Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
1.
Trends Pharmacol Sci ; 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38853101

ABSTRACT

The M2 muscarinic receptor (M2R) is a prototypic class A G protein-coupled receptor (GPCR). Interestingly, Fasciani et al. recently identified an internal translation start site within the M2 receptor mRNA, directing the expression of a C-terminal receptor fragment. Elevated during cellular stress, this polypeptide localizes to mitochondria where it inhibits oxidative phosphorylation.

2.
Nat Commun ; 15(1): 5129, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879678

ABSTRACT

Glucagon, a hormone released from pancreatic α-cells, is critical for maintaining euglycemia and plays a key role in the pathophysiology of diabetes. To stimulate the development of new classes of therapeutic agents targeting glucagon release, key α-cell signaling pathways that regulate glucagon secretion need to be identified. Here, we focused on the potential importance of α-cell Gs signaling on modulating α-cell function. Studies with α-cell-specific mouse models showed that activation of α-cell Gs signaling causes a marked increase in glucagon secretion. We also found that intra-islet adenosine plays an unexpected autocrine/paracrine role in promoting glucagon release via activation of α-cell Gs-coupled A2A adenosine receptors. Studies with α-cell-specific Gαs knockout mice showed that α-cell Gs also plays an essential role in stimulating the activity of the Gcg gene, thus ensuring proper islet glucagon content. Our data suggest that α-cell enriched Gs-coupled receptors represent potential targets for modulating α-cell function for therapeutic purposes.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs , Glucagon-Secreting Cells , Glucagon , Mice, Knockout , Signal Transduction , Glucagon/metabolism , Animals , Glucagon-Secreting Cells/metabolism , Mice , GTP-Binding Protein alpha Subunits, Gs/metabolism , Adenosine/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/genetics , Male , Mice, Inbred C57BL , Islets of Langerhans/metabolism
3.
Mol Metab ; 79: 101866, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38159884

ABSTRACT

OBJECTIVE: G-protein-coupled receptor (GPCR) kinases (GRKs) abrogate GPCR signaling by promoting receptor desensitization and internalization. Accumulating evidence suggests that GRK2 represents an important regulator of GPCR-mediated effects on systemic glucose metabolism, obesity, and insulin resistance. Despite the key role of the liver in maintaining euglycemia, the potential metabolic relevance of hepatic GRK2 has yet to be examined. Thus, the goal of this study was to explore the potential role of hepatic GRK2 in maintaining glucose homeostasis and other key metabolic functions. METHODS: To address this question, we generated mice that showed a ∼90% reduction in GRK2 protein expression selectively in hepatocytes (Hep-GRK2-KO mice) and subjected these mice, together with their control littermates, to systematic metabolic phenotyping studies. RESULTS: We found that Hep-GRK2-KO mice maintained on regular chow did not differ significantly from their control littermates in glycemia, glucose tolerance, insulin sensitivity, in vivo gluconeogenesis, and glucagon-induced hyperglycemia. We obtained similar findings when we analyzed Hep-GRK2-KO mice and control littermates consuming an obesogenic high-fat diet. Likewise, plasma levels of insulin, glucagon, free fatty acids, and ketone bodies remained unaffected by the lack of hepatocyte GRK2. The same was true when we examined the expression levels of key genes regulating hepatic glucose and fatty acid metabolism. CONCLUSION: In summary, our data suggest that hepatocyte GRK2 is dispensable for systemic glucose homeostasis and other key metabolic functions in both lean and obese mice. This finding suggests that drug development efforts aimed at inhibiting GRK2 to improve impaired glucose homeostasis and insulin sensitivity need to focus on other metabolically important tissues.


Subject(s)
Insulin Resistance , Animals , Mice , Glucagon/metabolism , Glucose/metabolism , Homeostasis , Insulin Resistance/physiology , Liver/metabolism
5.
Am J Physiol Renal Physiol ; 325(5): F618-F628, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37675459

