90 results on '"Willard FS"'
Search Results
2. The incretin co-agonist tirzepatide requires GIPR for hormone secretion from human islets.
- Author
-
El K, Douros JD, Willard FS, Novikoff A, Sargsyan A, Perez-Tilve D, Wainscott DB, Yang B, Chen A, Wothe D, Coupland C, Tschöp MH, Finan B, D'Alessio DA, Sloop KW, Müller TD, and Campbell JE
- Subjects
- Humans, Animals, Mice, Glucagon-Like Peptide Receptors agonists, Insulin metabolism, Cells, Cultured, Gastric Inhibitory Polypeptide pharmacology, Islets of Langerhans drug effects, Incretins pharmacology, Hypoglycemic Agents pharmacology
- Abstract
The incretins glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) mediate insulin responses that are proportionate to nutrient intake to facilitate glucose tolerance
1 . The GLP-1 receptor (GLP-1R) is an established drug target for the treatment of diabetes and obesity2 , whereas the therapeutic potential of the GIP receptor (GIPR) is a subject of debate. Tirzepatide is an agonist at both the GIPR and GLP-1R and is a highly effective treatment for type 2 diabetes and obesity3,4 . However, although tirzepatide activates GIPR in cell lines and mouse models, it is not clear whether or how dual agonism contributes to its therapeutic benefit. Islet beta cells express both the GLP-1R and the GIPR, and insulin secretion is an established mechanism by which incretin agonists improve glycemic control5 . Here, we show that in mouse islets, tirzepatide stimulates insulin secretion predominantly through the GLP-1R, owing to reduced potency at the mouse GIPR. However, in human islets, antagonizing GIPR activity consistently decreases the insulin response to tirzepatide. Moreover, tirzepatide enhances glucagon secretion and somatostatin secretion in human islets. These data demonstrate that tirzepatide stimulates islet hormone secretion from human islets through both incretin receptors., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
3. Mutual Cooperativity of Three Allosteric Sites on the Dopamine D1 Receptor.
- Author
-
Wang X, Hembre EJ, Goldsmith PJ, Beck JP, Svensson KA, Willard FS, and Bruns RF
- Subjects
- Humans, Allosteric Site physiology, Allosteric Regulation physiology, Receptors, G-Protein-Coupled, Dopamine metabolism, Receptors, Dopamine D1 metabolism
- Abstract
An amine-containing molecule called Compound A has been reported by a group from Bristol-Myers Squibb to act as a positive allosteric modulator (PAM) at the dopamine D1 receptor. We synthesized the more active enantiomer of Compound A (BMS-A1) and compared it with the D1 PAMs DETQ and MLS6585, which are known to bind to intracellular loop 2 and the extracellular portion of transmembrane helix 7, respectively. Results from D1/D5 chimeras indicated that PAM activity of BMS-A1 tracked with the presence of D1 sequence in the N-terminal/extracellular region of the D1 receptor, a unique location compared with either of the other PAMs. In pairwise combinations, BMS-A1 potentiated the small allo-agonist activity of each of the other PAMs, while the triple PAM combination (in the absence of dopamine) produced a cAMP response about 64% of the maximum produced by dopamine. Each of the pairwise PAM combinations produced a much larger leftward shift of the dopamine EC
50 than either single PAM alone. All three PAMs in combination produced a 1000-fold leftward shift of the dopamine curve. These results demonstrate the presence of three non-overlapping allosteric sites that cooperatively stabilize the same activated state of the human D1 receptor. SIGNIFICANCE STATEMENT: Deficiencies in dopamine D1 receptor activation are seen in Parkinson disease and other neuropsychiatric disorders. In this study, three positive allosteric modulators of the dopamine D1 receptor were found to bind to distinct and separate sites, interacting synergistically with each other and dopamine, with the triple combination causing a 1000-fold leftward shift of the response to dopamine. These results showcase multiple opportunities to modulate D1 tone and highlight new pharmacological approaches for allosteric modulation of G-protein-coupled receptors., (Copyright © 2023 by The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
4. LY3437943, a novel triple glucagon, GIP, and GLP-1 receptor agonist for glycemic control and weight loss: From discovery to clinical proof of concept.
