Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Islets ; 15(1): 2267725, 2023 12 31.
Article in English | MEDLINE | ID: mdl-37838950

ABSTRACT

The pancreatic ß cell synthesizes, packages, and secretes insulin in response to glucose-stimulation to maintain blood glucose homeostasis. Under diabetic conditions, a subset of ß cells fail and lose expression of key transcription factors (TFs) required for insulin secretion. Among these TFs is Pancreatic and duodenal homeobox 1 (PDX1), which recruits a unique subset of transcriptional coregulators to modulate its activity. Here we describe a novel interacting partner of PDX1, the Staphylococcal Nuclease and Tudor domain-containing protein (SND1), which has been shown to facilitate protein-protein interactions and transcriptional control through diverse mechanisms in a variety of tissues. PDX1:SND1 interactions were confirmed in rodent ß cell lines, mouse islets, and human islets. Utilizing CRISPR-Cas9 gene editing technology, we deleted Snd1 from the mouse ß cell lines, which revealed numerous differentially expressed genes linked to insulin secretion and cell proliferation, including limited expression of Glp1r. We observed Snd1 deficient ß cell lines had reduced cell expansion rates, GLP1R protein levels, and limited cAMP accumulation under stimulatory conditions, and further show that acute ablation of Snd1 impaired insulin secretion in rodent and human ß cell lines. Lastly, we discovered that PDX1:SND1 interactions were profoundly reduced in human ß cells from donors with type 2 diabetes (T2D). These observations suggest the PDX1:SND1 complex formation is critical for controlling a subset of genes important for ß cell function and is targeted in diabetes pathogenesis.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Animals , Humans , Mice , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Gene Expression , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tudor Domain
2.
Cell Rep ; 38(1): 110179, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986353

ABSTRACT

G protein-coupled receptors (GPCRs) in intestinal enteroendocrine cells (EECs) respond to nutritional, neural, and microbial cues and modulate the release of gut hormones. Here we show that Gpr17, an orphan GPCR, is co-expressed in glucagon-like peptide-1 (GLP-1)-expressing EECs in human and rodent intestinal epithelium. Acute genetic ablation of Gpr17 in intestinal epithelium improves glucose tolerance and glucose-stimulated insulin secretion (GSIS). Importantly, inducible knockout (iKO) mice and Gpr17 null intestinal organoids respond to glucose or lipid ingestion with increased secretion of GLP-1, but not the other incretin glucose-dependent insulinotropic polypeptide (GIP). In an in vitro EEC model, overexpression or agonism of Gpr17 reduces voltage-gated calcium currents and decreases cyclic AMP (cAMP) production, and these are two critical factors regulating GLP-1 secretion. Together, our work shows that intestinal Gpr17 signaling functions as an inhibitory pathway for GLP-1 secretion in EECs, suggesting intestinal GPR17 is a potential target for diabetes and obesity intervention.


Subject(s)
Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Intestinal Mucosa/metabolism , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Blood Glucose/analysis , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Diabetes Mellitus/pathology , Female , Gastric Inhibitory Polypeptide/metabolism , Glucose Tolerance Test , HEK293 Cells , HeLa Cells , Humans , Incretins/metabolism , Insulin/metabolism , Insulin Secretion/physiology , Intestinal Mucosa/cytology , Male , Mice , Mice, Knockout , Obesity/pathology , Receptors, Gastrointestinal Hormone/metabolism
3.
J Biol Chem ; 298(1): 101431, 2022 01.
Article in English | MEDLINE | ID: mdl-34801552

ABSTRACT

Insulin resistance impairs postprandial glucose uptake through glucose transporter type 4 (GLUT4) and is the primary defect preceding type 2 diabetes. We previously generated an insulin-resistant mouse model with human GLUT4 promoter-driven insulin receptor knockout (GIRKO) in the muscle, adipose, and neuronal subpopulations. However, the rate of diabetes in GIRKO mice remained low prior to 6 months of age on normal chow diet (NCD), suggesting that additional factors/mechanisms are responsible for adverse metabolic effects driving the ultimate progression of overt diabetes. In this study, we characterized the metabolic phenotypes of the adult GIRKO mice acutely switched to high-fat diet (HFD) feeding in order to identify additional metabolic challenges required for disease progression. Distinct from other diet-induced obesity (DIO) and genetic models (e.g., db/db mice), GIRKO mice remained leaner on HFD feeding, but developed other cardinal features of insulin resistance syndrome. GIRKO mice rapidly developed hyperglycemia despite compensatory increases in ß-cell mass and hyperinsulinemia. Furthermore, GIRKO mice also had impaired oral glucose tolerance and a limited glucose-lowering benefit from exendin-4, suggesting that the blunted incretin effect contributed to hyperglycemia. Secondly, GIRKO mice manifested severe dyslipidemia while on HFD due to elevated hepatic lipid secretion, serum triglyceride concentration, and lipid droplet accumulation in hepatocytes. Thirdly, GIRKO mice on HFD had increased inflammatory cues in the gut, which were associated with the HFD-induced microbiome alterations and increased serum lipopolysaccharide (LPS). In conclusion, our studies identified important gene/diet interactions contributing to diabetes progression, which might be leveraged to develop more efficacious therapies.