ABSTRACT

Acetylcholine plays an essential role in the regulation of detrusor muscle contractions, and antimuscarinics are widely used in the management of overactive bladder syndrome. However, several adverse effects limit their application and patients' compliance. Thus, this study aimed to further analyze the signal transduction of M2 and M3 receptors in the murine urinary bladder to eventually find more specific therapeutic targets. Experiments were performed on adult male wild-type, M2, M3, M2/M3, or Gαq/11 knockout (KO), and pertussis toxin (PTX)-treated mice. Contraction force and RhoA activity were measured in the urinary bladder smooth muscle (UBSM). Our results indicate that carbamoylcholine (CCh)-induced contractions were associated with increased activity of RhoA and were reduced in the presence of the Rho-associated kinase (ROCK) inhibitor Y-27632 in UBSM. CCh-evoked contractile responses and RhoA activation were markedly reduced in detrusor strips lacking either M2 or M3 receptors and abolished in M2/M3 KO mice. Inhibition of Gαi-coupled signaling by PTX treatment shifted the concentration-response curve of CCh to the right and diminished RhoA activation. CCh-induced contractile responses were markedly decreased in Gαq/11 KO mice; however, RhoA activation was unaffected. In conclusion, cholinergic detrusor contraction and RhoA activation are mediated by both M2 and M3 receptors. Furthermore, whereas both Gαi and Gαq/11 proteins mediate UBSM contraction, the activation at the RhoA-ROCK pathway appears to be linked specifically to Gαi. These findings may aid the identification of more specific therapeutic targets for bladder dysfunctions.NEW & NOTEWORTHY Muscarinic acetylcholine receptors are of utmost importance in physiological regulation of micturition and also in the development of voiding disorders. We demonstrate that the RhoA-Rho-associated kinase (ROCK) pathway plays a crucial role in contractions induced by cholinergic stimulation in detrusor muscle. Activation of RhoA is mediated by both M2 and M3 receptors as well as by Gi but not Gq/11 proteins. The Gi-RhoA-ROCK pathway may provide a novel therapeutic target for overactive voiding disorders.

6.
Cells ; 12(14)2023 07 19.
Article in English | MEDLINE | ID: mdl-37508553

ABSTRACT

Muscarinic acetylcholine receptors are well-known for their crucial involvement in hippocampus-dependent learning and memory, but the exact roles of the various receptor subtypes (M1-M5) are still not fully understood. Here, we studied how M1 and M3 receptors affect plasticity at the mossy fiber (MF)-CA3 pyramidal cell synapse. In hippocampal slices from M1/M3 receptor double knockout (M1/M3-dKO) mice, the signature short-term plasticity of the MF-CA3 synapse was not significantly affected. However, the rather unique NMDA receptor-independent and presynaptic form of long-term potentiation (LTP) of this synapse was much larger in M1/M3-deficient slices compared to wild-type slices in both field potential and whole-cell recordings. Consistent with its presynaptic origin, induction of MF-LTP strongly enhanced the excitatory drive onto single CA3 pyramidal cells, with the effect being more pronounced in M1/M3-dKO cells. In an earlier study, we found that the deletion of M2 receptors in mice disinhibits MF-LTP in a similar fashion, suggesting that endogenous acetylcholine employs both M1/M3 and M2 receptors to constrain MF-LTP. Importantly, such synergism was not observed for MF long-term depression (LTD). Low-frequency stimulation, which reliably induced LTD of MF synapses in control slices, failed to do so in M1/M3-dKO slices and gave rise to LTP instead. In striking contrast, loss of M2 receptors augmented LTD when compared to control slices. Taken together, our data demonstrate convergence of M1/M3 and M2 receptors on MF-LTP, but functional divergence on MF-LTD, with the net effect resulting in a well-balanced bidirectional plasticity of the MF-CA3 pyramidal cell synapse.


Subject(s)
Acetylcholine , Mossy Fibers, Hippocampal , Mice , Animals , Mossy Fibers, Hippocampal/physiology , Receptor, Muscarinic M1 , Mice, Knockout , Hippocampus , Pyramidal Cells/physiology , Receptor, Muscarinic M2/genetics
7.
Diabetes ; 72(7): 825-834, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37339353

ABSTRACT

The functional state of adipocytes plays a central role in regulating numerous important metabolic functions, including energy and glucose homeostasis. While white adipocytes store excess calories as fat (triglycerides) and release free fatty acids as a fuel source in times of need, brown and beige adipocytes (so-called thermogenic adipocytes) convert chemical energy stored in substrates (e.g., fatty acids or glucose) into heat, thus promoting energy expenditure. Like all other cell types, adipocytes express many G protein-coupled receptors (GPCRs) that are linked to four major functional classes of heterotrimeric G proteins (Gs, Gi/o, Gq/11, and G12/13). During the past few years, novel experimental approaches, including the use of chemogenetic strategies, have led to a series of important new findings regarding the metabolic consequences of activating or inhibiting distinct GPCR/G protein signaling pathways in white, brown, and beige adipocytes. This novel information should guide the development of novel drugs capable of modulating the activity of specific adipocyte GPCR signaling pathways for the treatment of obesity, type 2 diabetes, and related metabolic disorders.


