1232

            81. Kang H, Park C, Choi YK, Bae J, Kwon S, Kim J, et al.. Structural Basis for Y2 Receptor-Mediated Neuropeptide Y and Peptide YY Signaling. Structure. 2023. Vol. 31:44–57.e6

            82. Lin S, Boey D, Herzog H. NPY and Y Receptors: Lessons from Transgenic and Knockout Models. Neuropeptides. 2004. Vol. 38:189–200

            83. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, et al.. Gut Hormone PYY(3-36) Physiologically Inhibits Food Intake. Nature. 2002. Vol. 418:650–654

            84. Castan-Laurell I, Masri B, Valet P. The Apelin/APJ System as a Therapeutic Target in Metabolic Diseases. Expert Opinion on Therapeutic Targets. 2019. Vol. 23:215–225

            85. Pitkin SL, Maguire JJ, Bonner TI, Davenport AP. International Union of Basic and Clinical Pharmacology. LXXIV. Apelin Receptor Nomenclature, Distribution, Pharmacology, and Function. Pharmacological Reviews. 2010. Vol. 62:331–342

            86. Ma Y, Yue Y, Ma Y, Zhang Q, Zhou Q, Song Y, et al.. Structural Basis for Apelin Control of the Human Apelin Receptor. Structure. 2017. Vol. 25:858–866.e4

            87. Goidescu CM, Vida-Simiti LA. The Apelin-APJ System in the Evolution of Heart Failure. Clujul Medical. 2015. Vol. 88:3–8

            88. Krashes MJ, Lowell BB, Garfield AS. Melanocortin-4 Receptor-Regulated Energy Homeostasis. Nat Neurosci. 2016. Vol. 19:206–219

            89. Ste Marie L, Miura GI, Marsh DJ, Yagaloff K, Palmiter RD. A Metabolic Defect Promotes Obesity in Mice Lacking Melanocortin-4 Receptors. Proceedings of the National Academy of Sciences of the United States of America. 2000. Vol. 97:12339–12344

            90. Ersoy BA, Pardo L, Zhang S, Thompson DA, Millhauser G, Govaerts C, et al.. Mechanism of N-Terminal Modulation of Activity at the Melanocortin-4 Receptor GPCR. Nature Chemical Biology. 2012. Vol. 8:725–730

            91. Kühnen P, Krude H, Biebermann H. Melanocortin-4 Receptor Signalling: Importance for Weight Regulation and Obesity Treatment. Trends in Molecular Medicine. 2019. Vol. 25:136–148

            92. Kumar KG, Sutton GM, Dong JZ, Roubert P, Plas P, Halem HA, et al.. Analysis of the Therapeutic Functions of Novel Melanocortin Receptor Agonists in MC3R- and MC4R-Deficient C57BL/6J Mice. Peptides. 2009. Vol. 30:1892–1900

            93. Heyder NA, Kleinau G, Speck D, Schmidt A, Paisdzior S, Szczepek M, et al.. Structures of Active Melanocortin-4 Receptor-Gs-Protein Complexes with NDP-α-MSH and Setmelanotide. Cell Research. 2021. Vol. 31:1176–1189

            94. Israeli H, Degtjarik O, Fierro F, Chunilal V, Gill AK, Roth NJ, et al.. Structure Reveals the Activation Mechanism of the MC4 Receptor to Initiate Satiation Signaling. Science. 2021. Vol. 372:808–814

            95. Zhang H, Chen LN, Yang D, Mao C, Shen Q, Feng W, et al.. Structural Insights into Ligand Recognition and Activation of the Melanocortin-4 Receptor. Cell Research. 2021. Vol. 31:1163–1175

            96. Tao YX. The Melanocortin-4 Receptor: Physiology, Pharmacology, and Pathophysiology. Endocrine Reviews. 2010. Vol. 31:506–543

            97. Liu Z, Hruby VJ. MC4R Biased Signalling and the Conformational Basis of Biological Function Selections. Journal of Cellular and Molecular Medicine. 2022. Vol. 26:4125–4136

            98. Shao Z, Yin J, Chapman K, Grzemska M, Clark L, Wang J, et al.. High-Resolution Crystal Structure of the Human CB1 Cannabinoid Receptor. Nature. 2016. Vol. 540:602–606