Subject(s)
Diabetes Mellitus, Type 2 , Diet, High-Fat , Glucose Intolerance , Glucose Transporter Type 4 , Hyperglycemia , Insulin Resistance , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/metabolism , Hyperglycemia/blood , Hyperglycemia/etiology , Hyperglycemia/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
J Biol Chem ; 297(1): 100881, 2021 07.
Article in English | MEDLINE | ID: mdl-34144038

ABSTRACT

GPR17 is a G-protein-coupled receptor (GPCR) implicated in the regulation of glucose metabolism and energy homeostasis. Such evidence is primarily drawn from mouse knockout studies and suggests GPR17 as a potential novel therapeutic target for the treatment of metabolic diseases. However, links between human GPR17 genetic variants, downstream cellular signaling, and metabolic diseases have yet to be reported. Here, we analyzed GPR17 coding sequences from control and disease cohorts consisting of individuals with adverse clinical metabolic deficits including severe insulin resistance, hypercholesterolemia, and obesity. We identified 18 nonsynonymous GPR17 variants, including eight variants that were exclusive to the disease cohort. We characterized the protein expression levels, membrane localization, and downstream signaling profiles of nine GPR17 variants (F43L, V96M, V103M, D105N, A131T, G136S, R248Q, R301H, and G354V). These nine GPR17 variants had similar protein expression and subcellular localization as wild-type GPR17; however, they showed diverse downstream signaling profiles. GPR17-G136S lost the capacity for agonist-mediated cAMP, Ca2+, and ß-arrestin signaling. GPR17-V96M retained cAMP inhibition similar to GPR17-WT, but showed impaired Ca2+ and ß-arrestin signaling. GPR17-D105N displayed impaired cAMP and Ca2+ signaling, but unaffected agonist-stimulated ß-arrestin recruitment. The identification and functional profiling of naturally occurring human GPR17 variants from individuals with metabolic diseases revealed receptor variants with diverse signaling profiles, including differential signaling perturbations that resulted in GPCR signaling bias. Our findings provide a framework for structure-function relationship studies of GPR17 signaling and metabolic disease.


Subject(s)
Metabolic Syndrome/genetics , Mutation, Missense , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Calcium/metabolism , Cyclic AMP/metabolism , HEK293 Cells , Humans , Protein Transport , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism
5.
Nutr Diabetes ; 9(1): 29, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31611548

ABSTRACT

BACKGROUND: Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARH) control energy homeostasis by sensing hormonal and nutrient cues and activating secondary melanocortin sensing neurons. We identified the expression of a G protein-coupled receptor, Gpr17, in the ARH and hypothesized that it contributes to the regulatory function of POMC neurons on metabolism. METHODS: In order to test this hypothesis, we generated POMC neuron-specific Gpr17 knockout (PGKO) mice and determined their energy and glucose metabolic phenotypes on normal chow diet (NCD) and high-fat diet (HFD). RESULTS: Adult PGKO mice on NCD displayed comparable body composition and metabolic features measured by indirect calorimetry. By contrast, PGKO mice on HFD demonstrated a sexually dimorphic phenotype with female PGKO mice displaying better metabolic homeostasis. Notably, female PGKO mice gained significantly less body weight and adiposity (p < 0.01), which was associated with increased energy expenditure, locomotor activity, and respiratory quotient, while males did not have an overt change in energy homeostasis. Though PGKO mice of both sexes had comparable glucose and insulin tolerance, detailed analyses of liver gene expression and serum metabolites indicate that PGKO mice could have reduced gluconeogenesis and increased lipid utilization on HFD. To elucidate the central-based mechanism(s) underlying the better-preserved energy and glucose homeostasis in PGKO mice on HFD, we examined the electrophysiological properties of POMC neurons and found Gpr17 deficiency led to increased spontaneous action potentials. Moreover, PGKO mice, especially female knockouts, had increased POMC-derived alpha-melanocyte stimulating hormone and beta-endorphin despite a comparable level of prohormone POMC in their hypothalamic extracts. CONCLUSIONS: Gpr17 deficiency in POMC neurons protects metabolic homeostasis in a sex-dependent manner during dietary and aging challenges, suggesting that Gpr17 could be an effective anti-obesity target in specific populations with poor metabolic control.


