Abstract
DREADDs (Designer Receptors Exclusively Activated by a Designer Drug) are designer G protein-coupled receptors (GPCRs) that are widely used in the neuroscience field to modulate neuronal activity. In this review, we will focus on DREADD studies carried out with genetically engineered mice aimed at elucidating signaling pathways important for maintaining proper glucose and energy homeostasis. The availability of muscarinic receptor-based DREADDs endowed with selectivity for one of the four major classes of heterotrimeric G proteins (Gs, Gi, Gq, and G12) has been instrumental in dissecting the physiological and pathophysiological roles of distinct G protein signaling pathways in metabolically important cell types. The novel insights gained from this work should inform the development of novel classes of drugs useful for the treatment of several metabolic disorders including type 2 diabetes and obesity.
Keywords: G protein-coupled receptors, G proteins, type 2 diabetes, obesity, mutant mouse models, DREADD technology
G protein-coupled receptors (GPCRs) are cell surface receptors that are activated by extracellular ligands such as neurotransmitters, hormones, paracrine factors, and many other stimuli (Pierce et al., 2002). In humans, the GPCR superfamily consists of more than 800 distinct members (Sriram and Insel, 2018). Importantly, GPCRs are the target of a very large number of therapeutic agents (Sriram and Insel, 2018). Each individual GPCR interacts with one or more of the four major classes of heterotrimeric G proteins (Gs, Gi, Gq, and G12) which are named after the structural and functional properties of their α subunits (Wettschureck and Offermanns, 2005). The α subunit of heterotrimeric G proteins is tightly bound to a βγ complex in the inactive state of the protein. Receptor-activated free α subunits stimulate or inhibit distinct intracellular signaling pathways, but free βγ complexes can also modulate the activity of certain ion channels and effector proteins (Wettschureck and Offermanns, 2005).
Gene expression studies have shown that virtually each body cell expresses dozens of GPCRs that can modulate cellular functions by activating or inhibiting distinct intracellular signaling cascades (Regard et al., 2008). With almost no exception, each specific GPCR is expressed in multiple tissues and cell types (Regard et al., 2008). Moreover, highly selective ligands (agonists or antagonists) are not available for a very large number of GPCRs. For these reasons, identifying the in vivo metabolic roles of a particular GPCR expressed by a specific cell type represents a very difficult task.
General features of DREADDs (Designer Receptors Exclusively Activated by a Designer Drug)
In a pioneering study, Armbruster et al. (Armbruster et al., 2007) demonstrated that introduction of a pair of point mutations into the transmembrane core of different muscarinic acetylcholine (ACh) receptor subtype (M1-M4) rendered the resulting mutant receptors insensitive to activation by ACh or other endogenous ligands. However, by taking advantage of a yeast generic screen, the Roth lab identified a synthetic compound called clozapine-N-oxide (CNO) (Fig. 1) that was able to activate these mutant receptors with high potency and efficacy. Importantly, when used in the proper concentration/dose range, CNO is pharmacologically inert. For these reasons, Armbruster et al. (Armbruster et al., 2007) referred to these mutant muscarinic receptors as DREADDs (Designer Receptors Exclusively Activated by a Designer Drug). Two of these DREADDs, GqD and GiD (Fig. 2), are now widely used in the neuroscience field to activate or silence distinct sets of neurons, respectively (Rogan and Roth, 2011; Roth, 2016). However, it should be noted that treatment of mice with high doses of CNO (e.g. ≥10 mg/kg i.p) may cause off-target effects (Martinez et al., 2019).
FIGURE 1.
Chemical structures of CNO, clozapine, C21, and DCZ. All four agents can activate muscarinic receptor-based DREADDs with high potency and efficacy. However, clozapine acts on many other high-affinity targets in the CNS. Several studies have shown that CNO can be metabolized to clozapine. C21 and DCZ are CNO-derived, novel DREADD agonists that are not converted to clozapine in vivo (Chen et al., 2015; Nagai et al., 2020; Thompson et al., 2018). See text for details.
FIGURE 2.
Coupling properties of muscarinic receptor-based DREADDs. The structural features of the various DREADDs are described in the main body of the text. The four DREADDs shown here harbor the same two point mutations in the transmembrane core (Y->C3.33 and A->G5.46; Ballesteros-Weinstein GPCR nomenclature) (Armbruster et al., 2007; Guettier et al., 2009; Inoue et al., 2019). While acetylcholine, the endogenous muscarinic receptor agonist, shows little or no activity at these DREADDs, they can be activated by CNO with high potency and efficacy. The different G proteins are named after the α-subunit contained within the G protein heterotrimer. Activated Gα subunits regulate the activity of distinct intracellular signaling pathways, but free βγ complexes, which are released after G protein activation, can also modulate cellular functions. Gα proteins are grouped into four major subfamilies (Gs: αs, αolf; Gi: αi1, αi2, αi3, αo, etc.; Gq: αq, α11; G12: α12, α13). Abbreviations: Epac, exchange protein activated by cAMP; GIRK, G-protein-regulated inward-rectifier potassium channel; LARG, Leukemia-Associated RhoGEF; PLC, phospholipase C; PKA, protein kinase A; PKC, protein kinase C; RhoGEF, Rho guanine nucleotide exchange factor; ROCK; Rho-associated coiled-coil-containing protein kinase; VDCC, voltage-dependent Ca2+-channel.
The general concept underlying the development of DREADD technology dates back to earlier studies reporting the generation of so-called RASSLs (Receptors Activated Solely by Synthetic Ligands) (Conklin et al., 2008; Coward et al., 1998). However, the in vivo use of these first-generation designer GPCRs is complicated by the fact that RASSLs can still be activated by endogenous ligands and show strong constitutive activity when expressed in mutant mice (Conklin et al., 2008; Coward et al., 1998).
The M3 muscarinic receptor-derived DREADD, known as hM3Dq or simply GqD, is coupled to Gq-type G proteins (Armbruster et al., 2007; Wess et al., 2007) (Fig. 2). As a result, CNO binding to GqD leads to the activation of different isoforms of phospholipase Cβ, resulting in the breakdown of phosphatidylinositol and the generation of the second messengers, inositol 1,4,5- trisphosphate (IP3) and diacylglycerol (DAG). IP3 releases Ca2+ from intracellular stores, leading to elevated intracellular Ca2+ levels which regulate the activity of many intracellular signaling pathways. DAG activates different isoforms of PKC which also modulate the function of many important signaling proteins including ERK (Fig. 2). The GqD-mediated increase in intracellular Ca2+ levels is considered a key step that causes neuronal excitation (Rogan and Roth, 2011; Roth, 2016). It remains to be elucidated whether ligand activation of GqD leads to the activation of all G proteins of the Gq family (αq, α11, α14, and α15/16).
The second DREADD that is now extensively used in neuroscience research is derived from the M4 muscarinic receptor which is selectively linked to G proteins of the Gi family (Armbruster et al., 2007; Wess et al., 2007). CNO-induced activation of this DREADD, referred to as hM4Di or simply as GiD (Fig. 2), leads to the inhibition of adenylyl cyclase and the opening of G protein-coupled inwardly-rectifying potassium channels (GIRK) channels (Armbruster et al., 2007; Wess et al., 2007). This latter effect is thought to be responsible for the ability of activated GiD receptors to induce neuronal silencing. It needs to be explored whether CNO-mediated activation of GiD causes the activation of all members of the Gi/o family (αi1, αi2, αi3, αo, αz, αt, and αgust).
Subsequently, Guettier et al. (Guettier et al., 2009) reported the generation and pharmacologic properties of a Gs-coupled DREADD (GsD) (Fig. 2). The GsD construct was created by replacing the second and third intracellular loops of GqD with the corresponding β1-adrenoceptor sequences (Guettier et al., 2009). As expected, CNO treatment of cells expressing the GsD designer receptor resulted in increased cAMP levels (Guettier et al., 2009). In a very recent study, Inoue et al. (Inoue et al., 2019) described the development of the first G12-linked DREADDs (G12D) (Fig. 2). Structurally, these new designer receptors have a GqD backbone in which the third intracellular loops are derived from GPR132 or GPR183. Somewhat surprisingly, these G12 DREADDs do not activate G13 (Inoue et al., 2019). For this reason, these new designer receptors are particularly useful to study the consequences of G12 signaling in vivo.
The generation of mutant mice expressing a particular DREADD in a specific cell type has made it possible to study the in vivo consequences of activating distinct G protein signaling pathways in distinct cell types in vivo (Wess, 2016; Wess et al., 2013). As discussed above, such studies are difficult to perform by using classical pharmacological tools, since the expression of endogenous GPCRs is usually not restricted to a specific cell type (Regard et al., 2008). In this review, we will discuss DREADD studies that have shed light onto metabolic roles of activating specific classes of G proteins in distinct cell types known to be critical for glucose and energy homeostasis. The new insights gained from this research should stimulate the development of novel classes of drugs useful for the treatment of type 2 diabetes (T2D) and related metabolic disorders.
Novel tools useful for in vivo applications of DREADD technology
Novel DREADD ligands derived from CNO
Since the development of the first muscarinic receptor-based DREADDs more than a decade ago, CNO (Fig. 1) has become the agonist of choice for activating this class of designer GPCRs. Guettier et al. (Guettier et al., 2009) showed that acute treatment of mice with CNO (1 mg/kg i.p.) did not lead to a significant conversion of CNO to clozapine (Fig. 1) during a 2 hr observation period (clozapine is a potent psychoactive drug). However, several studies have shown that CNO can be back-transformed to clozapine in many species including humans and monkeys under different experimental conditions (Chang et al., 1998; Gomez et al., 2017; Jann et al., 1994; Manvich et al., 2018; Raper et al., 2017). Interestingly, clozapine is ~10-fold more potent than CNO in activating muscarinic receptor-based DREADDs (Armbruster et al., 2007). However, clozapine, which is in clinical use as an antipsychotic drug, affects many functions of the CNS by binding with high affinity to numerous cellular targets (Yadav et al., 2011). For this reason, DREADD studies involving the use of CNO should ideally always be conducted with four groups of experimental animals (DREADD animals treated with CNO or saline; wild-type (WT)/control animals treated with CNO or saline). A review of past DREADD studies indicates that one or more of these control groups were not consistently included in the experimental design. The use of all four control groups is of particular importance when high doses of CNO are administered. It should also be noted that CNO metabolism after acute vs. chronic CNO administration has not been studied systematically so far.
To prevent possible effects mediated by the conversion of CNO to clozapine, recent efforts (Chen et al., 2015; Thompson et al., 2018) led to the development of a CNO derivative, referred to as compound 21 (C21) (Fig. 1). This agent activates muscarinic receptor-based DREADDs with high efficacy and potency, shows very good bioavailability, and is not metabolized to clozapine. For this reason, C21 can be employed as an alternative to CNO for studies where metabolic conversion of CNO to clozapine is a major cause of concern.
Following systemic administration, the uptake of CNO into the brain has been shown to be relatively slow (Nagai et al., 2020). Interestingly, Nagai et al. (Nagai et al., 2020) recently reported that deschloroclozapine (DCZ) (Fig. 1), a clozapine derivative, represents an extremely potent DREADD agonist that quickly enters the brain and shows high biostability. Studies with mice and monkeys demonstrated that DCZ rapidly (within 10 min after injection) activates centrally expressed DREADDs (GqD or GiD), without detectable off-target effects (Nagai et al., 2020). These data suggest that DCZ represents a highly useful, novel agonist at muscarinic receptor-based DREADDs that is selective, stable, and has a fast onset of action. We recently demonstrated that very low doses of DCZ (e.g. 10 μg/kg i.p.) are also able to efficiently activate DREADDs expressed in peripheral tissues or cell types in mutant mice (J. Wess et al., unpublished data).
Fig. 1 shows that C21 and DCZ are structurally almost identical with clozapine, except for the lack of a chloro atom (C21 and DCZ) and a methyl moiety (C21). These modifications prevent back-metabolism to clozapine and greatly reduce the affinity of clozapine for its many CNS target proteins/receptors, while maintaining high brain penetrance (Chen et al., 2015; Nagai et al., 2020; Thompson et al., 2018).