            99. Li X, Shen L, Hua T, Liu ZJ. Structural and Functional Insights into Cannabinoid Receptors. Trends in Pharmacological Sciences. 2020. Vol. 41:665–677

            100. Shao Z, Yan W, Chapman K, Ramesh K, Ferrell AJ, Yin J, et al.. Structure of an Allosteric Modulator Bound to the CB1 Cannabinoid Receptor. Nature Chemical Biology. 2019. Vol. 15:1199–1205

            101. Janero DR, Makriyannis A. Cannabinoid Receptor Antagonists: Pharmacological Opportunities, Clinical Experience, and Translational Prognosis. Expert Opinion on Emerging Drugs. 2009. Vol. 14:43–65

            102. Adams MH, Lammon CB, Stover LM. Responding to Tricyclic Antidepressant Overdose. Dimensions of Critical Care Nursing. 1998. Vol. 17:67–74

            103. Fay JF, Farrens DL. A Key Agonist-Induced Conformational Change in the Cannabinoid Receptor CB1 is Blocked by the Allosteric Ligand Org 27569. Journal of Biological Chemistry. 2012. Vol. 287:33873–33882

            104. Fay JF, Farrens DL. Structural Dynamics and Energetics Underlying Allosteric Inactivation of the Cannabinoid Receptor CB1. Proceedings of the National Academy of Sciences of the United States of America. 2015. Vol. 112:8469–8474

            105. Yang X, Wang X, Xu Z, Wu C, Zhou Y, Wang Y, et al.. Molecular Mechanism of Allosteric Modulation for the Cannabinoid Receptor CB1. Nature Chemical Biology. 2022. Vol. 18:831–840

            106. Krishna Kumar K, Robertson MJ, Thadhani E, Wang H, Suomivuori CM, Powers AS, et al.. Structural Basis for Activation of CB1 by an Endocannabinoid Analog. Nature Communications. 2023. Vol. 14:2672

            107. Miranda K, Becker W, Busbee PB, Dopkins N, Abdulla OA, Zhong Y, et al.. Yin and Yang of Cannabinoid CB1 Receptor: CB1 Deletion in Immune Cells Causes Exacerbation While Deletion in Non-Immune Cells Attenuates Obesity. iScience. 2022. Vol. 25:104994

            108. Dvorácskó S, Herrerias A, Oliverio A, Bhattacharjee P, Pommerolle L, Liu Z, et al.. Cannabinoformins: Designing Biguanide-Embedded, Orally Available, Peripherally Selective Cannabinoid-1 Receptor Antagonists for Metabolic Syndrome Disorders. Journal of Medicinal Chemistry. 2023. Vol. 66:11985–12004

            109. Gentry PR, Sexton PM, Christopoulos A. Novel Allosteric Modulators of G Protein-Coupled Receptors. Journal of Biological Chemistry. 2015. Vol. 290:19478–19488

            110. Guo S, Zhao T, Yun Y, Xie X. Recent Progress in Assays for GPCR Drug Discovery. American Journal of Physiology Cell Physiology. 2022. Vol. 323:C583–C594

            111. Hauser AS, Avet C, Normand C, Mancini A, Inoue A, Bouvier M, et al.. Common Coupling Map Advances GPCR-G Protein Selectivity. Elife. 2022. Vol. 11:e74107

            112. Avet C, Mancini A, Breton B, Le Gouill C, Hauser AS, Normand C, et al.. Effector Membrane Translocation Biosensors Reveal G Protein And Betaarrestin Coupling Profiles of 100 Therapeutically Relevant GPCRs. Elife. 2022. Vol. 11:e74101

            113. Andersen A, Lund A, Knop FK, Vilsboll T. Glucagon-Like Peptide 1 in Health And Disease. Nature Reviews Endocrinology. 2018. Vol. 14:390–403

            114. Wright SC, Motso A, Koutsilieri S, Beusch CM, Sabatier P, Berghella A, et al.. GLP-1R Signaling Neighborhoods Associate with the Susceptibility to Adverse Drug Reactions of Incretin Mimetics. Nature Communications. 2023. Vol. 14:6243