Subject(s)
Body Weight/physiology , Brain/metabolism , Insulin Resistance/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Receptors, G-Protein-Coupled/metabolism , Aging/metabolism , Animals , Diet, High-Fat , Energy Metabolism/physiology , Female , Homeostasis/physiology , Liver/metabolism , Male , Mice , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Sex Factors
6.
Sensors (Basel) ; 19(15)2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31344821

ABSTRACT

Purinergic signals, such as extracellular adenosine triphosphate (ATP) and adenosine diphosphate (ADP), mediate intercellular communication and stress responses throughout mammalian tissues, but the dynamics of their release and clearance are still not well understood. Although physiochemical methods provide important insight into physiology, genetically encoded optical sensors have proven particularly powerful in the quantification of signaling in live specimens. Indeed, genetically encoded luminescent and fluorescent sensors provide new insights into ATP-mediated purinergic signaling. However, new tools to detect extracellular ADP are still required. To this end, in this study, we use protein engineering to generate a new genetically encoded sensor that employs a high-affinity bacterial ADP-binding protein and reports a change in occupancy with a change in the Förster-type resonance energy transfer (FRET) between cyan and yellow fluorescent proteins. We characterize the sensor in both protein solution studies, as well as live-cell microscopy. This new sensor responds to nanomolar and micromolar concentrations of ADP and ATP in solution, respectively, and in principle it is the first fully-genetically encoded sensor with sufficiently high affinity for ADP to detect low levels of extracellular ADP. Furthermore, we demonstrate that tethering the sensor to the cell surface enables the detection of physiologically relevant nucleotide release induced by hypoosmotic shock as a model of tissue edema. Thus, we provide a new tool to study purinergic signaling that can be used across genetically tractable model systems.


Subject(s)
Adenosine Diphosphate/isolation & purification , Adenosine Triphosphate/isolation & purification , Biosensing Techniques , Edema/diagnosis , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Cell Communication/genetics , Edema/genetics , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Humans , Luminescent Proteins/chemistry , Osmotic Pressure , Protein Binding/genetics
7.
PLoS One ; 12(11): e0187481, 2017.
Article in English | MEDLINE | ID: mdl-29121644

ABSTRACT

Extracellular adenosine triphosphate (ATP) is a key purinergic signal that mediates cell-to-cell communication both within and between organ systems. We address the need for a robust and minimally invasive approach to measuring extracellular ATP by re-engineering the ATeam ATP sensor to be expressed on the cell surface. Using this approach, we image real-time changes in extracellular ATP levels with a sensor that is fully genetically-encoded and does not require an exogenous substrate. In addition, the sensor is ratiometric to allow for reliable quantitation of extracellular ATP fluxes. Using live-cell microscopy, we characterize sensor performance when expressed on cultured Neuro2A cells, and we measure both stimulated release of ATP and its clearance by ectonucleotidases. Thus, this proof-of-principle demonstrates a first-generation sensor to report extracellular ATP dynamics that may be useful for studying purinergic signaling in living specimens.


Subject(s)
5'-Nucleotidase/chemistry , Adenosine Triphosphate/analysis , Biosensing Techniques/methods , HEK293 Cells , Humans , Microscopy, Fluorescence/methods
8.
J Chem Inf Model ; 57(2): 322-334, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28068084

ABSTRACT

Adenylyl cyclase 2 (AC2) is one of nine membrane-bound isoforms of adenylyl cyclase that converts ATP into cyclic AMP (cAMP), an important second messenger molecule. Upregulation of AC2 is linked to cancers like pancreatic and small intestinal neuroendocrine tumors (NETs). The structures of the various isoforms of adenylyl cyclases are highly homologous, posing a significant challenge to drug discovery efforts for an effective, isoform-selective modulator of AC2. In a previous study, a screen identified a potential isoform-selective and noncompetitive inhibitor of AC2, SKF83566. In the present study, molecular modeling is used to explore the mode of inhibition of AC2 by SKF83566 and to investigate the active enantiomer of SKF83566. Homology models of hAC2 were built based on canine AC5-C1a and rat AC2-C2a templates. With these models, a combination of flexible docking, molecular dynamics simulations, and free energy calculations using the MM/GBSA methodology suggested an allosteric mechanism in which (S)-SKF83566 binds to an allosteric site near ATP and alters the protein conformation of the ATP binding site, potentially preventing the adenosine moiety of ATP from forming an archlike shape to form cAMP. The predicted binding preference for the (S)-SKF83566 enantiomer and the predicted free energy are consistent with the experimental data.