Non-muscarinic receptor-based DREADDs
During the past decade, additional DREADDs containing different GPCR backbones have also been developed. Several years ago, Vardy et al. (Vardy et al., 2015) described the generation and pharmacological characterization of a κ-opioid-derived DREADD (KORD) that is selectively activated by salvinorin B, a pharmacologically inert compound. However, salvinorin B is able to bind to the κ-opioid receptor with modest affinity (>100 nM) (Vardy et al., 2015). For this reason, salvinorin B should be used at low doses that can activate the KORD designer receptor but do not cause any undesired side effects via interaction with WT k-opioid receptors. Since this new DREADD is activated by a ligand other than CNO, it allows the regulation of neuronal activity and/or behavior in a multiplexed and bidirectional fashion. For example, co-expression of the GqD and KORD designer receptors made it possible to activate (via CNO/GqD) and inhibit (via salvinorin B/KORD) neuronal activity in a sequential fashion (Vardy et al., 2015).
Moreover, Hudson et al. (Hudson et al., 2012) succeeded in generating a DREADD variant of the free fatty acid receptor 2 (FFA2). The WT FFA2 is activated by short chain fatty acids (SCFAs) and couples to both Gq- and Gi-type proteins (Bolognini et al., 2016). The FFA2 DREADD, which shows a similar coupling profile, does no longer respond effectively to SCFAs but can be selectively activated by several other ligands, including sorbic acid (Hudson et al., 2012). Recently, Bolognini et al. (Bolognini et al., 2019) used a FFA2-DREADD knock-in mouse strain to study the physiological roles of FFA2 in the gut and white adipose tissue. Analysis of this novel mutant mouse strain demonstrated that DREADD knock-in mice can serve as highly useful tools to elucidate the physiological functions and therapeutic potential of GPCRs for which highly selective ligands are not available (Bolognini et al., 2019).
Mouse models that allow the expression of DREADDs in a conditional fashion
The recent development of a new series of DREADD mutant mice has made it possible to express various DREADDs (GqD, GiD, and GsD) in specific cell types or tissues in a Cre-dependent fashion. Zhu et al. (Zhu et al., 2016) first generated mutant mouse strains harboring the GqD or GiD sequences under the control of a strong ubiquitous promoter, which is separated from the DREADD sequences by a stop signal that is flanked by loxP sites. Using an analogous approach, Akhmedov et al. (Akhmedov et al., 2017a) developed a mutant mouse strain in which the expression of GsD requires the presence of Cre recombinase. In all these new mouse strains, the DREADD construct (preceded by the promoter and floxed stop sequences) was inserted into the ROSA26 locus. When these mice are crossed with specific Cre driver lines, Cre recombinase removes the stop signal, leading to DREADD expression in Cre-expressing cells. This approach allows the conditional expression of specific DREADDs in a cell type-specific fashion.
DREADDs that display signaling bias
As is the case with most GPCRs, is likely that ligand-activated DREADDs do not only interact with heterotrimeric G proteins but can also recruit β-arrestins (βarr1 and βarr2). This has been clearly demonstrated for the CNO-activated GqD designer receptor (Alvarez-Curto et al., 2011; Hu et al., 2016; Nakajima and Wess, 2012). The two β-arrestins are known to play a key role in receptor desensitization and internalization (Pierce and Lefkowitz, 2001). However, many studies suggest that βarr1 and βarr2 can also act as signaling proteins in their own right (Beaulieu et al., 2009; DeFea, 2011; Gurevich and Gurevich, 2014; Luttrell and Gesty-Palmer, 2010; Rajagopal et al., 2010; Shukla et al., 2011). For this reason, the in vivo consequences of activating a particular DREADD in a specific cell type or tissue may not only depend on activated G proteins but may also involve β-arrestin-dependent signaling. To assess the relative importance of these two signaling branches, mutant mice that selectively lack specific G protein α-subunits or β-arrestins in DREADD-expressing cells would prove highly valuable tools. Accumulating evidence indicates that the use of so-called ‘biased’ GPCR agonists which initiate signaling preferentially through either G proteins or β-arrestins may prove advantageous in a number of pathophysiological conditions (Gurevich and Gurevich, 2019; Kenakin and Christopoulos, 2013; Luttrell et al., 2015; Rajagopal et al., 2010; Shukla et al., 2011; Smith et al., 2018; Wootten et al., 2018). This concept is based on findings that the therapeutic effect of a particular GPCR agonist may arise from activating one signaling pathway, whereas the other signaling branch may mediate undesirable side effects.
The development of two mutationally modified versions of GqD, M3D-Gq and M3D-arr, which display biased coupling properties (Hu et al., 2016; Nakajima and Wess, 2012), may prove helpful to explore to which extent β-arrestins contribute to DREADD-mediated responses in vivo. The ligand-activated M3D-Gq designer receptor promotes signaling via Gq-type G proteins but does cause the recruitment of β-arrestins (Hu et al., 2016). On the other hand, the M3D-arr DREADD shows the opposite activity profile (Hu et al., 2016; Nakajima and Wess, 2012).
Caveats
Chronic CNO treatment of mutant mice expressing GqD in pancreatic β-cells (Jain et al., 2013) or GsD and GiD in adipocytes (Wang et al., 2019; Wang et al., 2020) did not lead to a waning of DREADD-mediated effects over time. However, additional studies are required to confirm whether this is also true for other cell types and other physiological/behavioral responses. It should also be noted DREADD-based experimental strategies mimic drug effects on target GPCRs, but that other experimental approaches (e.g., optogenetics) need to be applied when a more precise temporal control of cellular signaling is required. At present, little is known about the potential impact of DREADD overexpression on the expression of endogenous receptors or signaling molecules. Surprisingly, expression of GiD in sensory C-fibers of mice disrupted endogenous inhibitory GPCR expression and second-messenger coupling, most likely due to CNO-independent signaling of the GiD in dorsal root ganglion (DRG) neurons (Saloman et al., 2016). For this reason, experimental data obtained with systems where DREADDs display constitutive activity need to be interpreted with particular caution.
Metabolically important signaling pathways identified by using DREADD technology
In the following, we will review studies that employed DREADD technology to elucidate the metabolic roles of distinct GPCR signaling pathways operative in metabolically important cell types in regulating whole-body glucose and energy homeostasis. The focus will be on work carried out with three muscarinic receptor-based DREADDs (GqD, GsD, and GiD). The novel information resulting from this research should be of considerable translational usefulness for designing new pharmacological strategies for the treatment of major metabolic disorders, including T2D and obesity.
Pancreatic β- and α-cells
Pancreatic islets are composed of multiple cell types, including β- and α-cells which synthesize and release the hormones insulin and glucagon, respectively. These hormones play essential roles in maintaining euglycemia and modulate many other important metabolic functions. T2D is characterized by impaired β-cell function, leading to insufficient insulin release to overcome peripheral insulin resistance (Hudish et al., 2019; Roden and Shulman, 2019). For this reason, drugs capable of improving impaired β-cell function have great clinical potential for the management of T2D (Hudish et al., 2019; Roden and Shulman, 2019). Different experimental approaches, including RNA-seq studies, have demonstrated that pancreatic β-cells express numerous GPCRs that are linked to different G protein signaling pathways (Ahren, 2009; Amisten et al., 2013; Riddy et al., 2018; Xin et al., 2016).
β-cell-specific GsD mice
To examine the metabolic importance of receptor-mediated activation of Gs in β-cell function and whole-body glucose homeostasis, Guettier et al. (Guettier et al., 2009) generated and analyzed mice that selectively expressed the GsD construct in pancreatic β-cells (β-GsD mice) (Fig. 3a).
FIGURE 3.
Summary of in vivo ‘DREADD studies’ investigating signaling pathways regulating glucose and energy homeostasis. This cartoon summarizes data obtained with mutant mice expressing DREADDs in pancreatic β- and α-cells (a), adipocytes (b), hepatocytes (c), skeletal muscle cells (d), and AgRP (e) and POMC neurons (f) of the arcuate nucleus of the hypothalamus. For simplicity, only some key signaling factors/pathways and metabolic phenotypes are listed. See text for details.
Acute CNO treatment of β-GsD mice led to a pronounced increase in insulin secretion, improved glucose tolerance, and several other beneficial metabolic effects. In β-GsD mice, the GsD receptor also showed some degree of constitutive activity (Guettier et al., 2009). Interestingly, expression of the GsD receptor in mouse hepatocytes in vivo also resulted in a significant degree of CNO-independent signaling (Akhmedov et al., 2017b). However, when expressed in other cell types, including adipocytes (Wang et al., 2019) and AgRP neurons (Nakajima et al., 2016), the GsD receptor did not exhibit constitutive activity. It is likely that actual DREADD expression levels, together with cell type-specific factors, are responsible for these seemingly discrepant findings.
The glucagon-like peptide-1(GLP-1) receptor is a Gs-coupled receptor (like GsD) that is endogenously expressed at relatively high levels by pancreatic β-cells (Müller et al., 2019). As a result, GLP-1 receptor agonists, such as exenatide or semaglutide, have emerged as a very promising class of novel antidiabetic agents (Chia and Egan, 2020; Drucker, 2018). The same is true for inhibitors of dipeptidyl peptidase 4 (DPP4 inhibitors or gliptins; e.g. sitagliptin) which interfere with the degradation of GLP-1, glucose-dependent insulinotropic polypeptide (GIP), and several other bioactive peptides (Drucker and Nauck, 2006; Müller et al., 2019).
While stimulation of β-cell Gs signaling causes multiple beneficial metabolic effects, mice selectively lacking Gαs in pancreatic β-cells (β-Gs-KO mice) showed profound metabolic deficits (Xie et al., 2007). β-Gs-KO mice displayed severe hypoinsulinemia, hyperglycemia, and glucose intolerance. Moreover, the lack of β-cell Gαs led to greatly reduced islet insulin content, associated with a marked reduction in β-cell mass (Xie et al., 2007). These findings further highlight the importance of β-cell Gs signaling in β-cell function and whole-body glucose homeostasis.
β-cell-specific GqD mice
The development of a mutant mouse strain selectively expressing the GqD construct in pancreatic β-cells (β-GqD mice) made it possible to study the metabolic outcome of selectively stimulating Gq signaling in β-cells in vivo (Guettier et al., 2009; Jain et al., 2013) (Fig. 3a). Acute CNO treatment of β-GqD mice strongly promoted insulin secretion and led to a dramatic improvement in glucose tolerance (Guettier et al., 2009). Chronic CNO treatment of β-GqD mice caused a significant increase in pancreatic insulin content and β-cell mass, associated with the up-regulation of many genes critical for β-cell function and proliferation (Jain et al., 2013). In addition, chronically enhanced β-cell Gq signaling greatly improved glucose homeostasis in mouse models of diabetes, most likely due to Gq-mediated activation of insulin receptor substrate 2 (IRS2) signaling (Jain et al., 2013). In agreement with these findings, the Gq-coupled free fatty acid receptor 1 (FFAR1), which is expressed at relatively high levels by pancreatic β-cells (Tomita et al., 2014), is considered a promising target for the development of novel antidiabetic drugs (Li et al., 2020).
Interestingly, a recent study (Makhmutova et al., 2020) analyzing mice expressing GqD selectively in β-cells demonstrated that β-cells can communicate with vagal sensory neurons, most likely using serotonin as a signaling molecule. Since serotonin is co-released with insulin from β-cells, it may act as a reporter of the secretory state of β-cells via vagal afferent nerves.
As expected, mutant mice that lacked both Gαq and Gα11 selectively in pancreatic β-cells (β-Gq/11-KO mice) displayed impaired glucose tolerance and insulin secretion (Sassmann et al., 2010). Somewhat surprisingly, the ability of glucose to stimulate insulin release was also reduced in these mutant mice. The authors demonstrated that this deficit was caused by the loss of β cell-autonomous potentiation of insulin secretion through factors co-secreted with insulin (Sassmann et al., 2010). Such factors include uridine diphosphate which acts on Gq-coupled P2Y6 receptors and extracellular calcium that stimulates β-cell calcium-sensing receptors. These observations further highlight the metabolic importance of Gq-dependent signaling pathways operative in pancreatic β-cells.