Subject(s)
Adipocytes, Beige , Diabetes Mellitus, Type 2 , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Receptors, G-Protein-Coupled/metabolism , Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Glucose/metabolism , Thermogenesis , Adipocytes, Brown/metabolism , Energy Metabolism
8.
Trends Pharmacol Sci ; 44(8): 492-494, 2023 08.
Article in English | MEDLINE | ID: mdl-37208206

ABSTRACT

The third intracellular loop of G-protein-coupled receptors (GPCRs) shows remarkable diversity in sequence and overall length. Sadler and colleagues recently demonstrated that this domain acts as an 'autoregulator' of receptor activity and that its length contributes to receptor/G-protein coupling selectivity. These observations may prove useful for developing novel therapeutics.


Subject(s)
Receptors, G-Protein-Coupled , Humans , Amino Acid Sequence
9.
Pharmacol Rev ; 75(5): 854-884, 2023 09.
Article in English | MEDLINE | ID: mdl-37028945

ABSTRACT

The two ß-arrestins, ß-arrestin-1 and -2 (systematic names: arrestin-2 and -3, respectively), are multifunctional intracellular proteins that regulate the activity of a very large number of cellular signaling pathways and physiologic functions. The two proteins were discovered for their ability to disrupt signaling via G protein-coupled receptors (GPCRs) via binding to the activated receptors. However, it is now well recognized that both ß-arrestins can also act as direct modulators of numerous cellular processes via either GPCR-dependent or -independent mechanisms. Recent structural, biophysical, and biochemical studies have provided novel insights into how ß-arrestins bind to activated GPCRs and downstream effector proteins. Studies with ß-arrestin mutant mice have identified numerous physiologic and pathophysiological processes regulated by ß-arrestin-1 and/or -2. Following a short summary of recent structural studies, this review primarily focuses on ß-arrestin-regulated physiologic functions, with particular focus on the central nervous system and the roles of ß-arrestins in carcinogenesis and key metabolic processes including the maintenance of glucose and energy homeostasis. This review also highlights potential therapeutic implications of these studies and discusses strategies that could prove useful for targeting specific ß-arrestin-regulated signaling pathways for therapeutic purposes. SIGNIFICANCE STATEMENT: The two ß-arrestins, structurally closely related intracellular proteins that are evolutionarily highly conserved, have emerged as multifunctional proteins able to regulate a vast array of cellular and physiological functions. The outcome of studies with ß-arrestin mutant mice and cultured cells, complemented by novel insights into ß-arrestin structure and function, should pave the way for the development of novel classes of therapeutically useful drugs capable of regulating specific ß-arrestin functions.


Subject(s)
Arrestins , Signal Transduction , Mice , Animals , beta-Arrestins/metabolism , Arrestins/chemistry , Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/metabolism
10.
Nucleic Acids Res ; 50(16): 9195-9211, 2022 09 09.
Article in English | MEDLINE | ID: mdl-36018801

ABSTRACT

Enhancers establish proximity with distant target genes to regulate temporospatial gene expression and specify cell identity. Lim domain binding protein 1 (LDB1) is a conserved and widely expressed protein that functions as an enhancer looping factor. Previous studies in erythroid cells and neuronal cells showed that LDB1 forms protein complexes with different transcription factors to regulate cell-specific gene expression. Here, we show that LDB1 regulates expression of liver genes by occupying enhancer elements and cooperating with hepatic transcription factors HNF4A, FOXA1, TCF7 and GATA4. Using the glucose transporter SLC2A2 gene, encoding GLUT2, as an example, we find that LDB1 regulates gene expression by mediating enhancer-promoter interactions. In vivo, we find that LDB1 deficiency in primary mouse hepatocytes dysregulates metabolic gene expression and changes the enhancer landscape. Conditional deletion of LDB1 in adult mouse liver induces glucose intolerance. However, Ldb1 knockout hepatocytes show improved liver pathology under high-fat diet conditions associated with increased expression of genes related to liver fatty acid metabolic processes. Thus, LDB1 is linked to liver metabolic functions under normal and obesogenic conditions.