Subject(s)
Adenylyl Cyclases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Adenylyl Cyclases/chemistry , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Humans , Protein Conformation , Sequence Homology, Amino Acid , Stereoisomerism
9.
Eur J Pharmacol ; 763(Pt B): 223-32, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25981304

ABSTRACT

Heterologous sensitization of adenylyl cyclase (also referred to as superactivation, sensitization, or supersensitization of adenylyl cyclase) is a cellular adaptive response first described 40 years ago in the laboratory of Dr. Marshall Nirenberg. This apparently paradoxical cellular response occurs following persistent activation of Gαi/o-coupled receptors and causes marked enhancement in the activity of adenylyl cyclases, thereby increasing cAMP production. Since our last review in 2005, significant progress in the field has led to a better understanding of the relevance of, and the cellular biochemical processes that occur during the development and expression of heterologous sensitization. In this review we will discuss the recent advancements in the field and the mechanistic hypotheses on heterologous sensitization.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Animals , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Humans
10.
J Pharmacol Exp Ther ; 352(1): 53-60, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25332454

ABSTRACT

The yellow fever mosquito, Aedes aegypti, vectors disease-causing agents that adversely affect human health, most notably the viruses causing dengue and yellow fever. The efficacy of current mosquito control programs is challenged by the emergence of insecticide-resistant mosquito populations, suggesting an urgent need for the development of chemical insecticides with new mechanisms of action. One recently identified potential insecticide target is the A. aegypti D1-like dopamine receptor, AaDOP2. The focus of the present study was to evaluate AaDOP2 antagonism both in vitro and in vivo using assay technologies with increased throughput. The in vitro assays revealed AaDOP2 antagonism by four distinct chemical scaffolds from tricyclic antidepressant or antipsychotic chemical classes, and elucidated several structure-activity relationship trends that contributed to enhanced antagonist potency, including lipophilicity, halide substitution on the tricyclic core, and conformational rigidity. Six compounds displayed previously unparalleled potency for in vitro AaDOP2 antagonism, and among these, asenapine, methiothepin, and cis-(Z)-flupenthixol displayed subnanomolar IC50 values and caused rapid toxicity to A. aegypti larvae and/or adults in vivo. Our study revealed a significant correlation between in vitro potency for AaDOP2 antagonism and in vivo toxicity, suggesting viability of AaDOP2 as an insecticidal target. Taken together, this study expanded the repertoire of known AaDOP2 antagonists, enhanced our understanding of AaDOP2 pharmacology, provided further support for rational targeting of AaDOP2, and demonstrated the utility of efficiency-enhancing in vitro and in vivo assay technologies within our genome-to-lead pipeline for the discovery of next-generation insecticides.


Subject(s)
Aedes , Antidepressive Agents , Antipsychotic Agents , Dopamine Antagonists , Insect Proteins/antagonists & inhibitors , Mosquito Control/methods , Receptors, Dopamine/metabolism , Aedes/physiology , Animals , Drug Evaluation, Preclinical , Female , HEK293 Cells , High-Throughput Screening Assays , Humans , Larva , Small Molecule Libraries , Yellow Fever/transmission
11.
J Vis Exp ; (83): e51218, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24514897