α-cell-specific GiD mice
Pancreatic α-cells store and release glucagon which functions to maintain euglycemia and to counteract the actions of insulin on the liver and other tissues (Ahren, 2015; Quesada et al., 2008; Unger and Cherrington, 2012). It is well documented that α-cell dysfunction contributes to deficits in glucose homeostasis in diabetic individuals (Ahren, 2015; Lee et al., 2016; Quesada et al., 2008; Wendt and Eliasson, 2020). Like pancreatic β-cells, α-cells express many GPCRs that differ in their G protein coupling properties (Ahren, 2009; Amisten et al., 2013; Riddy et al., 2018; Xin et al., 2016).
To investigate the in vivo metabolic consequences of simulating Gi signaling in α-cells, Zhu et al. (Zhu et al., 2019) analyzed mutant mice that express the GiD construct selectively in α-cells (⍺-GiD mice) (Fig. 3a). Interestingly, acute CNO treatment of ⍺-GiD mice led to profound reductions in both plasma glucagon and insulin levels. CNO-treated ⍺-GiD mice also showed impairments in glucose-stimulated insulin secretion (GSIS) and glucose tolerance, supporting the concept that glucagon released from pancreatic α-cells can promote insulin secretion (Zhu et al., 2019).
In agreement with two recent studies (Capozzi et al., 2019a; Svendsen et al., 2018), Zhu et al. (Zhu et al., 2019) also showed that intraislet glucagon primarily acts on β-cell GLP-1 receptors to stimulate insulin secretion in a paracrine fashion. Since glucagon elevates blood glucose levels by stimulating hepatic glucose production (HGP) under fasting conditions (Ali and Drucker, 2009), the ability of intraislet glucagon to promote insulin release in the presence of glucose suggests that glucagon has opposing physiological functions depending on the nutritional state of the organism (Capozzi et al., 2019b). In the light of these new findings, the development of therapeutic approaches aimed at suppressing glucagon signaling to treat hyperglycemia needs to be reassessed.
L-cells
L-cell-specific GqD mice
L-cells are enteroendocrine cells that are primarily found in the ileum and large intestine (colon). Following food intake, L-cells secrete the hormones GLP-1 and peptide YY (PYY), as well as several other bioactive peptides (Gribble and Reimann, 2016; Spreckley and Murphy, 2015). GLP-1 exerts multiple biological actions, including stimulation of insulin release from pancreatic β-cells, delayed gastric emptying, and reduced appetite. PYY also causes anorectic effects and reduces gastrointestinal motility. L-cell function is regulated by the activity of a considerable number of GPCRs (Gribble and Reimann, 2016).
Recently, Lewis et al. (Lewis et al., 2020) generated a mutant mouse line that expressed the GqD designer receptor in an inducible fashion selectively in mouse distal colonic L cells. Interestingly, CNO treatment of these mutant mice led to a significant increase in plasma GLP-1 and PYY levels, improved glucose tolerance, and decreased food intake. These data suggest that agonists that can act on Gq-coupled receptors expressed by colonic L-cells may prove useful to improve impaired glucose homeostasis under pathophysiological conditions. Such receptors may include FFAR1 and the AT1a angiotensin II and V1b vasopressin receptor subtypes (Billing et al., 2018).
Adipocytes
Adipocytes play a key role in regulating glucose and energy homeostasis under physiological and pathophysiological conditions including T2D (Guilherme et al., 2008). In obesity, adipose tissue is infiltrated by macrophages, resulting in the secretion of inflammatory adipokines and other factors that result in impaired glucose homeostasis (Gonzalez-Muniesa et al., 2017; Guh et al., 2009; Kusminski et al., 2016; Saltiel and Olefsky, 2017). It is well documented that adipocytes express many GPCRs endowed with different coupling properties (Amisten et al., 2015; Regard et al., 2008). While some of these receptors have been studied in considerable detail (e.g. β-adrenergic receptors), the metabolic functions of most of these GPCRs have not been studied systematically.
Adipocyte-specific GsD mice
To explore the in vivo metabolic consequences of selectively activating Gs signaling in adipocytes, two groups (Caron et al., 2019; Wang et al., 2019) recently studied mutant mice that expressed the GsD construct selectively in adipocytes (adipo-GsD mice) (Fig. 3b). Both studies demonstrated that acute CNO injection of lean or obese adipo-GsD mice resulted in increased lipolysis, elevated plasma free fatty acid (FFA) and plasma insulin levels, decreased blood glucose levels, and improved glucose tolerance (Caron et al., 2019; Wang et al., 2019). The beneficial effects on glucose homeostasis were most likely caused by increased insulin release caused by elevated plasma FFA levels. Consistent with this notion, it has been shown that FFAs can promote insulin secretion by activating β-cell FFAR1 receptors (Alquier et al., 2009; Hauke et al., 2018; Schnell et al., 2007). While Caron el al. (Caron et al., 2019) only reported the outcome of acute CNO administration studies, Wang et al. (Wang et al., 2019) also examined the metabolic consequences of chronic CNO exposure. Interestingly, chronic CNO treatment of adipo-GsD mice maintained on a high-fat diet (HFD) significantly slowed the development of adiposity, resulting in multiple metabolic benefits including improved glucose tolerance, increased whole-body energy expenditure, and reduced food intake (Wang et al., 2019). The reduced adiposity displayed by CNO-treated adipo-GsD mice maintained on an obesogenic diet is most likely due to a combination of enhanced energy expenditure and reduced appetite. Chemogenetic activation of Gs signaling also enhanced the thermogenic activity of both brown and white fat, a response predicted to increase energy expenditure (Wang et al., 2019). At present, it remains unclear which signaling pathways and molecules play a role in reducing food intake in CNO-treated adipo-GsD mice.
In agreement with the metabolic phenotypes displayed by CNO-treated adipo-GsD mice, mutant mice lacking Gαs selectively in adipocytes (adipo-Gs-KO mice) showed impaired brown adipose tissue (BAT) function and reduced lipolysis (Li et al., 2016). However, adipo-Gs-KO mice displayed unchanged body weight and energy expenditure, but improved insulin sensitivity and glucose metabolism, possibly due to reduced plasma FFA levels. One possible explanation for the observation that both adipo-Gs-KO mice and CNO-treated adipo-GsD mice showed beneficial metabolic phenotypes may be due to compensatory changes in adipo-Gs-KO mice that lack Gαs throughout development.
The metabolic effects displayed by CNO-treated adipo-GsD mice are similar to those observed after treatment of mice or rats with β3-adrenergic receptor (β3-AR)-selective agonists such CL316,243 (Collins and Surwit, 2001; Ghorbani and Himms-Hagen, 1997; Himms-Hagen et al., 1994; Susulic et al., 1995; Xiao et al., 2015). These effects have been shown to be mediated by activation of Gs-coupled adipocyte β3-ARs (Gavrilova et al., 2000; Grujic et al., 1997) which are abundantly expressed in mouse adipocytes (Collins et al., 1999).
In contrast, β3-ARs are expressed at relatively low levels in human adipocytes where the β1- and β2-ARs are the predominant β-AR subtypes (Robidoux et al., 2004). Despite this altered β-AR expression pattern, acute treatment of healthy male subjects with mirabegron (200 mg orally), a selective agonist at human β3-ARs (Igawa and Michel, 2013), enhanced basal metabolic rate (Cypess et al., 2015). More recently, O’Mara et al. (O’Mara et al., 2020) reported that chronic mirabegron treatment (100 mg per day for 8 weeks) of healthy female subjects stimulated BAT metabolic activity and resting energy expenditure, increased insulin sensitivity, and improved glucose tolerance, suggesting that selective β3-AR agonists might prove useful as novel antidiabetic drugs.. However, two recent studies indicated that the mirabegron-induced metabolic improvements may involve mechanisms that are independent of β3-AR-mediated BAT activation. Finlin et al. (Finlin et al., 2020) reported that treatment of obese, insulin-resistant individuals with mirabegron (50 mg per day for 12 weeks) causes improved glucose homeostasis and that this effect is most likely due to enhanced skeletal muscle oxidative capacity and improved β-cell function (BAT activity was unchanged). These beneficial metabolic effects appear to be secondary to mirabegron’s ability to decrease adipose inflammation and remodeling (Finlin et al., 2020). Another recent study demonstrated that oral administration of mirabegron stimulated BAT thermogenesis only at the maximal allowable dose (200 mg) which also led to the activation of β1- and β2-ARs (Blondin et al., 2020). The authors reported that this high dose of mirabegron causes cardiovascular side effects and stimulates lipolysis in white adipocytes and that mirabegron-induced lipolysis and thermogenesis in BAT are most likely mediated by activation of β2-ARs (Blondin et al., 2020). Cardiovascular side effects following a high dose of mirabegron have also been observed in at least two other studies (Cypess et al., 2015; Malik et al., 2012). Clearly, additional studies are needed to reconcile the seemingly discrepant findings obtained with mirabegron in human clinical trials. Differences in the mirabegron doses used, length of drug application, and the metabolic status of the individuals included in the various studies are all likely factors that may contribute to the different outcomes of these studies.
Adipocyte-specific GiD mice
Adipocytes also express many Gi-coupled receptors (Regard et al., 2008; Wang et al., 2020). In order to explore the in vivo metabolic outcome of selectively activating Gi signaling in adipocytes, Wang et al. (Wang et al., 2020) recently developed and analyzed mice that expressed the GiD designer receptor selectively in adipocytes (adipo-GiD mice) (Fig. 3b). CNO-injected adipo-GiD mice displayed robust reductions in plasma FFA, glycerol, and triglyceride levels, indicating that Gi-mediated signaling inhibits lipolysis in adipocytes. Moreover, independent of the diet that the mice consumed (regular chow or HFD), acute or chronic CNO treatment of adipo-GiD mice led to improved glucose tolerance and enhanced insulin sensitivity, most likely secondary to reduced plasma FFA levels (Wang et al., 2020).
In contrast to the observations by Wang et al. (Wang et al., 2020), a related study (Caron et al., 2019) reported that CNO treatment did not affect FFA release from adipose explants derived from adipo-GiD mice. One possible explanation for these discrepant findings is that chemogenetic activation of Gi signaling is more efficient in vivo where adipose tissue function is regulated by many other factors and neuronal pathways.
On the other hand, mice that lacked functional Gi-type proteins in adipocytes (adipo-Gi-KO mice) showed significant deficits in glucose tolerance and insulin sensitivity (as compared to control littermates) when maintained on a HFD. More detailed studies revelated that increased plasma FFA levels caused by enhanced lipolysis in adipocytes greatly reduced the responsiveness of peripheral tissues to insulin (Wang et al., 2020). In agreement with this finding, previous studies have shown that increased plasma FFA levels can trigger insulin resistance and inflammation, explaining why obesity is closely linked to insulin resistance (Boden, 2008). Taken together, these data convincingly demonstrate that adipocyte Gi signaling plays a central role in regulating lipolysis and whole-body glucose homeostasis.
Adipocytes express several endogenous Gi-coupled receptors, including the CB1 cannabinoid receptor, hydroxycarboxylic acid receptors-1 and −2 (HCA1/GPR81 and HCA2 /GPR109A), and various chemokine and orphan receptors (Regard et al., 2008; Wang et al., 2020). Based on the studies discussed above, these receptors represent potential targets for reducing plasma FFA levels to improve glucose homeostasis in T2D.
Adipocyte-specific expression of GqD
A recent study (Klepac et al., 2016) reported that CNO treatment of GqD-expressing murine brown and white adipocytes inhibited adipocyte differentiation. The authors also found that endothelin acting on Gq-coupled ETA endothelin receptors functions as an autocrine activator of Gq signaling in brown adipocytes. These data suggest that drugs able to inhibit Gq signaling in brown or beige fat may prove useful to enhance energy expenditure for therapeutic purposes.
