Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 296: 100503, 2021.
Article in English | MEDLINE | ID: mdl-33684444

ABSTRACT

G protein-coupled receptors (GPCRs) signal through activation of G proteins and subsequent modulation of downstream effectors. More recently, signaling mediated by ß-arrestin has also been implicated in important physiological functions. This has led to great interest in the identification of biased ligands that favor either G protein or ß-arrestin-signaling pathways. However, nearly all screening techniques for measuring ß-arrestin recruitment have required C-terminal receptor modifications that can in principle alter protein interactions and thus signaling. Here, we have developed a novel luminescence-based assay to measure ß-arrestin recruitment to the membrane or early endosomes by unmodified receptors. Our strategy uses NanoLuc, an engineered luciferase from Oplophorus gracilirostris (deep-sea shrimp) that is smaller and brighter than other well-established luciferases. Recently, several publications have explored functional NanoLuc split sites for use in complementation assays. We have identified a unique split site within NanoLuc and fused the corresponding N-terminal fragment to either a plasma membrane or early endosome tether and the C-terminal fragment to ß-arrestins, which form the basis for the MeNArC and EeNArC assays, respectively. Upon receptor activation, ß-arrestin is recruited to the membrane and subsequently internalized in an agonist concentration-dependent manner. This recruitment promotes complementation of the two NanoLuc fragments, thereby reconstituting functional NanoLuc, allowing for quantification of ß-arrestin recruitment with a single luminescence signal. Our assay avoids potential artifacts related to C-terminal receptor modification and has promise as a new generic assay for measuring ß-arrestin recruitment to diverse GPCR types in heterologous or native cells.


Subject(s)
Cell Membrane/metabolism , Luciferases/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism , Biological Assay/methods , Cells, Cultured , Humans , Ligands , Protein Binding , Signal Transduction , beta-Arrestins/chemistry
2.
J Investig Med ; 65(8): 1116-1124, 2017 12.
Article in English | MEDLINE | ID: mdl-28784695

ABSTRACT

Critical aspects of maintaining glucose homeostasis in the face of chronic insulin resistance and type 2 diabetes (T2D) are increased insulin secretion and adaptive expansion of beta cell mass. Nutrient and hormone sensing G protein-coupled receptors are important mediators of these properties. A growing body of evidence now suggests that the G protein-coupled receptor, free fatty acid receptor 2 (FFA2), is capable of contributing to the maintenance of glucose homeostasis by acting at the pancreatic beta cell as well as at other metabolically active tissues. We have previously demonstrated that Gαq/11-biased agonism of FFA2 can potentiate glucose stimulated insulin secretion (GSIS) as well as promote beta cell proliferation. However, the currently available Gαq/11-biased agonists for FFA2 exhibit low potency, making them difficult to examine in vivo. This study sought to identify Gαq/11-biased FFA2-selective agonists with potent GSIS-stimulating effects. To do this, we generated an FFA2 homology model that was used to screen a library of 10 million drug-like compounds. Although FFA2 and the related short chain fatty acid receptor FFA3 share 52% sequence similarity, our virtual screen identified over 50 compounds with predicted selectivity and increased potency for FFA2 over FFA3. Subsequent in vitro calcium mobilization assays and GSIS assays resulted in the identification of a compound that can potentiate GSIS via activation of Gαq/11 with 100-fold increased potency compared with previously described Gαq/11-biased FFA2 agonists. These methods and findings provide a foundation for future discovery efforts to identify biased FFA2 agonists as potential T2D therapeutics.


Subject(s)
Insulin/metabolism , Molecular Docking Simulation , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Structural Homology, Protein , Animals , Binding Sites , Calcium/metabolism , Cell Line , Computer Simulation , Glucose/pharmacology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Ligands , Mice , Protein Structure, Secondary , Small Molecule Libraries/chemistry
3.
Sci Rep ; 6: 28159, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27324831

ABSTRACT

The regulation of pancreatic ß cell mass is a critical factor to help maintain normoglycemia during insulin resistance. Nutrient-sensing G protein-coupled receptors (GPCR) contribute to aspects of ß cell function, including regulation of ß cell mass. Nutrients such as free fatty acids (FFAs) contribute to precise regulation of ß cell mass by signaling through cognate GPCRs, and considerable evidence suggests that circulating FFAs promote ß cell expansion by direct and indirect mechanisms. Free Fatty Acid Receptor 2 (FFA2) is a ß cell-expressed GPCR that is activated by short chain fatty acids, particularly acetate. Recent studies of FFA2 suggest that it may act as a regulator of ß cell function. Here, we set out to explore what role FFA2 may play in regulation of ß cell mass. Interestingly, Ffar2(-/-) mice exhibit diminished ß cell mass at birth and throughout adulthood, and increased ß cell death at adolescent time points, suggesting a role for FFA2 in establishment and maintenance of ß cell mass. Additionally, activation of FFA2 with Gαq/11-biased agonists substantially increased ß cell proliferation in in vitro and ex vivo proliferation assays. Collectively, these data suggest that FFA2 may be a novel therapeutic target to stimulate ß cell growth and proliferation.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Pancreas/pathology , Receptors, Cell Surface/metabolism , Animals , Cell Survival , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Volatile/metabolism , Humans , Insulin Resistance , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Signal Transduction
4.
Mol Endocrinol ; 29(7): 1055-66, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26075576