ABSTRACT

Sensitization of adenylyl cyclase (AC) signaling has been implicated in a variety of neuropsychiatric and neurologic disorders including substance abuse and Parkinson's disease. Acute activation of Gαi/o-linked receptors inhibits AC activity, whereas persistent activation of these receptors results in heterologous sensitization of AC and increased levels of intracellular cAMP. Previous studies have demonstrated that this enhancement of AC responsiveness is observed both in vitro and in vivo following the chronic activation of several types of Gαi/o-linked receptors including D2 dopamine and µ opioid receptors. Although heterologous sensitization of AC was first reported four decades ago, the mechanism(s) that underlie this phenomenon remain largely unknown. The lack of mechanistic data presumably reflects the complexity involved with this adaptive response, suggesting that nonbiased approaches could aid in identifying the molecular pathways involved in heterologous sensitization of AC. Previous studies have implicated kinase and Gbγ signaling as overlapping components that regulate the heterologous sensitization of AC. To identify unique and additional overlapping targets associated with sensitization of AC, the development and validation of a scalable cAMP sensitization assay is required for greater throughput. Previous approaches to study sensitization are generally cumbersome involving continuous cell culture maintenance as well as a complex methodology for measuring cAMP accumulation that involves multiple wash steps. Thus, the development of a robust cell-based assay that can be used for high throughput screening (HTS) in a 384 well format would facilitate future studies. Using two D2 dopamine receptor cellular models (i.e. CHO-D2L and HEK-AC6/D2L), we have converted our 48-well sensitization assay (>20 steps 4-5 days) to a five-step, single day assay in 384-well format. This new format is amenable to small molecule screening, and we demonstrate that this assay design can also be readily used for reverse transfection of siRNA in anticipation of targeted siRNA library screening.


Subject(s)
Adenylyl Cyclases/metabolism , High-Throughput Screening Assays/methods , RNA, Small Interfering/analysis , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Animals , CHO Cells , Cricetulus , Cyclic AMP/metabolism , HEK293 Cells , Humans , Miniaturization/methods , RNA, Small Interfering/genetics , Rats , Receptors, Dopamine D2/metabolism , Transfection
12.
J Pharmacol Exp Ther ; 347(2): 276-87, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24008337

ABSTRACT

Adenylyl cyclase (AC) isoforms are implicated in several physiologic processes and disease states, but advancements in the therapeutic targeting of AC isoforms have been limited by the lack of potent and isoform-selective small-molecule modulators. The discovery of AC isoform-selective small molecules is expected to facilitate the validation of AC isoforms as therapeutic targets and augment the study of AC isoform function in vivo. Identification of chemical probes for AC2 is particularly important because there are no published genetic deletion studies and few small-molecule modulators. The present report describes the development and implementation of an intact-cell, small-molecule screening approach and subsequent validation paradigm for the discovery of AC2 inhibitors. The NIH clinical collections I and II were screened for inhibitors of AC2 activity using PMA-stimulated cAMP accumulation as a functional readout. Active compounds were subsequently confirmed and validated as direct AC2 inhibitors using orthogonal and counterscreening assays. The screening effort identified SKF-83566 [8-bromo-2,3,4,5-tetrahydro-3-methyl-5-phenyl-1H-3-benzazepin-7-ol hydrobromide] as a selective AC2 inhibitor with superior pharmacological properties for selective modulation of AC2 compared with currently available AC inhibitors. The utility of SKF-83566 as a small-molecule probe to study the function of endogenous ACs was demonstrated in C2C12 mouse skeletal muscle cells and human bronchial smooth muscle cells.


Subject(s)
2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/analogs & derivatives , Adenylyl Cyclase Inhibitors , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Small Molecule Libraries/pharmacology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/chemistry , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Adenylyl Cyclases/genetics , Animals , Cell Membrane/enzymology , Cell Membrane/immunology , Cyclic AMP/metabolism , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Mice , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/immunology , Sf9 Cells , Small Molecule Libraries/chemistry , Spodoptera , Transfection
13.
Cell Mol Neurobiol ; 33(4): 551-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23504261

ABSTRACT

Activator of G protein signaling 3 (AGS3) binds Gα(i) subunits in the GDP-bound state, implicating AGS3 as an important regulator of Gα(i)-linked receptor (e.g., D2 dopamine and µ-opioid) signaling. We examined the ability of AGS3 to modulate recombinant adenylyl cyclase (AC) type 1 and 2 signaling in HEK293 cells following both acute and persistent activation of the D(2L) dopamine receptor (D(2L)DR). AGS3 expression modestly enhanced the potency of acute quinpirole-induced D(2L)DR modulation of AC1 or AC2 activity. AGS3 also promoted desensitization of D(2L)DR-mediated inhibition of AC1, whereas desensitization of D(2L)DR-mediated AC2 activation was significantly attenuated. Additionally, AGS3 reduced D(2L)DR-mediated sensitization of AC1 and AC2. These data suggest that AGS3 is involved in altering G protein signaling in a complex fashion that is effector-specific and dependent on the duration of receptor activation.