Hepatocytes
The liver plays a central role in many key metabolic functions, including the regulation of glucose homeostasis. Hepatic glucose fluxes are regulated by insulin, glucagon, and many other hormones and neurotransmitters. In T2D, HGP is increased, particularly in the fasting state (Lin and Accili, 2011; Postic et al., 2004; Unger and Cherrington, 2012). Hepatocytes express dozens of GPCRs linked to distinct G protein signaling pathways (Exton, 1987; Regard et al., 2008). Among these receptors, the metabolic functions of the hepatic glucagon receptor (GCGR) have been studied in great detail. Glucagon binding to the GCGR leads to the activation of Gs, which eventually causes increased cAMP levels and a series of subsequent events that enhance HGP via glycogen breakdown (glycogenolysis) and gluconeogenesis. Thus, the major role of glucagon is to raise blood glucose levels under hypoglycemic conditions (D’Alessio, 2011; Estall and Drucker, 2006; Jiang and Zhang, 2003).
Hepatocyte-specific GsD mice
Akhmedov et al. (Akhmedov et al., 2017a) succeeded in generating a mutant mouse line (Hep-GsD mice) that expressed the GsD construct selectively in hepatocytes (Fig. 3c). As expected, CNO treatment of Hep-GsD mice triggered enhanced hepatic cAMP signaling, glycogenolysis, and changes in gene expression similar to these observed after stimulation of hepatic GCGRs (Akhmedov et al., 2017a). These CNO-induced changes in hepatocyte metabolism resulted in increased blood glucose levels under both fed and fasting conditions, suggesting that inhibition of hepatic Gs signaling might prove useful to lower pathologically elevated blood glucose levels.
In agreement with this notion, Chen et al. (Chen et al., 2005) demonstrated that liver-specific Gαs KO mice (Hep-Gs-KO mice) displayed reduced blood glucose levels and increased glucose tolerance. These phenotypes were opposite to those observed with Hep-GsD mice (Akhmedov et al., 2017a), strongly suggesting that the metabolic outcomes of activating hepatic GsD receptors were indeed mediated by Gs. However, Hep-Gs-KO mice showed greatly increased glucagon and GLP-1 levels and pancreatic α-cell hyperplasia, complicating the interpretation of the metabolic phenotypes displayed by these mutant mice.
Hepatocyte-specific GiD mice
Hepatocytes also express several Gi-linked GPCRs such as certain α2-adrenergic receptor subtypes (Exton, 1985) and the CB1 cannabinoid receptor (Liu et al., 2012; Osei-Hyiaman et al., 2008). To study the metabolic outcomes of stimulating Gi-dependent signaling pathways in hepatocytes, Rossi et al. (Rossi et al., 2018) developed a mutant mouse line that selectively expressed the GiD construct in hepatocytes (Hep-GiD mice) (Fig. 3c). Somewhat surprisingly, CNO treatment of GiD-expressing hepatocytes led to an increase in glucose output (Rossi et al., 2018). In agreement with these in vitro data, CNO-injected Hep-GiD mice displayed elevated blood glucose levels, impaired glucose tolerance, and increased rates of gluconeogenesis and glycogenolysis in vivo (Rossi et al., 2018). On the other hand, mutant mice that lacked functional Gi proteins in hepatocytes (Hep-Gi-KO mice) showed in vivo metabolic phenotypes that were opposite to those caused by CNO treatment of Hep-GiD mice. Moreover, Hep-Gi-KO mice displayed significantly improved glucose homeostasis when maintained on an obesogenic diet (Rossi et al., 2018). Additional studies led to the identification of a signaling pathway that links the stimulation of hepatocyte Gi signaling to the production of reactive oxygen species (ROS), eventually leading to elevated HGP and impaired glucose homeostasis in a JNK-dependent fashion (Rossi et al., 2018).
Rossi et al. (Rossi et al., 2018) also showed that injection of WT mice with anandamide, a cannabinoid receptor agonist, led to impaired glucose tolerance and that this detrimental metabolic effect was mediated by hepatic Gi proteins (Rossi et al., 2018). This observation is in agreement with earlier work demonstrating that signaling initiated by hepatocyte CB1 receptors causes severe metabolic impairments including reduced glucose tolerance (Liu et al., 2012; Osei-Hyiaman et al., 2008).
Taken together, these data strongly suggest that drugs able to inhibit hepatic Gi signaling may prove useful to restore euglycemia in T2D.
Hepatocyte-specific GqD mice
Hepatocytes also express GPCRs that are coupled to G proteins of the Gq family. The best known examples are the AT1a angiotensin II receptor, different α1-adrenergic receptor subtypes, and the V1a and V1b vasopressin receptors (Exton et al., 1981; Li et al., 2013; Lin and Accili, 2011; Regard et al., 2008).
Li et al. (Li et al., 2013) developed a mouse strain that expressed the GqD construct selectively in hepatocytes (Hep-GqD mice) (Fig. 3c). Acute CNO treatment of Hep-GqD mice, but not of WT control mice, markedly increased blood glucose levels, due to increases in the rates of both gluconeogenesis and glycogenolysis (Li et al., 2013). Previous studies suggest that Gq-mediated increases in cytoplasmic Ca2+ levels play a central role in Gq-dependent increases in HGP (Exton, 1987). Gq-mediated changes in gene expression may contribute to the stimulation of gluconeogenesis observed with CNO-injected Hep-GqD mice (Li et al., 2013). Studies with a mouse strain expressing a Gq-biased DREADD (M3D-Gq) in hepatocytes indicated that the increase in HGP caused by GqD activation does not require the recruitment of β-arrestins (Hu et al., 2016).
In summary, these data support the concept that agents that can block the activity of Gq-linked GPCRs endogenously expressed by hepatocytes, including, for example, members of the V1 vasopressin receptor family (Exton, 1987; Li et al., 2013), may prove beneficial to improve glucose homeostasis for therapeutic purposes.
Skeletal muscle
In the postprandial state, skeletal muscle (SKM) is the major site of glucose uptake, and SKM insulin resistance is considered to be the key event in the development of hyperglycemia in T2D (DeFronzo and Tripathy, 2009). Like other cell types, SKM cells express dozens of GPCRs that differ in their G protein coupling preference (Jean-Baptiste et al., 2005; Regard et al., 2008). To date, the potential roles of these receptors in regulating SKM function and whole-body glucose homeostasis have not been investigated systematically.
SKM-specific GqD mice
To study the metabolic effects of stimulating receptor-mediated Gq signaling in SKM, Bone et al. (Bone et al., 2019) analyzed mutant mice that expressed the GqD designer receptor in SKM cells only (SKM-GqD mice) (Fig. 3d). Following CNO administration, lean and obese SKM-GqD mice showed improved glucose homeostasis, most likely due to increased glucose uptake into SKM. On the other hand, obese mutant mice lacking Gαq/11 selectively in SKM displayed even more severe deficits in glucose homeostasis (Bone et al., 2019). Additional studies indicated that Gq-mediated activation of SKM AMPK triggers enhanced GLUT4 translocation to the plasma membrane, thus promoting glucose uptake in an insulin-independent fashion (Bone et al., 2019). These data should be of considerable clinical relevance since SKM glucose uptake is impaired in T2D due to peripheral insulin resistance.
Central AgRP and POMC neurons
The use of DREADD technology and optogenetic techniques has led to major novel insights into the neuronal circuits that control energy and glucose homeostasis in the CNS (Krashes, 2017; Sternson and Eiselt, 2017). Specifically, these studies have shown that various neuronal subpopulations within the hypothalamus are of particular importance in regulating key metabolic functions (Krashes, 2017; Sternson and Eiselt, 2017). The arcuate nucleus of the hypothalamus (ARC) contains two neuronal subpopulations that play central roles in the control of food intake and energy homeostasis (Morton et al., 2014). One of these neuronal subpopulations, so-called AgRP neurons, contains agouti-related protein (AgRP), neuropeptide Y (NPY), and GABA. After activation of these neurons, AgRP is released and inhibits the activity of central melanocortin receptors (predominantly melanocortin-4 receptors [MC4Rs]) to promote food intake and increase energy expenditure (Cone, 2006; Morton et al., 2014). The second major set of ARC neurons is referred to as pro-opiomelanocortin (POMC) neurons. Following their activation, these neurons release α-melanocyte stimulating hormone (α-MSH) which suppresses feeding behavior by binding to and activating central melanocortin receptors (primarily MC4Rs) that are expressed in different areas of the brain (Cone, 2006; Morton et al., 2014).
During the past decade, chemogenetic and optogenetic studies have identified several other neuronal subpopulations that modulate energy homeostasis and other important metabolic processes. Many of these studies have been reviewed recently (Atasoy and Sternson, 2018; Burnett and Krashes, 2016).
AgRP neurons
Early studies have shown that diphtheria toxin-mediated ablation of ARC AgRP neurons causes rapid cessation of feeding in mice (Gropp et al., 2005; Luquet et al., 2005), indicating that this neuronal subpopulation plays a key role play in regulating food intake. The development of optogenetic and chemogenetic strategies has made it possible to selectively modify the activity of AgRP neurons under different experimental conditions. Such studies have led to a wealth of novel information about how the activity of AgRP neurons is regulated under physiological and pathophysiological conditions and which metabolic processes are modulated by these neurons.
In the following, we will briefly summarize studies that used DREADD technology to study the function and regulation of AgRP neurons. Like all other neurons, AgRP neurons express a number of functionally diverse GPCRs (Cowley et al., 2003; Ren et al., 2012). Metabolic studies with mice expressing the GqD, GiD, and GsD designer receptors selectively in AgRP neurons have yielded novel information about the ability of different functional classes of GPCRs to regulate the activity of these neurons and whole body energy homeostasis in general (Fig. 3e).
AgRP neuron-specific GqD and GiD mice
DREADDs have emerged as powerful novel tools for neuroscience research, primarily due to the fact that CNO treatment of GqD-expressing neurons produces rapid depolarization and increased neuronal firing rate (Rogan and Roth, 2011; Roth, 2016). On the other hand, exposure of GiD-expressing neurons to CNO results in neuronal hyperpolarization and decreased firing rate (Rogan and Roth, 2011; Roth, 2016). As expected, CNO treatment of mutant mice expressing the GqD designer receptor selectively in AgRP neurons resulted in neuronal excitation (Krashes et al., 2011). The resulting release of AgRP, NPY, and GABA triggered a rapid increase in food intake and reduced energy expenditure (Krashes et al., 2011). The same laboratory (Krashes et al., 2013) also showed that the release of either NPY or GABA alone was sufficient to promote acute feeding in CNO-treated AgRP-GqD mice. In contrast, AgRP release enhanced food intake over a delayed but prolonged period of time (Krashes et al., 2013). CNO treatment of AgRP-GiD mice led to reduced food intake at the beginning of the dark cycle when mice are feeding most actively (Krashes et al., 2011).
Chemogenetic activation (via GqD) or inhibition (via GiD) of AgRP neurons has no significant effect on blood glucose levels and glucose tolerance (Üner et al., 2019). However, GqD-mediated stimulation of AgRP neurons in diabetic ob/ob mice attenuates the ability of leptin to inhibit feeding but does not interfere with the ability of leptin to lower blood glucose levels (Üner et al., 2019). These observations indicate that changes in the activity of AgRP neurons regulate food intake, but are insufficient to modulate glucose homeostasis in normal and diabetic mice.
Although acute chemogenetic activation of AgRP neurons promotes both food intake and weight gain (Ewbank et al., 2020; Krashes et al., 2011; Krashes et al., 2013), chronic, GqD-mediated stimulation of AgRP neurons causes a return of food intake to baseline levels within one week (Ewbank et al., 2020). Moreover, after chronic chemogenetic activation of AgRP neurons for two months, body weight also returns to baseline levels, most likely due to the activation of compensatory neuronal pathways that counteract the metabolic effects of chronic stimulation of AgRP neurons (Ewbank et al., 2020).
A recent study (Bunner et al., 2020) showed that a single bout of acute exercise increases the activity of ARC AgRP neurons, resulting in increased food intake. This orexigenic effect was abolished after CNO treatment of mice expressing the inhibitory GiD receptor in AgRP neurons (Bunner et al., 2020), indicating that the exercise-induced orexigenic effect is dependent on the activation of AgRP neurons.