ABSTRACT

G protein-coupled receptors have been well described to contribute to the regulation of glucose-stimulated insulin secretion (GSIS). The short-chain fatty acid-sensing G protein-coupled receptor, free fatty acid receptor 2 (FFAR2), is expressed in pancreatic ß-cells, and in rodents, its expression is altered during insulin resistance. Thus, we explored the role of FFAR2 in regulating GSIS. First, assessing the phenotype of wild-type and Ffar2(-/-) mice in vivo, we observed no differences with regard to glucose homeostasis on normal or high-fat diet, with a marginally significant defect in insulin secretion in Ffar2(-/-) mice during hyperglycemic clamps. In ex vivo insulin secretion studies, we observed diminished GSIS from Ffar2(-/-) islets relative to wild-type islets under high-glucose conditions. Further, in the presence of acetate, the primary endogenous ligand for FFAR2, we observed FFAR2-dependent potentiation of GSIS, whereas FFAR2-specific agonists resulted in either potentiation or inhibition of GSIS, which we found to result from selective signaling through either Gαq/11 or Gαi/o, respectively. Lastly, in ex vivo insulin secretion studies of human islets, we observed that acetate and FFAR2 agonists elicited different signaling properties at human FFAR2 than at mouse FFAR2. Taken together, our studies reveal that FFAR2 signaling occurs by divergent G protein pathways that can selectively potentiate or inhibit GSIS in mouse islets. Further, we have identified important differences in the response of mouse and human FFAR2 to selective agonists, and we suggest that these differences warrant consideration in the continued investigation of FFAR2 as a novel type 2 diabetes target.


Subject(s)
Acetates/metabolism , Insulin/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Diet, High-Fat , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Glucose Clamp Technique , Glucose Tolerance Test , Humans , Insulin/pharmacology , Insulin Secretion , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Cell Surface/agonists , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/deficiency , Signal Transduction/drug effects , Species Specificity
5.
J Med Chem ; 53(9): 3502-16, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20380377

ABSTRACT

The inhibition of LTB(4) binding to and activation of G-protein-coupled receptors BLT1 and BLT2 is the premise of a treatment for several inflammatory diseases. In a lead optimization effort starting with the leukotriene B(4) (LTB(4)) receptor antagonist (2), members of a series of 3,5-diarylphenyl ethers were found to be highly potent inhibitors of LTB(4) binding to BLT1 and BLT2 receptors, with varying levels of selectivity depending on the substitution. In addition, compounds 33 and 38 from this series have good in vitro ADME properties, good oral bioavailability, and efficacy after oral delivery in guinea pig LTB(4) and nonhuman primate allergen challenge models. Further profiling in a rat non-GLP toxicity experiment provided the rationale for differentiation and selection of one compound (33) for clinical development.


Subject(s)
Drug Discovery , Leukotriene Antagonists/chemistry , Phenyl Ethers/pharmacology , Receptors, Leukotriene B4/antagonists & inhibitors , Animals , Drug Evaluation, Preclinical , Guinea Pigs , HL-60 Cells , Humans , Leukotriene Antagonists/pharmacology , Phenyl Ethers/chemistry , Primates , Protein Binding , Rats , Receptors, G-Protein-Coupled/metabolism , Receptors, Leukotriene B4/metabolism , Structure-Activity Relationship
6.
Prostaglandins Other Lipid Mediat ; 92(1-4): 33-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20214997

ABSTRACT

Asthma, chronic obstructive pulmonary disease (COPD) and acute lung injury/acute respiratory distress syndrome (ALI/ARDS) are characterized by neutrophilic inflammation and elevated levels of leukotriene B4 (LTB4). However, the exact role of LTB4 pathways in mediating pulmonary neutrophilia and the potential therapeutic application of LTB4 receptor antagonists in these diseases remains controversial. Here we show that a novel dual BLT1 and BLT2 receptor antagonist, RO5101576, potently inhibited LTB4-evoked calcium mobilization in HL-60 cells and chemotaxis of human neutrophils. RO5101576 significantly attenuated LTB4-evoked pulmonary eosinophilia in guinea pigs. In non-human primates, RO5101576 inhibited allergen and ozone-evoked pulmonary neutrophilia, with comparable efficacy to budesonide (allergic responses). RO5101576 had no effects on LPS-evoked neutrophilia in guinea pigs and cigarette smoke-evoked neutrophilia in mice and rats. In toxicology studies RO5101576 was well-tolerated. Theses studies show differential effects of LTB4 receptor antagonism on neutrophil responses in vivo and suggest RO5101576 may represent a potential new treatment for pulmonary neutrophilia in asthma.


Subject(s)
Benzodioxoles/pharmacology , Phenylpropionates/pharmacology , Pneumonia/drug therapy , Primates , Receptors, Leukotriene B4/antagonists & inhibitors , Animals , Benzodioxoles/therapeutic use , Benzodioxoles/toxicity , Dogs , Drug-Related Side Effects and Adverse Reactions , Female , Guinea Pigs , HL-60 Cells , Humans , Hypersensitivity/complications , Lipopolysaccharides/pharmacology , Lung/drug effects , Male , Mice , Ozone/pharmacology , Phenylpropionates/therapeutic use , Phenylpropionates/toxicity , Pneumonia/chemically induced , Pneumonia/complications , Pneumonia/metabolism , Rats , Receptors, Leukotriene B4/metabolism , Smoking/adverse effects , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...