Subject(s)
Adenylyl Cyclases/metabolism , Carrier Proteins/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction , Animals , HEK293 Cells , Humans , Illicit Drugs/pharmacology , Isoenzymes/metabolism , Rats , Signal Transduction/drug effects
14.
Methods Enzymol ; 521: 259-79, 2013.
Article in English | MEDLINE | ID: mdl-23351744

ABSTRACT

Emerging evidence indicates that G protein-coupled receptor (GPCR) signaling is mediated by receptor-receptor interactions at multiple levels. Thus, understanding the biochemistry and pharmacology of those receptor complexes is an important part of delineating the fundamental processes associated with GPCR-mediated signaling in human disease. A variety of experimental approaches have been used to explore these complexes, including bimolecular fluorescence complementation (BiFC) and multicolor BiFC (mBiFC). BiFC approaches have recently been used to explore the composition, cellular localization, and drug modulation of GPCR complexes. The basic methods for applying BiFC and mBiFC to study GPCRs in living cells are the subject of the present chapter.


Subject(s)
Protein Interaction Mapping/methods , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/metabolism , Animals , Fluorescence , Fluorometry/methods , Humans , Microscopy, Fluorescence/methods , Protein Multimerization , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection/methods
15.
Bioorg Med Chem ; 20(21): 6366-74, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018094

ABSTRACT

This work describes the identification of a novel class of octahydrobenzo[f]quinolines as dopamine D(3)-selective full agonists. We developed a facile method that utilizes Suzuki coupling for easy incorporations of various substituted pendant rings into the scaffold. A small focused library of octahydrobenzo[f]quinolines 5 was synthesized, and these compounds demonstrated at least 14-fold D(2)-like selectivity over D(1) in native porcine striatal tissue. Furthermore, n-propyl analog 5f was found to be a high affinity (K(i)=1.1 nM) D(3) dopamine full agonist with 145-fold selectivity over the D(2) receptor and about 840-fold selectivity over the D(1) receptor.


Subject(s)
Quinolines/pharmacology , Receptors, Dopamine D3/agonists , Binding, Competitive/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ligands , Models, Molecular , Molecular Conformation , Quinolines/chemical synthesis , Quinolines/chemistry , Receptors, Dopamine D3/metabolism , Structure-Activity Relationship
16.
J Biol Chem ; 283(52): 36698-710, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18984596

ABSTRACT

Heterotrimeric G-protein Galpha subunits and GoLoco motif proteins are key members of a conserved set of regulatory proteins that influence invertebrate asymmetric cell division and vertebrate neuroepithelium and epithelial progenitor differentiation. GoLoco motif proteins bind selectively to the inhibitory subclass (Galphai) of Galpha subunits, and thus it is assumed that a Galphai.GoLoco motif protein complex plays a direct functional role in microtubule dynamics underlying spindle orientation and metaphase chromosomal segregation during cell division. To address this hypothesis directly, we rationally identified a point mutation to Galphai subunits that renders a selective loss-of-function for GoLoco motif binding, namely an asparagine-to-isoleucine substitution in the alphaD-alphaE loop of the Galpha helical domain. This GoLoco-insensitivity ("GLi") mutation prevented Galphai1 association with all human GoLoco motif proteins and abrogated interaction between the Caenorhabditis elegans Galpha subunit GOA-1 and the GPR-1 GoLoco motif. In contrast, the GLi mutation did not perturb any other biochemical or signaling properties of Galphai subunits, including nucleotide binding, intrinsic and RGS protein-accelerated GTP hydrolysis, and interactions with Gbetagamma dimers, adenylyl cyclase, and seven transmembrane-domain receptors. GoLoco insensitivity rendered Galphai subunits unable to recruit GoLoco motif proteins such as GPSM2/LGN and GPSM3 to the plasma membrane, and abrogated the exaggerated mitotic spindle rocking normally seen upon ectopic expression of wild type Galphai subunits in kidney epithelial cells. This GLi mutation should prove valuable in establishing the physiological roles of Galphai.GoLoco motif protein complexes in microtubule dynamics and spindle function during cell division as well as to delineate potential roles for GoLoco motifs in receptor-mediated signal transduction.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Spindle Apparatus , Amino Acid Motifs , Amino Acid Sequence , Animals , Caenorhabditis elegans , Cell Membrane/metabolism , Humans , Microtubules/metabolism , Models, Molecular , Molecular Sequence Data , Point Mutation , Rats , Sequence Homology, Amino Acid , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...