The activity of mouse AgRP neurons can also affect other central functions and behaviors including anxiety. For example, a recent study (Li et al., 2019) demonstrated that GqD-mediated activation of AgRP neurons reduces anxiety in fed mice, while GiD-mediated inhibition of AgRP neurons blocks fasting-induced anxiolytic activity. These data indicate that hypothalamic AgRP neurons can also modulate complex behaviors such anxiety.
ARC AgRP neurons also regulate the activity of the sympathetic nervous system and cardiovascular function (Cassaglia et al., 2011; Steculorum et al., 2016). For example, a recent study (Jiang et al., 2020) showed that acute, GqD-mediated activation of AgRP neurons results in a decrease in renal sympathetic nerve activity in conscious mice. Somewhat surprisingly, chronic, GqD-mediated activation of AgRP neurons leads to an increase in blood pressure specifically during the light phase when mice are mostly inactive (Jiang et al., 2020). These findings suggest that the activity state of AgRP neurons regulates sympathetic outflow and can cause major effects on cardiovascular function.
Several of the studies summarized above raise caveats regarding the potential development of drugs aimed at inhibiting the activity of AgRP neurons for therapeutic purposes. Major obstacles are that AgRP neurons do not only regulate food intake but also modulate other complex behaviors (e.g., anxiety) and cardiovascular functions and that the metabolic benefits of this strategy may disappear over time (Ewbank et al., 2020).
AgRP neuron-specific GsD mice
To explore the potential regulation of AgRP neurons by receptor-dependent Gs signaling, Nakajima et al. (Nakajima et al., 2016) generated mice that selectively expressed the GsD construct in AgRP neurons (AgRP-GsD mice) (Fig. 3e). CNO-injected AgRP-GsD mice display a long-lasting increase in food intake that is accompanied by a significant increase in body weight, an effect that is mediated almost exclusively by AgRP release (Nakajima et al., 2016). Additional mechanistic studies suggested that increasing Gs signaling in AgRP neurons promotes the expression of the Klf4 transcription factor in AgRP neurons. Klf4 stimulates the expression of AgRP, ultimately leading to an increase in AgRP release and enhanced appetite. Interestingly, stimulation of Gs-linked PAC1 receptors endogenously expressed by AgRP neurons can mimic the orexigenic effects displayed by CNO-treated AgRP-GsD mice (Nakajima et al., 2016). These observations raise the possibility that drugs able to interfere with Gs signaling in AgRP neurons may prove useful to reduce appetite for therapeutic purposes.
POMC neuron-specific GqD and GiD mice
Like AgRP neurons, POMC neurons play an important role in modulating food intake and energy homeostasis (Morton et al., 2014). Activation and inhibition of POMC neurons by using either DREADD technology or optogenetic techniques reduces and increases food intake, respectively (Aponte et al., 2011; Atasoy et al., 2012). However, these effects occur with considerable delay suggesting that POMC neurons are primarily involved in the long-term regulation of feeding.
In the adult rodent brain, most POMC neurons are located in the ARC (Padilla et al., 2012). However, POMC neurons are also present in the nucleus tractus solitarius (NTS) of the medulla and several other brain regions (Padilla et al., 2012). Interestingly, GqD-mediated activation of NTS POMC neurons results in an immediate reduction in food intake (Zhan et al., 2013). In contrast, only chronic, but not acute, chemogenetic stimulation of ARC POMC neurons suppresses food intake (Zhan et al., 2013), indicating that POMC neurons in the ARC and NTS have different functional roles. While NTS POMC neurons appear to respond to acute satiety signals, ARC POMC neurons seem to be involved in long-term regulation of feeding behavior and energy homeostasis.
The activity of POMC neurons is regulated by two major circulating hormones, leptin and insulin. Leptin depolarizes and activates POMC neurons, leading to the suppression of food intake (Morton et al., 2014). By contrast, insulin inhibits POMC neurons, thus limiting α-MSH release (Morton et al., 2014). However, under certain experimental conditions, POMC neurons can also be activated by insulin (Qiu et al., 2018; Qiu et al., 2014). A recent study demonstrated that the proportion of POMC neurons that is activated by insulin depends on the activity of the phosphatase TCPTP, which is increased after fasting but reduced in the fed state (Dodd et al., 2018). These authors also reported that prolonged GiD-mediated inhibition of ARC POMC neurons leads to increased HGP and impaired whole-body insulin sensitivity. On the other hand, chronic, GqD-mediated activation of ARC POMC neurons lowers HGP and improves hepatic glucose metabolism (Dodd et al., 2018). Taken together, these observations suggest that changes in the proportion of POMC neurons that are activated or inhibited by insulin may directly affect peripheral glucose metabolism. Importantly, this study highlights the fact that changes in the activity of POMC neurons can affect the function of metabolically important peripheral cell types such as hepatocytes of the liver.
In agreement with this concept, a recent study (Brandt et al., 2018) demonstrated that chemogenetic activation of ARC POMC neurons stimulates the activity of sympathetic nerves supplying the liver, thus causing changes in liver metabolism (e.g., activation of mTOR signaling) that prime the liver for incoming nutrients. The authors also reported that sensory food perception activates ARC POMC neurons, resulting in similar hepatic effects which appear to be dependent on the activation of MC4Rs (Brandt et al., 2018). These observations indicate that POMC neurons play a central role in a circuit linking sensory food anticipation to changes in hepatocyte function that prime the liver for incoming nutrients.
A related study showed that GiD-mediated inhibition of ARC POMC neurons promotes food intake and lowers blood glucose levels in normoglycemic mice (Uner et al., 2019). Additional experimentation revealed that the hypoglycemic effect caused by inhibition of POMC neurons is independent of changes in food intake (Uner et al., 2019). Although the neuronal circuits underlying this hypoglycemic response remain to be elucidated, these findings suggest that POMC neurons represent a potential target for novel drugs aimed at treating hyperglycemia.
The activity of POMC neurons can also affect cardiovascular parameters including blood pressure. A recent study reported that chronic GqD-mediated activation of mouse ARC POMC neurons leads to a significant reduction in blood pressure (Jiang et al., 2020). The molecular and cellular mechanisms underlying this effect remain to be explored in future studies.
Conclusions and future directions
The studies summarized in this short review clearly indicate that DREADD-based approaches are highly useful to assess the in vivo metabolic consequences of activating specific GPCR/G protein signaling cascades in metabolically distinct cell types. The novel information derived from these studies should stimulate efforts to develop novel classes of drugs useful for the treatment of several metabolic disorders including T2D and obesity (Table 1). Endogenous GPCRs need be identified that can be targeted to mimic the beneficial effects observed with the DREADD mutant mice. Additional studies are required to ensure that the metabolically important signaling pathways identified by the use of DREADD technology in mice are also operative in human tissues. Finally, it remains to be explored whether receptor (DREADD)-mediated recruitment of β-arrestins contributes to the physiological effects caused by DREADD activation in different cell types. Such information is important for the design of novel drugs endowed with pharmacological profiles that ensure maximum efficacy and a favorable side effect profile.
Table 1.
Potential therapeutic strategies to improve glucose hemostasis and/or reduce obesity, as suggested by the use of DREADD technology in mice
Cell type | DREADD | Major phenotypes after DREADD activation | Drugs predicted to improve glucose hemostasis and/or reduce obesity | Refs. |
---|---|---|---|---|
β-cells | GqD | Increase in insulin secretion, β-cell proliferation, and β-cell mass | GPCR agonists that stimulate β-cell Gq | (Guettier et al., 2009) (Jain et al., 2013) |
GsD | Enhanced insulin release and increased β-cell mass | GPCR agonists that activate β-cell Gs | (Guettier et al., 2009) | |
α-cells | GiD | Reduced glucagon secretion Deficits in glucose tolerance and GSIS |
GPCR agonists that enhance α-cell activity | (Zhu et al., 2019) |
L-cells (colon) | GqD | Improved glucose tolerance Reduced food intake |
GPCR agonists that stimulate L-cell Gq | (Lewis et al., 2020) |
Adipocytes | GsD | Increased lipolysis and energy expenditure Reduced food intake and improved glucose homeostasis |
GPCR agonists that stimulate adipocyte Gs | (Wang et al., 2019) (Caron et al., 2019) |
GiD | Impaired lipolysis Improved insulin and glucose tolerance |
GPCR agonists that activate adipocyte Gi | (Wang et al., 2020) | |
GqD | Inhibition of brown adipocyte differentiation | GPCR antagonists that block Gq signaling in brown/beige fat | (Klepac et al., 2016) | |
Hepatocytes | GsD | Enhanced glycogenolysis and hyperglycemia | GPCR antagonists that block hepatocyte Gs signaling | (Akhmedov et al., 2017a) |
GiD | Elevated HGP and hyperglycemia Impaired glucose tolerance |
GPCR antagonists that interfere with hepatocyte Gi signaling | (Rossi et al., 2018) | |
GqD | Increased HGP and hyperglycemia | GPCR antagonists that inhibit hepatocyte Gq signaling | (Li et al., 2013) | |
Skeletal muscle cells | GqD | Enhanced glucose uptake into sk. musc. Improved glucose homeostasis |
GPCR agonists that stimulate skeletal muscle Gq | (Bone et al., 2019) |
AgRP neurons | GqD | Enhanced food intake Weight gain |
GPCR antagonists suppressing Gq signaling in AgRP neurons | (Krashes et al., 2011) |
GiD | Reduced food intake | GPCR agonists enhancing Gi signaling in AgRP neurons | (Krashes et al., 2011) | |
GsD | Enhanced food intake Increase in body weight |
GPCR antagonists inhibiting Gs signaling in AgRP neurons | (Nakajima et al., 2016) | |
POMC neurons | GqD | Reduced food intake | GPCR agonists promotin Gq signaling in POMC neurons | (Zhan et al., 2013) (Atasoy et al., 2012) |
GiD | Increased food intake | GPCR antagonists suppressing Gi signaling in POMC neurons | (Krashes et al., 2011) | |
GiD | Increased food intake | GPCR agonists stimulating Gi signaling in POMC neurons | (Uner et al., 2019) |
Abbreviations: HGP, hepatic glucose production; GSIS, glucose-stimulated insulin secretion.
Acknowledgements - Conflict of interest disclosure
We are grateful for the contributions of many past members of the Wess laboratory who carried out some of the studies reviewed in this article. We apologize to those individuals whose work we were unable to discuss in this short review because of space constraints. The work carried out in the Wess laboratory reviewed in this article was supported by the Intramural Research Program of the NIH, NIDDK, Bethesda, Maryland, USA. The authors have no conflict of interest to declare.