Subject(s)
DNA-Binding Proteins , Transcription Factors , Mice , Animals , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM Domain Proteins/metabolism , Gene Expression , Hepatocytes/metabolism , Liver/metabolism
11.
Nat Commun ; 13(1): 1652, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35351896

ABSTRACT

Obesity is the major driver of the global epidemic in type 2 diabetes (T2D). In individuals with obesity, impaired insulin action leads to increased lipolysis in adipocytes, resulting in elevated plasma free fatty acid (FFA) levels that promote peripheral insulin resistance, a hallmark of T2D. Here we show, by using a combined genetic/biochemical/pharmacologic approach, that increased adipocyte lipolysis can be prevented by selective activation of adipocyte Gq signaling in vitro and in vivo (in mice). Activation of this pathway by a Gq-coupled designer receptor or by an agonist acting on an endogenous adipocyte Gq-coupled receptor (CysLT2 receptor) greatly improved glucose and lipid homeostasis in obese mice or in mice with adipocyte insulin receptor deficiency. Our findings identify adipocyte Gq signaling as an essential regulator of whole-body glucose and lipid homeostasis and should inform the development of novel classes of GPCR-based antidiabetic drugs.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Adipocytes/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Homeostasis , Lipids , Lipolysis , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism
12.
Int J Mol Sci ; 23(1)2022 Jan 02.
Article in English | MEDLINE | ID: mdl-35008921

ABSTRACT

The two ß-arrestins (ß-arrestin-1 and -2; alternative names: arrestin-2 and -3, respectively) are well known for their ability to inhibit signaling via G protein-coupled receptors. However, ß-arrestins can also act as signaling molecules in their own right. Although the two proteins share a high degree of sequence and structural homology, early studies with cultured cells indicated that ß-arrestin-1 and -2 are not functionally redundant. Recently, the in vivo metabolic roles of the two ß-arrestins have been studied using mutant mice selectively lacking either ß-arrestin-1 or -2 in cell types that are of particular relevance for regulating glucose and energy homeostasis. These studies demonstrated that the ß-arrestin-1 and -2 mutant mice displayed distinct metabolic phenotypes in vivo, providing further evidence for the functional heterogeneity of these two highly versatile signaling proteins.


Subject(s)
Signal Transduction , beta-Arrestin 1/metabolism , beta-Arrestin 2/metabolism , Animals , Diabetes Mellitus/metabolism , Disease Models, Animal , Energy Metabolism , Glucose/metabolism , Mice , Obesity/metabolism , Receptors, G-Protein-Coupled/metabolism
13.
Nat Commun ; 13(1): 22, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013148

ABSTRACT

Activation of the sympathetic nervous system causes pronounced metabolic changes that are mediated by multiple adrenergic receptor subtypes. Systemic treatment with ß2-adrenergic receptor agonists results in multiple beneficial metabolic effects, including improved glucose homeostasis. To elucidate the underlying cellular and molecular mechanisms, we chronically treated wild-type mice and several newly developed mutant mouse strains with clenbuterol, a selective ß2-adrenergic receptor agonist. Clenbuterol administration caused pronounced improvements in glucose homeostasis and prevented the metabolic deficits in mouse models of ß-cell dysfunction and insulin resistance. Studies with skeletal muscle-specific mutant mice demonstrated that these metabolic improvements required activation of skeletal muscle ß2-adrenergic receptors and the stimulatory G protein, Gs. Unbiased transcriptomic and metabolomic analyses showed that chronic ß2-adrenergic receptor stimulation caused metabolic reprogramming of skeletal muscle characterized by enhanced glucose utilization. These findings strongly suggest that agents targeting skeletal muscle metabolism by modulating ß2-adrenergic receptor-dependent signaling pathways may prove beneficial as antidiabetic drugs.


Subject(s)
Cellular Reprogramming/drug effects , Clenbuterol/pharmacology , Hypoglycemic Agents/pharmacology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Animals , Biochemical Phenomena , Clenbuterol/metabolism , Female , Glucose/metabolism , Homeostasis , Insulin Resistance , Male , Metabolic Diseases , Metabolomics , Mice , Mice, Knockout , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction
14.
Annu Rev Physiol ; 84: 17-40, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34705480

ABSTRACT

ß-Arrestin-1 and -2 (also known as arrestin-2 and -3, respectively) are ubiquitously expressed cytoplasmic proteins that dampen signaling through G protein-coupled receptors. However, ß-arrestins can also act as signaling molecules in their own right. To investigate the potential metabolic roles of the two ß-arrestins in modulating glucose and energy homeostasis, recent studies analyzed mutant mice that lacked or overexpressed ß-arrestin-1 and/or -2 in distinct, metabolically important cell types. Metabolic analysis of these mutant mice clearly demonstrated that both ß-arrestins play key roles in regulating the function of most of these cell types, resulting in striking changes in whole-body glucose and/or energy homeostasis. These studies also revealed that ß-arrestin-1 and -2, though structurally closely related, clearly differ in their metabolic roles under physiological and pathophysiological conditions. These new findings should guide the development of novel drugs for the treatment of various metabolic disorders, including type 2 diabetes and obesity.