            115. Leon N, LaCoursiere R, Yarosh D, Patel RS. Lixisenatide (Adlyxin): A Once-Daily Incretin Mimetic Injection for Type-2 Diabetes. Pharmacy and Therapeutics. 2017. Vol. 42:676–711

            116. Saxena AR, Gorman DN, Esquejo RM, Bergman A, Chidsey K, Buckeridge C, et al.. Danuglipron (PF-06882961) in type 2 Diabetes: a Randomized, Placebo-Controlled, Multiple Ascending-Dose Phase 1 Trial. Nat Med. 2021. Vol. 27:1079–1087

            117. Ferraino KE, Cora N, Pollard CM, Sizova A, Maning J, Lymperopoulos A. Adrenal Angiotensin II Type 1 Receptor Biased Signaling: The Case for “Biased” Inverse Agonism for Effective Aldosterone Suppression. Cell Signal. 2021. Vol. 82:109967

            118. Lymperopoulos A, Borges JI, Carbone AM, Cora N, Sizova A. Cardiovascular Angiotensin II Type 1 Receptor Biased Signaling: Focus on Non-Gq-, Non-Betaarrestin-Dependent Signaling. Pharmacological Research. 2021. Vol. 174:105943

            119. Zhang Y, Zhao Y, Yang L, Cai Y, Shangguan X, Huang R. Factors associated with pharmacists’ knowledge regarding high-alert medications: a convenience sample survey in China. Acta Materia Medica. 2022. Jul 20;

            120. Zhang M, Otsuki K, Li W. Molecular networking as a natural products discovery strategy. Acta Materia Medica. 2023. Vol. 2:126–141. [Cross Ref]

            121. Sloop KW, Emmerson PJ, Statnick MA, Willard FS. The Current State of GPCR-Based Drug Discovery to Treat Metabolic Disease. British Journal of Pharmacology. 2018. Vol. 175:4060–4071

            122. Riddy DM, Delerive P, Summers RJ, Sexton PM, Langmead CJ. G Protein-Coupled Receptors Targeting Insulin Resistance, Obesity, and Type 2 Diabetes Mellitus. Pharmacological Reviews. 2018. Vol. 70:39–67

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            02 March 2024
            : 3
            : 1
            : 31-45
            Affiliations
            [a ]State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
            [b ]State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
            [c ]Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
            [d ]Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
            Author notes

            1These authors contributed equally.

            Article
            10.15212/AMM-2023-0041
            c98d4f3f-2248-4a2e-afd7-66298dfc428e
            Copyright © 2024 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 26 October 2023
            : 17 January 2024
            : 04 February 2024
            Page count
            Figures: 7, Tables: 1, References: 122, Pages: 15
            Funding
            Funded by: National Natural Science Foundation of China
            Award ID: 82204479
            Funded by: National Natural Science Foundation of China
            Award ID: 82073715
            Funded by: Guangdong Basic and Applied Basic Research Foundation
            Award ID: 2021A1515110381
            Funded by: Fundamental Research Funds for the Central Public Welfare Research Institutes
            Award ID: ZZ14-YQ-056
            Funded by: Research Fund of University of Macau
            Award ID: MYRG2020-00091-ICMS
            Funded by: Research Fund of University of Macau
            Award ID: MYRG2022-00177-ICMS
            Funded by: Science and Technology Development Fund, Macao SAR
            Award ID: FDCT 0064/2021/AGJ
            Funded by: Science and Technology Development Fund, Macao SAR
            Award ID: 005/2023/SKL
            Financial support by the National Natural Science Foundation of China (82204479 and 82073715), Guangdong Basic and Applied Basic Research Foundation (2021A1515110381), the Fundamental Research Funds for the Central Public Welfare Research Institutes (ZZ14-YQ-056), the Research Fund of University of Macau (MYRG2020-00091-ICMS and MYRG2022-00177-ICMS), and the Science and Technology Development Fund, Macao SAR (FDCT 0064/2021/AGJ and 005/2023/SKL) are gratefully acknowledged.
            Categories
            Review Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            drug target,obesity,GPCR,energy homeostasis,biased signaling

            Comments

            Comment on this article