List of abbreviations
- ACh
acetylcholine
- AgRP
agouti-related protein
- CNO
clozapine-N-oxide
- DCZ
deschloroclozapine
- DREADD
Designer Receptor Exclusively Activated by a Designer Drug
- FFA
free fatty acid
- FFAR
free fatty acid receptor 1
- GCGR
glucagon receptor
- GIRK channel
G-protein-regulated inward-rectifier potassium channel
- GLP-1
glucagon-like peptide-1
- GPCR
G protein-coupled receptor
- GSIS
glucose-stimulated insulin secretion
- HFD
high-fat diet
- HGP
hepatic glucose production
- IP3
inositol trisphosphate
- IRS2
insulin receptor substrate 2
- MC4 receptor
melanocortin 4 receptor
- NPY
agouti-related peptide
- NPY
neuropeptide Y
- POMC
pro-opiomelanocortin
- PTX
pertussis toxin
- PYY
peptide YY
- RASSL
Receptor Activated Solely by Synthetic Ligands
- ROS
reactive oxygen species
- SKM
skeletal muscle
- T2D
type 2 diabetes
References
- Ahren B (2009). Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes. Nat Rev Drug Discov 8, 369–385. [DOI] [PubMed] [Google Scholar]
- Ahren B (2015). Glucagon--Early breakthroughs and recent discoveries. Peptides 67, 74–81. [DOI] [PubMed] [Google Scholar]
- Akhmedov D, Mendoza-Rodriguez MG, Rajendran K, Rossi M, Wess J, and Berdeaux R (2017a). Gs-DREADD knock-in mice for tissue-specific, temporal stimulation of cAMP signaling. Molecular and cellular biology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akhmedov D, Mendoza-Rodriguez MG, Rajendran K, Rossi M, Wess J, and Berdeaux R (2017b). Gs-DREADD Knock-In Mice for Tissue-Specific, Temporal Stimulation of Cyclic AMP Signaling. Molecular and cellular biology 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ali S, and Drucker DJ (2009). Benefits and limitations of reducing glucagon action for the treatment of type 2 diabetes. Am J Physiol Endocrinol Metab 296, E415–421. [DOI] [PubMed] [Google Scholar]
- Alquier T, Peyot ML, Latour MG, Kebede M, Sorensen CM, Gesta S, Ronald Kahn C, Smith RD, Jetton TL, Metz TO, et al. (2009). Deletion of GPR40 impairs glucose-induced insulin secretion in vivo in mice without affecting intracellular fuel metabolism in islets. Diabetes 58, 2607–2615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alvarez-Curto E, Prihandoko R, Tautermann CS, Zwier JM, Pediani JD, Lohse MJ, Hoffmann C, Tobin AB, and Milligan G (2011). Developing chemical genetic approaches to explore G protein-coupled receptor function: validation of the use of a receptor activated solely by synthetic ligand (RASSL). Mol Pharmacol 80, 1033–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amisten S, Neville M, Hawkes R, Persaud SJ, Karpe F, and Salehi A (2015). An atlas of G-protein coupled receptor expression and function in human subcutaneous adipose tissue. Pharmacol Ther 146, 61–93. [DOI] [PubMed] [Google Scholar]
- Amisten S, Salehi A, Rorsman P, Jones PM, and Persaud SJ (2013). An atlas and functional analysis of G-protein coupled receptors in human islets of Langerhans. Pharmacol Ther 139, 359–391. [DOI] [PubMed] [Google Scholar]
- Aponte Y, Atasoy D, and Sternson SM (2011). AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without training. Nature neuroscience 14, 351–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Armbruster BN, Li X, Pausch MH, Herlitze S, and Roth BL (2007). Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand. Proc Natl Acad Sci U S A 104, 5163–5168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atasoy D, Betley JN, Su HH, and Sternson SM (2012). Deconstruction of a neural circuit for hunger. Nature 488, 172–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atasoy D, and Sternson SM (2018). Chemogenetic Tools for Causal Cellular and Neuronal Biology. Physiological reviews 98, 391–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beaulieu JM, Gainetdinov RR, and Caron MG (2009). Akt/GSK3 signaling in the action of psychotropic drugs. Annu Rev Pharmacol Toxicol 49, 327–347. [DOI] [PubMed] [Google Scholar]
- Billing LJ, Smith CA, Larraufie P, Goldspink DA, Galvin S, Kay RG, Howe JD, Walker R, Pruna M, Glass L, et al. (2018). Co-storage and release of insulin-like peptide-5, glucagon-like peptide-1 and peptideYY from murine and human colonic enteroendocrine cells. Molecular metabolism 16, 65–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blondin DP, Nielsen S, Kuipers EN, Severinsen MC, Jensen VH, Miard S, Jespersen NZ, Kooijman S, Boon MR, Fortin M, et al. (2020). Human Brown Adipocyte Thermogenesis Is Driven by β2-AR Stimulation. Cell Metab 32, 287–300.e287. [DOI] [PubMed] [Google Scholar]
- Boden G (2008). Obesity and free fatty acids. Endocrinol Metab Clin North Am 37, 635–646, viii-ix. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolognini D, Barki N, Butcher AJ, Hudson BD, Sergeev E, Molloy C, Moss CE, Bradley SJ, Le Gouill C, Bouvier M, et al. (2019). Chemogenetics defines receptor-mediated functions of short chain free fatty acids. Nat Chem Biol 15, 489–498. [DOI] [PubMed] [Google Scholar]
- Bolognini D, Tobin AB, Milligan G, and Moss CE (2016). The Pharmacology and Function of Receptors for Short-Chain Fatty Acids. Mol Pharmacol 89, 388–398. [DOI] [PubMed] [Google Scholar]
- Bone DBJ, Meister J, Knudsen JR, Dattaroy D, Cohen A, Lee R, Lu H, Metzger D, Jensen TE, and Wess J (2019). Skeletal Muscle-Specific Activation of Gq Signaling Maintains Glucose Homeostasis. Diabetes 68, 1341–1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brandt C, Nolte H, Henschke S, Engström Ruud L, Awazawa M, Morgan DA, Gabel P, Sprenger HG, Hess ME, Günther S, et al. (2018). Food Perception Primes Hepatic ER Homeostasis via Melanocortin-Dependent Control of mTOR Activation. Cell 175, 1321–1335.e1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bunner W, Landry T, Laing BT, Li P, Rao Z, Yuan Y, and Huang H (2020). ARC(AgRP/NPY) Neuron Activity Is Required for Acute Exercise-Induced Food Intake in Un-Trained Mice. Front Physiol 11, 411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burnett CJ, and Krashes MJ (2016). Resolving Behavioral Output via Chemogenetic Designer Receptors Exclusively Activated by Designer Drugs. J Neurosci 36, 9268–9282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capozzi ME, Svendsen B, Encisco SE, Lewandowski SL, Martin MD, Lin H, Jaffe JL, Coch RW, Haldeman JM, MacDonald PE, et al. (2019a). beta Cell tone is defined by proglucagon peptides through cAMP signaling. JCI insight 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capozzi ME, Wait JB, Koech J, Gordon AN, Coch RW, Svendsen B, Finan B, D’Alessio DA, and Campbell JE (2019b). Glucagon lowers glycemia when beta-cells are active. JCI insight 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caron A, Reynolds RP, Castorena CM, Michael NJ, Lee CE, Lee S, Berdeaux R, Scherer PE, and Elmquist JK (2019). Adipocyte Gs but not Gi signaling regulates whole-body glucose homeostasis. Molecular metabolism. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cassaglia PA, Hermes SM, Aicher SA, and Brooks VL (2011). Insulin acts in the arcuate nucleus to increase lumbar sympathetic nerve activity and baroreflex function in rats. J Physiol 589, 1643–1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang WH, Lin SK, Lane HY, Wei FC, Hu WH, Lam YW, and Jann MW (1998). Reversible metabolism of clozapine and clozapine N-oxide in schizophrenic patients. Prog Neuropsychopharmacol Biol Psychiatry 22, 723–739. [DOI] [PubMed] [Google Scholar]
- Chen M, Gavrilova O, Zhao WQ, Nguyen A, Lorenzo J, Shen L, Nackers L, Pack S, Jou W, and Weinstein LS (2005). Increased glucose tolerance and reduced adiposity in the absence of fasting hypoglycemia in mice with liver-specific Gs alpha deficiency. J Clin Invest 115, 3217–3227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, Choo H, Huang XP, Yang X, Stone O, Roth BL, and Jin J (2015). The first structure-activity relationship studies for designer receptors exclusively activated by designer drugs. ACS Chem Neurosci 6, 476–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chia CW, and Egan JM (2020). Incretins in obesity and diabetes. Ann N Y Acad Sci 1461, 104–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collins S, Daniel KW, and Rohlfs EM (1999). Depressed expression of adipocyte beta-adrenergic receptors is a common feature of congenital and diet-induced obesity in rodents. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity 23, 669–677. [DOI] [PubMed] [Google Scholar]
- Collins S, and Surwit RS (2001). The beta-adrenergic receptors and the control of adipose tissue metabolism and thermogenesis. Recent progress in hormone research 56, 309–328. [DOI] [PubMed] [Google Scholar]
- Cone RD (2006). Studies on the physiological functions of the melanocortin system. Endocr Rev 27, 736–749. [DOI] [PubMed] [Google Scholar]
- Conklin BR, Hsiao EC, Claeysen S, Dumuis A, Srinivasan S, Forsayeth JR, Guettier JM, Chang WC, Pei Y, McCarthy KD, et al. (2008). Engineering GPCR signaling pathways with RASSLs. Nat Methods 5, 673–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coward P, Wada HG, Falk MS, Chan SD, Meng F, Akil H, and Conklin BR (1998). Controlling signaling with a specifically designed Gi-coupled receptor. Proc Natl Acad Sci U S A 95, 352–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cowley MA, Smith RG, Diano S, Tschop M, Pronchuk N, Grove KL, Strasburger CJ, Bidlingmaier M, Esterman M, Heiman ML, et al. (2003). The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron 37, 649–661. [DOI] [PubMed] [Google Scholar]
- Cypess AM, Weiner LS, Roberts-Toler C, Franquet Elia E, Kessler SH, Kahn PA, English J, Chatman K, Trauger SA, Doria A, et al. (2015). Activation of human brown adipose tissue by a beta3-adrenergic receptor agonist. Cell Metab 21, 33–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Alessio D (2011). The role of dysregulated glucagon secretion in type 2 diabetes. Diabetes Obes Metab 13 Suppl 1, 126–132. [DOI] [PubMed] [Google Scholar]
- DeFea KA (2011). Beta-arrestins as regulators of signal termination and transduction: how do they determine what to scaffold? Cellular signalling 23, 621–629. [DOI] [PubMed] [Google Scholar]
- DeFronzo RA, and Tripathy D (2009). Skeletal muscle insulin resistance is the primary defect in type 2 diabetes. Diabetes care 32 Suppl 2, S157–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dodd GT, Michael NJ, Lee-Young RS, Mangiafico SP, Pryor JT, Munder AC, Simonds SE, Brüning JC, Zhang ZY, Cowley MA, et al. (2018). Insulin regulates POMC neuronal plasticity to control glucose metabolism. Elife 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drucker DJ (2018). Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab 27, 740–756. [DOI] [PubMed] [Google Scholar]
- Drucker DJ, and Nauck MA (2006). The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368, 1696–1705. [DOI] [PubMed] [Google Scholar]
- Estall JL, and Drucker DJ (2006). Glucagon and glucagon-like peptide receptors as drug targets. Curr Pharm Des 12, 1731–1750. [DOI] [PubMed] [Google Scholar]
- Ewbank SN, Campos CA, Chen JY, Bowen AJ, Padilla SL, Dempsey JL, Cui JY, and Palmiter RD (2020). Chronic G(q) signaling in AgRP neurons does not cause obesity. Proc Natl Acad Sci U S A 117, 20874–20880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Exton JH (1985). Mechanisms involved in alpha-adrenergic phenomena. Am J Physiol 248, E633–647. [DOI] [PubMed] [Google Scholar]
- Exton JH (1987). Mechanisms of hormonal regulation of hepatic glucose metabolism. Diabetes Metab Rev 3, 163–183. [DOI] [PubMed] [Google Scholar]
- Exton JH, Blackmore PF, El-Refai MF, Dehaye JP, Strickland WG, Cherrington AD, Chan TM, Assimacopoulos-Jeannet FD, and Chrisman TD (1981). Mechanisms of hormonal regulation of liver metabolism. Adv Cyclic Nucleotide Res 14, 491–505. [PubMed] [Google Scholar]