Subject(s)
Diabetes Mellitus, Type 2 , Glucose , Animals , Glucose/metabolism , Homeostasis , Humans , Mice , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism
15.
Mol Metab ; 55: 101415, 2022 01.
Article in English | MEDLINE | ID: mdl-34883278

ABSTRACT

OBJECTIVE: The goal of this study was to determine the glucometabolic effects of acute activation of Gs signaling in skeletal muscle (SKM) in vivo and its contribution to whole-body glucose homeostasis. METHODS: To address this question, we studied mice that express a Gs-coupled designer G protein-coupled receptor (Gs-DREADD or GsD) selectively in skeletal muscle. We also identified two Gs-coupled GPCRs that are endogenously expressed by SKM at relatively high levels (ß2-adrenergic receptor and CRF2 receptor) and studied the acute metabolic effects of activating these receptors in vivo by highly selective agonists (clenbuterol and urocortin 2 (UCN2), respectively). RESULTS: Acute stimulation of GsD signaling in SKM impaired glucose tolerance in lean and obese mice by decreasing glucose uptake selectively into SKM. The acute metabolic effects following agonist activation of ß2-adrenergic and, potentially, CRF2 receptors appear primarily mediated by altered insulin release. Clenbuterol injection improved glucose tolerance by increasing insulin secretion in lean mice. In SKM, clenbuterol stimulated glycogen breakdown. UCN2 injection resulted in decreased glucose tolerance associated with lower plasma insulin levels. The acute metabolic effects of UCN2 were not mediated by SKM Gs signaling. CONCLUSIONS: Selective activation of Gs signaling in SKM causes an acute increase in blood glucose levels. However, acute in vivo stimulation of endogenous Gs-coupled receptors enriched in SKM has only a limited impact on whole-body glucose homeostasis, most likely due to the fact that these receptors are also expressed by pancreatic islets where they modulate insulin release.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , Muscle, Skeletal/metabolism , Signal Transduction/drug effects , Animals , Clenbuterol/pharmacology , Diabetes Mellitus, Type 2/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/physiology , Glucose/metabolism , Glucose Intolerance/metabolism , Homeostasis/drug effects , Insulin/metabolism , Insulin Resistance/physiology , Insulin Secretion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/physiology , Obesity/metabolism , Receptors, Adrenergic, beta-2/metabolism
16.
Endocrinology ; 163(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34871353

ABSTRACT

G protein-coupled receptors (GPCRs) are the target of ~30% to 35% of all US Food and Drug Administration-approved drugs. The individual members of the GPCR superfamily couple to 1 or more functional classes of heterotrimeric G proteins. The physiological outcome of activating a particular GPCR in vivo depends on the pattern of receptor distribution and the type of G proteins activated by the receptor. Based on the structural and functional properties of their α-subunits, heterotrimeric G proteins are subclassified into 4 major families: Gs, Gi/o, Gq/11, and G12/13. Recent studies with genetically engineered mice have yielded important novel insights into the metabolic roles of Gi/o-type G proteins. For example, recent data indicate that Gi signaling in pancreatic α-cells plays a key role in regulating glucagon release and whole body glucose homeostasis. Receptor-mediated activation of hepatic Gi signaling stimulates hepatic glucose production, suggesting that inhibition of hepatic Gi signaling could prove clinically useful to reduce pathologically elevated blood glucose levels. Activation of adipocyte Gi signaling reduces plasma free fatty acid levels, thus leading to improved insulin sensitivity in obese, glucose-intolerant mice. These new data suggest that Gi-coupled receptors that are enriched in metabolically important cell types represent potential targets for the development of novel drugs useful for the treatment of type 2 diabetes and related metabolic disorders.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Proteins/metabolism , Glucose/metabolism , Homeostasis , Models, Animal , Signal Transduction , Adipocytes/metabolism , Animals , Humans , Mice
17.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34752420