- Finlin BS, Memetimin H, Zhu B, Confides AL, Vekaria HJ, El Khouli RH, Johnson ZR, Westgate PM, Chen J, Morris AJ, et al. (2020). The beta3-adrenergic receptor agonist mirabegron improves glucose homeostasis in obese humans. J Clin Invest. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gavrilova O, Marcus-Samuels B, and Reitman ML (2000). Lack of responses to a beta3-adrenergic agonist in lipoatrophic A-ZIP/F-1 mice. Diabetes 49, 1910–1916. [DOI] [PubMed] [Google Scholar]
- Ghorbani M, and Himms-Hagen J (1997). Appearance of brown adipocytes in white adipose tissue during CL 316,243-induced reversal of obesity and diabetes in Zucker fa/fa rats. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity 21, 465–475. [DOI] [PubMed] [Google Scholar]
- Gomez JL, Bonaventura J, Lesniak W, Mathews WB, Sysa-Shah P, Rodriguez LA, Ellis RJ, Richie CT, Harvey BK, Dannals RF, et al. (2017). Chemogenetics revealed: DREADD occupancy and activation via converted clozapine. Science 357, 503–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez-Muniesa P, Martinez-Gonzalez MA, Hu FB, Despres JP, Matsuzawa Y, Loos RJF, Moreno LA, Bray GA, and Martinez JA (2017). Obesity. Nature reviews Disease primers 3, 17034. [DOI] [PubMed] [Google Scholar]
- Gribble FM, and Reimann F (2016). Enteroendocrine Cells: Chemosensors in the Intestinal Epithelium. Annu Rev Physiol 78, 277–299. [DOI] [PubMed] [Google Scholar]
- Gropp E, Shanabrough M, Borok E, Xu AW, Janoschek R, Buch T, Plum L, Balthasar N, Hampel B, Waisman A, et al. (2005). Agouti-related peptide-expressing neurons are mandatory for feeding. Nature neuroscience 8, 1289–1291. [DOI] [PubMed] [Google Scholar]
- Grujic D, Susulic VS, Harper ME, Himms-Hagen J, Cunningham BA, Corkey BE, and Lowell BB (1997). Beta3-adrenergic receptors on white and brown adipocytes mediate beta3-selective agonist-induced effects on energy expenditure, insulin secretion, and food intake. A study using transgenic and gene knockout mice. J Biol Chem 272, 17686–17693. [DOI] [PubMed] [Google Scholar]
- Guettier JM, Gautam D, Scarselli M, Ruiz de Azua I, Li JH, Rosemond E, Ma X, Gonzalez FJ, Armbruster BN, Lu H, et al. (2009). A chemical-genetic approach to study G protein regulation of beta cell function in vivo. Proc Natl Acad Sci U S A 106, 19197–19202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guh DP, Zhang W, Bansback N, Amarsi Z, Birmingham CL, and Anis AH (2009). The incidence of co-morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC public health 9, 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guilherme A, Virbasius JV, Puri V, and Czech MP (2008). Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol 9, 367–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gurevich VV, and Gurevich EV (2014). Overview of different mechanisms of arrestin-mediated signaling. Curr Protoc Pharmacol 67, Unit 2.10.11–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gurevich VV, and Gurevich EV (2019). Plethora of functions packed into 45 kDa arrestins: biological implications and possible therapeutic strategies. Cell Mol Life Sci 76, 4413–4421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hauke S, Keutler K, Phapale P, Yushchenko DA, and Schultz C (2018). Endogenous Fatty Acids Are Essential Signaling Factors of Pancreatic beta-Cells and Insulin Secretion. Diabetes 67, 1986–1998. [DOI] [PubMed] [Google Scholar]
- Himms-Hagen J, Cui J, Danforth E Jr., Taatjes DJ, Lang SS, Waters BL, and Claus TH (1994). Effect of CL-316,243, a thermogenic beta 3-agonist, on energy balance and brown and white adipose tissues in rats. Am J Physiol 266, R1371–1382. [DOI] [PubMed] [Google Scholar]
- Hu J, Stern M, Gimenez LE, Wanka L, Zhu L, Rossi M, Meister J, Inoue A, Beck-Sickinger AG, Gurevich VV, et al. (2016). A G Protein-biased Designer G Protein-coupled Receptor Useful for Studying the Physiological Relevance of Gq/11-dependent Signaling Pathways. J Biol Chem 291, 7809–7820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hudish LI, Reusch JE, and Sussel L (2019). beta Cell dysfunction during progression of metabolic syndrome to type 2 diabetes. J Clin Invest 129, 4001–4008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hudson BD, Christiansen E, Tikhonova IG, Grundmann M, Kostenis E, Adams DR, Ulven T, and Milligan G (2012). Chemically engineering ligand selectivity at the free fatty acid receptor 2 based on pharmacological variation between species orthologs. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 26, 4951–4965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Igawa Y, and Michel MC (2013). Pharmacological profile of beta3-adrenoceptor agonists in clinical development for the treatment of overactive bladder syndrome. Naunyn-Schmiedeberg’s archives of pharmacology 386, 177–183. [DOI] [PubMed] [Google Scholar]
- Inoue A, Raimondi F, Kadji FMN, Singh G, Kishi T, Uwamizu A, Ono Y, Shinjo Y, Ishida S, Arang N, et al. (2019). Illuminating G-Protein-Coupling Selectivity of GPCRs. Cell 177, 1933–1947.e1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jain S, Ruiz de Azua I, Lu H, White MF, Guettier JM, and Wess J (2013). Chronic activation of a designer G(q)-coupled receptor improves beta cell function. J Clin Invest 123, 1750–1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jann MW, Lam YW, and Chang WH (1994). Rapid formation of clozapine in guinea-pigs and man following clozapine-N-oxide administration. Arch Int Pharmacodyn Ther 328, 243–250. [PubMed] [Google Scholar]
- Jean-Baptiste G, Yang Z, Khoury C, Gaudio S, and Greenwood MT (2005). Peptide and non-peptide G-protein coupled receptors (GPCRs) in skeletal muscle. Peptides 26, 1528–1536. [DOI] [PubMed] [Google Scholar]
- Jiang G, and Zhang BB (2003). Glucagon and regulation of glucose metabolism. Am J Physiol Endocrinol Metab 284, E671–678. [DOI] [PubMed] [Google Scholar]
- Jiang J, Morgan DA, Cui H, and Rahmouni K (2020). Activation of hypothalamic AgRP and POMC neurons evokes disparate sympathetic and cardiovascular responses. Am J Physiol Heart Circ Physiol 319, H1069–h1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenakin T, and Christopoulos A (2013). Signalling bias in new drug discovery: detection, quantification and therapeutic impact. Nat Rev Drug Discov 12, 205–216. [DOI] [PubMed] [Google Scholar]
- Klepac K, Kilic A, Gnad T, Brown LM, Herrmann B, Wilderman A, Balkow A, Glode A, Simon K, Lidell ME, et al. (2016). The Gq signalling pathway inhibits brown and beige adipose tissue. Nat Commun 7, 10895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krashes MJ (2017). Untangling Appetite Circuits with Optogenetics and Chemogenetics. In Appetite and Food Intake: Central Control, nd, and Harris RBS, eds. (Boca Raton (FL): CRC Press/Taylor & Francisc 2017 by Taylor & Francis Group, LLC.), pp. 91–116. [PubMed] [Google Scholar]
- Krashes MJ, Koda S, Ye C, Rogan SC, Adams AC, Cusher DS, Maratos-Flier E, Roth BL, and Lowell BB (2011). Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J Clin Invest 121, 1424–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krashes MJ, Shah BP, Koda S, and Lowell BB (2013). Rapid versus delayed stimulation of feeding by the endogenously released AgRP neuron mediators GABA, NPY, and AgRP. Cell Metab 18, 588–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kusminski CM, Bickel PE, and Scherer PE (2016). Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat Rev Drug Discov 15, 639–660. [DOI] [PubMed] [Google Scholar]
- Lee YH, Wang MY, Yu XX, and Unger RH (2016). Glucagon is the key factor in the development of diabetes. Diabetologia 59, 1372–1375. [DOI] [PubMed] [Google Scholar]
- Lewis JE, Miedzybrodzka EL, Foreman RE, Woodward ORM, Kay RG, Goldspink DA, Gribble FM, and Reimann F (2020). Selective stimulation of colonic L cells improves metabolic outcomes in mice. Diabetologia 63, 1396–1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li C, Hou Y, Zhang J, Sui G, Du X, Licinio J, Wong ML, and Yang Y (2019). AGRP neurons modulate fasting-induced anxiolytic effects. Transl Psychiatry 9, 111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li JH, Jain S, McMillin SM, Cui Y, Gautam D, Sakamoto W, Lu H, Jou W, McGuinness OP, Gavrilova O, et al. (2013). A novel experimental strategy to assess the metabolic effects of selective activation of a G(q)-coupled receptor in hepatocytes in vivo. Endocrinology 154, 3539–3551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li YQ, Shrestha YB, Chen M, Chanturiya T, Gavrilova O, and Weinstein LS (2016). Gsalpha deficiency in adipose tissue improves glucose metabolism and insulin sensitivity without an effect on body weight. Proc Natl Acad Sci U S A 113, 446–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z, Zhou Z, and Zhang L (2020). Current status of GPR40/FFAR1 modulators in medicinal chemistry (2016–2019): a patent review. Expert Opin Ther Pat 30, 27–38. [DOI] [PubMed] [Google Scholar]
- Lin HV, and Accili D (2011). Hormonal regulation of hepatic glucose production in health and disease. Cell Metab 14, 9–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J, Zhou L, Xiong K, Godlewski G, Mukhopadhyay B, Tam J, Yin S, Gao P, Shan X, Pickel J, et al. (2012). Hepatic cannabinoid receptor-1 mediates diet-induced insulin resistance via inhibition of insulin signaling and clearance in mice. Gastroenterology 142, 1218–1228 e1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luquet S, Perez FA, Hnasko TS, and Palmiter RD (2005). NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 310, 683–685. [DOI] [PubMed] [Google Scholar]
- Luttrell LM, and Gesty-Palmer D (2010). Beyond desensitization: physiological relevance of arrestin-dependent signaling. Pharmacol Rev 62, 305–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luttrell LM, Maudsley S, and Bohn LM (2015). Fulfilling the Promise of “Biased” G Protein-Coupled Receptor Agonism. Mol Pharmacol 88, 579–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Makhmutova M, Weitz J, Tamayo A, Pereira E, Boulina M, Almaça J, Rodriguez-Diaz R, and Caicedo A (2020). Pancreatic β-Cells Communicate With Vagal Neurons. Gastroenterology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malik M, van Gelderen EM, Lee JH, Kowalski DL, Yen M, Goldwater R, Mujais SK, Schaddelee MP, de Koning P, Kaibara A, et al. (2012). Proarrhythmic safety of repeat doses of mirabegron in healthy subjects: a randomized, double-blind, placebo-, and active-controlled thorough QT study. Clin Pharmacol Ther 92, 696–706. [DOI] [PubMed] [Google Scholar]
- Manvich DF, Webster KA, Foster SL, Farrell MS, Ritchie JC, Porter JH, and Weinshenker D (2018). The DREADD agonist clozapine N-oxide (CNO) is reverse-metabolized to clozapine and produces clozapine-like interoceptive stimulus effects in rats and mice. Sci Rep 8, 3840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinez VK, Saldana-Morales F, Sun JJ, Zhu PJ, Costa-Mattioli M, and Ray RS (2019). Off-Target Effects of Clozapine-N-Oxide on the Chemosensory Reflex Are Masked by High Stress Levels. Front Physiol 10, 521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morton GJ, Meek TH, and Schwartz MW (2014). Neurobiology of food intake in health and disease. Nat Rev Neurosci 15, 367–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Müller TD, Finan B, Bloom SR, D’Alessio D, Drucker DJ, Flatt PR, Fritsche A, Gribble F, Grill HJ, Habener JF, et al. (2019). Glucagon-like peptide 1 (GLP-1). Molecular metabolism 30, 72–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nagai Y, Miyakawa N, Takuwa H, Hori Y, Oyama K, Ji B, Takahashi M, Huang XP, Slocum ST, DiBerto JF, et al. (2020). Deschloroclozapine, a potent and selective chemogenetic actuator enables rapid neuronal and behavioral modulations in mice and monkeys. Nature neuroscience. [DOI] [PubMed] [Google Scholar]
- Nakajima K, Cui Z, Li C, Meister J, Cui Y, Fu O, Smith AS, Jain S, Lowell BB, Krashes MJ, et al. (2016). Gs-coupled GPCR signalling in AgRP neurons triggers sustained increase in food intake. Nat Commun 7, 10268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakajima K, and Wess J (2012). Design and functional characterization of a novel, arrestin-biased designer G protein-coupled receptor. Mol Pharmacol 82, 575–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Mara AE, Johnson JW, Linderman JD, Brychta RJ, McGehee S, Fletcher LA, Fink YA, Kapuria D, Cassimatis TM, Kelsey N, et al. (2020). Chronic mirabegron treatment increases human brown fat, HDL cholesterol, and insulin sensitivity. J Clin Invest. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Osei-Hyiaman D, Liu J, Zhou L, Godlewski G, Harvey-White J, Jeong WI, Batkai S, Marsicano G, Lutz B, Buettner C, et al. (2008). Hepatic CB1 receptor is required for development of diet-induced steatosis, dyslipidemia, and insulin and leptin resistance in mice. J Clin Invest 118, 3160–3169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Padilla SL, Reef D, and Zeltser LM (2012). Defining POMC neurons using transgenic reagents: impact of transient Pomc expression in diverse immature neuronal populations. Endocrinology 153, 1219–1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pierce KL, and Lefkowitz RJ (2001). Classical and new roles of beta-arrestins in the regulation of G-protein-coupled receptors. Nat Rev Neurosci 2, 727–733. [DOI] [PubMed] [Google Scholar]
- Pierce KL, Premont RT, and Lefkowitz RJ (2002). Seven-transmembrane receptors. Nat Rev Mol Cell Biol 3, 639–650. [DOI] [PubMed] [Google Scholar]
- Postic C, Dentin R, and Girard J (2004). Role of the liver in the control of carbohydrate and lipid homeostasis. Diabetes Metab 30, 398–408. [DOI] [PubMed] [Google Scholar]
- Qiu J, Wagner EJ, Rønnekleiv OK, and Kelly MJ (2018). Insulin and leptin excite anorexigenic pro-opiomelanocortin neurones via activation of TRPC5 channels. J Neuroendocrinol 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu J, Zhang C, Borgquist A, Nestor CC, Smith AW, Bosch MA, Ku S, Wagner EJ, Rønnekleiv OK, and Kelly MJ (2014). Insulin excites anorexigenic proopiomelanocortin neurons via activation of canonical transient receptor potential channels. Cell Metab 19, 682–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quesada I, Tuduri E, Ripoll C, and Nadal A (2008). Physiology of the pancreatic alpha-cell and glucagon secretion: role in glucose homeostasis and diabetes. The Journal of endocrinology 199, 5–19. [DOI] [PubMed] [Google Scholar]
- Rajagopal S, Rajagopal K, and Lefkowitz RJ (2010). Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nat Rev Drug Discov 9, 373–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raper J, Morrison RD, Daniels JS, Howell L, Bachevalier J, Wichmann T, and Galvan A (2017). Metabolism and Distribution of Clozapine-N-oxide: Implications for Nonhuman Primate Chemogenetics. ACS Chem Neurosci 8, 1570–1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Regard JB, Sato IT, and Coughlin SR (2008). Anatomical profiling of G protein-coupled receptor expression. Cell 135, 561–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren H, Orozco IJ, Su Y, Suyama S, Gutierrez-Juarez R, Horvath TL, Wardlaw SL, Plum L, Arancio O, and Accili D (2012). FoxO1 target Gpr17 activates AgRP neurons to regulate food intake. Cell 149, 1314–1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riddy DM, Delerive P, Summers RJ, Sexton PM, and Langmead CJ (2018). G Protein-Coupled Receptors Targeting Insulin Resistance, Obesity, and Type 2 Diabetes Mellitus. Pharmacol Rev 70, 39–67. [DOI] [PubMed] [Google Scholar]
- Robidoux J, Martin TL, and Collins S (2004). Beta-adrenergic receptors and regulation of energy expenditure: a family affair. Annu Rev Pharmacol Toxicol 44, 297–323. [DOI] [PubMed] [Google Scholar]
- Roden M, and Shulman GI (2019). The integrative biology of type 2 diabetes. Nature 576, 51–60. [DOI] [PubMed] [Google Scholar]
- Rogan SC, and Roth BL (2011). Remote control of neuronal signaling. Pharmacol Rev 63, 291–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rossi M, Zhu L, McMillin SM, Pydi SP, Jain S, Wang L, Cui Y, Lee RJ, Cohen AH, Kaneto H, et al. (2018). Hepatic Gi signaling regulates whole-body glucose homeostasis. J Clin Invest 128, 746–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roth BL (2016). DREADDs for Neuroscientists. Neuron 89, 683–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saloman JL, Scheff NN, Snyder LM, Ross SE, Davis BM, and Gold MS (2016). Gi-DREADD Expression in Peripheral Nerves Produces Ligand-Dependent Analgesia, as well as Ligand-Independent Functional Changes in Sensory Neurons. J Neurosci 36, 10769–10781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saltiel AR, and Olefsky JM (2017). Inflammatory mechanisms linking obesity and metabolic disease. J Clin Invest 127, 1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sassmann A, Gier B, Gröne HJ, Drews G, Offermanns S, and Wettschureck N (2010). The Gq/G11-mediated signaling pathway is critical for autocrine potentiation of insulin secretion in mice. J Clin Invest 120, 2184–2193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schnell S, Schaefer M, and Schofl C (2007). Free fatty acids increase cytosolic free calcium and stimulate insulin secretion from beta-cells through activation of GPR40. Molecular and cellular endocrinology 263, 173–180. [DOI] [PubMed] [Google Scholar]
- Shukla AK, Xiao K, and Lefkowitz RJ (2011). Emerging paradigms of beta-arrestin-dependent seven transmembrane receptor signaling. Trends Biochem Sci 36, 457–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith JS, Lefkowitz RJ, and Rajagopal S (2018). Biased signalling: from simple switches to allosteric microprocessors. Nat Rev Drug Discov 17, 243–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spreckley E, and Murphy KG (2015). The L-Cell in Nutritional Sensing and the Regulation of Appetite. Front Nutr 2, 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sriram K, and Insel PA (2018). G Protein-Coupled Receptors as Targets for Approved Drugs: How Many Targets and How Many Drugs? Mol Pharmacol 93, 251–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steculorum SM, Ruud J, Karakasilioti I, Backes H, Engstrom Ruud L, Timper K, Hess ME, Tsaousidou E, Mauer J, Vogt MC, et al. (2016). AgRP Neurons Control Systemic Insulin Sensitivity via Myostatin Expression in Brown Adipose Tissue. Cell 165, 125–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sternson SM, and Eiselt AK (2017). Three Pillars for the Neural Control of Appetite. Annu Rev Physiol 79, 401–423. [DOI] [PubMed] [Google Scholar]
- Susulic VS, Frederich RC, Lawitts J, Tozzo E, Kahn BB, Harper ME, Himms-Hagen J, Flier JS, and Lowell BB (1995). Targeted disruption of the beta 3-adrenergic receptor gene. J Biol Chem 270, 29483–29492. [DOI] [PubMed] [Google Scholar]
- Svendsen B, Larsen O, Gabe MBN, Christiansen CB, Rosenkilde MM, Drucker DJ, and Holst JJ (2018). Insulin Secretion Depends on Intra-islet Glucagon Signaling. Cell reports 25, 1127–1134.e1122. [DOI] [PubMed] [Google Scholar]
- Thompson KJ, Khajehali E, Bradley SJ, Navarrete JS, Huang XP, Slocum S, Jin J, Liu J, Xiong Y, Olsen RHJ, et al. (2018). DREADD Agonist 21 Is an Effective Agonist for Muscarinic-Based DREADDs in Vitro and in Vivo. ACS Pharmacol Transl Sci 1, 61–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tomita T, Hosoda K, Fujikura J, Inagaki N, and Nakao K (2014). The G-Protein-Coupled Long-Chain Fatty Acid Receptor GPR40 and Glucose Metabolism. Frontiers in endocrinology 5, 152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uner AG, Kecik O, Quaresma PGF, De Araujo TM, Lee H, Li W, Kim HJ, Chung M, Bjorbaek C, and Kim YB (2019). Role of POMC and AgRP neuronal activities on glycaemia in mice. Sci Rep 9, 13068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Üner AG, Keçik O, Quaresma PGF, De Araujo TM, Lee H, Li W, Kim HJ, Chung M, Bjørbæk C, and Kim YB (2019). Role of POMC and AgRP neuronal activities on glycaemia in mice. Sci Rep 9, 13068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Unger RH, and Cherrington AD (2012). Glucagonocentric restructuring of diabetes: a pathophysiologic and therapeutic makeover. J Clin Invest 122, 4–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vardy E, Robinson JE, Li C, Olsen RH, DiBerto JF, Giguere PM, Sassano FM, Huang XP, Zhu H, Urban DJ, et al. (2015). A New DREADD Facilitates the Multiplexed Chemogenetic Interrogation of Behavior. Neuron 86, 936–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L, Pydi SP, Cui Y, Zhu L, Meister J, Gavrilova O, Berdeaux R, Fortin JP, Bence KK, Vernochet C, et al. (2019). Selective activation of Gs signaling in adipocytes causes striking metabolic improvements in mice. Molecular metabolism. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L, Pydi SP, Zhu L, Barella LF, Cui Y, Gavrilova O, Bence KK, Vernochet C, and Wess J (2020). Adipocyte G(i) signaling is essential for maintaining whole-body glucose homeostasis and insulin sensitivity. Nat Commun 11, 2995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wendt A, and Eliasson L (2020). Pancreatic alpha-cells - The unsung heroes in islet function. Semin Cell Dev Biol. [DOI] [PubMed] [Google Scholar]
- Wess J (2016). Use of Designer G Protein-Coupled Receptors to Dissect Metabolic Pathways. Trends in endocrinology and metabolism: TEM 27, 600–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wess J, Eglen RM, and Gautam D (2007). Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development. Nat Rev Drug Discov 6, 721–733. [DOI] [PubMed] [Google Scholar]
- Wess J, Nakajima K, and Jain S (2013). Novel designer receptors to probe GPCR signaling and physiology. Trends Pharmacol Sci 34, 385–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wettschureck N, and Offermanns S (2005). Mammalian G proteins and their cell type specific functions. Physiological reviews 85, 1159–1204. [DOI] [PubMed] [Google Scholar]
- Wootten D, Christopoulos A, Marti-Solano M, Babu MM, and Sexton PM (2018). Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat Rev Mol Cell Biol 19, 638–653. [DOI] [PubMed] [Google Scholar]
- Xiao C, Goldgof M, Gavrilova O, and Reitman ML (2015). Anti-obesity and metabolic efficacy of the beta3-adrenergic agonist, CL316243, in mice at thermoneutrality compared to 22 degrees C. Obesity (Silver Spring, Md) 23, 1450–1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie T, Chen M, Zhang QH, Ma Z, and Weinstein LS (2007). Beta cell-specific deficiency of the stimulatory G protein alpha-subunit Gsalpha leads to reduced beta cell mass and insulin-deficient diabetes. Proc Natl Acad Sci U S A 104, 19601–19606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xin Y, Kim J, Okamoto H, Ni M, Wei Y, Adler C, Murphy AJ, Yancopoulos GD, Lin C, and Gromada J (2016). RNA Sequencing of Single Human Islet Cells Reveals Type 2 Diabetes Genes. Cell Metab 24, 608–615. [DOI] [PubMed] [Google Scholar]
- Yadav PN, Abbas AI, Farrell MS, Setola V, Sciaky N, Huang XP, Kroeze WK, Crawford LK, Piel DA, Keiser MJ, et al. (2011). The presynaptic component of the serotonergic system is required for clozapine’s efficacy. Neuropsychopharmacology 36, 638–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhan C, Zhou J, Feng Q, Zhang JE, Lin S, Bao J, Wu P, and Luo M (2013). Acute and long-term suppression of feeding behavior by POMC neurons in the brainstem and hypothalamus, respectively. J Neurosci 33, 3624–3632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu H, Aryal DK, Olsen RH, Urban DJ, Swearingen A, Forbes S, Roth BL, and Hochgeschwender U (2016). Cre-dependent DREADD (Designer Receptors Exclusively Activated by Designer Drugs) mice. Genesis (New York, NY : 2000) 54, 439–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu L, Dattaroy D, Pham J, Wang L, Barella LF, Cui Y, Wilkins KJ, Roth BL, Hochgeschwender U, Matschinsky FM, et al. (2019). Intra-islet glucagon signaling is critical for maintaining glucose homeostasis. JCI insight 5. [DOI] [PMC free article] [PubMed] [Google Scholar]