ABSTRACT

Glucagon, a hormone released from pancreatic α cells, plays a key role in maintaining euglycemia. New insights into the signaling pathways that control glucagon secretion may stimulate the development of novel therapeutic agents. In this study, we investigated the potential regulation of α cell function by G proteins of the Gq family. The use of a chemogenetic strategy allowed us to selectively activate Gq signaling in mouse α cells in vitro and in vivo. Acute stimulation of α cell Gq signaling led to elevated plasma glucagon levels, accompanied by increased insulin release and improved glucose tolerance. Moreover, chronic activation of this pathway greatly improved glucose tolerance in obese mice. We also identified an endogenous Gq-coupled receptor (vasopressin 1b receptor; V1bR) that was enriched in mouse and human α cells. Agonist-induced activation of the V1bR strongly stimulated glucagon release in a Gq-dependent fashion. In vivo studies indicated that V1bR-mediated glucagon release played a key role in the counterregulatory hyperglucagonemia under hypoglycemic and glucopenic conditions. These data indicate that α cell Gq signaling represents an important regulator of glucagon secretion, resulting in multiple beneficial metabolic effects. Thus, drugs that target α cell-enriched Gq-coupled receptors may prove useful to restore euglycemia in various pathophysiological conditions.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hypoglycemic Agents/metabolism , Signal Transduction/immunology , Animals , Humans , Male , Mice
19.
Biochem Pharmacol ; 192: 114739, 2021 10.
Article in English | MEDLINE | ID: mdl-34418353

ABSTRACT

Hepatic insulin resistance (IR) and enhanced hepatic glucose production (HGP) are key features of type 2 diabetes (T2D), contributing to fasting hyperglycemia. Adenosine receptors (ARs) are G protein-coupled and expressed in hepatocytes. Here, we explored the role of hepatic Gi/o-coupled A1AR on insulin resistance and glucose fluxes associated with obesity. We generated a mouse model with hepatocyte-specific deletion of A1AR (A1LΔ/Δ), which was compared with whole body knockout of A1AR or A1AR/A3AR (both Gi-coupled). Selective deletion of hepatic A1AR resulted in a modest improvement in insulin sensitivity. In addition, HFD A1LΔ/Δ mice showed decreased fasting glucose levels. Hyperinsulinemic-euglycemic clamp studies demonstrated enhanced insulin sensitivity with no change in HGP in HFD A1LΔ/Δ mice. Similar to A1LΔ/Δ, fasting blood glucose levels were significantly reduced in whole body A1Δ/Δ and A1Δ/ΔA3Δ/Δ compared to wild-type mice. Taken together, our data support the concept that blocking hepatic A1AR may decrease fasting blood glucose levels without directly affecting hepatocyte glucose metabolism and insulin sensitivity.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Hepatocytes/metabolism , Insulin Resistance/physiology , Receptor, Adenosine A1/deficiency , Animals , Diabetes Mellitus, Experimental/genetics , Diet, High-Fat/adverse effects , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A1/genetics
20.
Nat Commun ; 12(1): 3385, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099679

ABSTRACT

Obesity is the key driver of peripheral insulin resistance, one of the key features of type 2 diabetes (T2D). In insulin-resistant individuals, the expansion of beta-cell mass is able to delay or even prevent the onset of overt T2D. Here, we report that beta-arrestin-1 (barr1), an intracellular protein known to regulate signaling through G protein-coupled receptors, is essential for beta-cell replication and function in insulin-resistant mice maintained on an obesogenic diet. Specifically, insulin-resistant beta-cell-specific barr1 knockout mice display marked reductions in beta-cell mass and the rate of beta-cell proliferation, associated with pronounced impairments in glucose homeostasis. Mechanistic studies suggest that the observed metabolic deficits are due to reduced Pdx1 expression levels caused by beta-cell barr1 deficiency. These findings indicate that strategies aimed at enhancing barr1 activity and/or expression in beta-cells may prove useful to restore proper glucose homeostasis in T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/pathology , Obesity/metabolism , beta-Arrestin 1/metabolism , Animals , Blood Glucose/metabolism , Cell Proliferation , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Homeodomain Proteins/metabolism , Humans , Insulin Resistance , Male , Mice , Mice, Knockout , Obesity/etiology , Obesity/pathology , Trans-Activators/metabolism , beta-Arrestin 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...