- Author
-
Coskun T, Urva S, Roell WC, Qu H, Loghin C, Moyers JS, O'Farrell LS, Briere DA, Sloop KW, Thomas MK, Pirro V, Wainscott DB, Willard FS, Abernathy M, Morford L, Du Y, Benson C, Gimeno RE, Haupt A, and Milicevic Z
- Subjects
- Animals, Body Weight, Gastric Inhibitory Polypeptide metabolism, Glucagon-Like Peptide-1 Receptor metabolism, Glycemic Control, Mice, Mice, Obese, Receptors, Glucagon metabolism, Weight Loss, Glucagon metabolism, Receptors, Gastrointestinal Hormone metabolism
- Abstract
With an increasing prevalence of obesity, there is a need for new therapies to improve body weight management and metabolic health. Multireceptor agonists in development may provide approaches to fulfill this unmet medical need. LY3437943 is a novel triple agonist peptide at the glucagon receptor (GCGR), glucose-dependent insulinotropic polypeptide receptor (GIPR), and glucagon-like peptide-1 receptor (GLP-1R). In vitro, LY3437943 shows balanced GCGR and GLP-1R activity but more GIPR activity. In obese mice, administration of LY3437943 decreased body weight and improved glycemic control. Body weight loss was augmented by the addition of GCGR-mediated increases in energy expenditure to GIPR- and GLP-1R-driven calorie intake reduction. In a phase 1 single ascending dose study, LY3437943 showed a safety and tolerability profile similar to other incretins. Its pharmacokinetic profile supported once-weekly dosing, and a reduction in body weight persisted up to day 43 after a single dose. These findings warrant further clinical assessment of LY3437943., Competing Interests: Declaration of interests All authors are employees and stockholders of Eli Lilly and Company and have no other conflicts of interest to declare. C.L. is a former employee of Eli Lilly and Company., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
5. Structural determinants of dual incretin receptor agonism by tirzepatide.
- Author
-
Sun B, Willard FS, Feng D, Alsina-Fernandez J, Chen Q, Vieth M, Ho JD, Showalter AD, Stutsman C, Ding L, Suter TM, Dunbar JD, Carpenter JW, Mohammed FA, Aihara E, Brown RA, Bueno AB, Emmerson PJ, Moyers JS, Kobilka TS, Coghlan MP, Kobilka BK, and Sloop KW
- Subjects
- Gastric Inhibitory Polypeptide metabolism, Gastric Inhibitory Polypeptide pharmacology, Gastric Inhibitory Polypeptide therapeutic use, Glucagon-Like Peptide-1 Receptor metabolism, Humans, Incretins pharmacology, Diabetes Mellitus, Type 2 drug therapy, Diabetes Mellitus, Type 2 metabolism, Receptors, Gastrointestinal Hormone agonists, Receptors, Gastrointestinal Hormone metabolism, Receptors, Gastrointestinal Hormone therapeutic use
- Abstract
SignificanceTirzepatide is a dual agonist of the glucose-dependent insulinotropic polypeptide receptor (GIPR) and the glucagon-like peptide-1 receptor (GLP-1R), which are incretin receptors that regulate carbohydrate metabolism. This investigational agent has proven superior to selective GLP-1R agonists in clinical trials in subjects with type 2 diabetes mellitus. Intriguingly, although tirzepatide closely resembles native GIP in how it activates the GIPR, it differs markedly from GLP-1 in its activation of the GLP-1R, resulting in less agonist-induced receptor desensitization. We report how cryogenic electron microscopy and molecular dynamics simulations inform the structural basis for the unique pharmacology of tirzepatide. These studies reveal the extent to which fatty acid modification, combined with amino acid sequence, determines the mode of action of a multireceptor agonist.
- Published
- 2022
- Full Text
- View/download PDF
6. Discovery of an Orally Efficacious Positive Allosteric Modulator of the Glucagon-like Peptide-1 Receptor.
- Author
-
Willard FS, Wainscott DB, Showalter AD, Stutsman C, Ma W, Cardona GR, Zink RW, Corkins CM, Chen Q, Yumibe N, Agejas J, Cumming GR, Minguez JM, Jiménez A, Mateo AI, Castaño AM, Briere DA, Sloop KW, and Bueno AB
- Subjects
- Administration, Oral, Animals, Blood Glucose analysis, Drug Discovery, Glucagon-Like Peptide-1 Receptor chemistry, Glucagon-Like Peptide-1 Receptor metabolism, Humans, Hypoglycemic Agents administration & dosage, Mice, Models, Molecular, Rats, Sprague-Dawley, Rats, Allosteric Regulation drug effects, Glucagon-Like Peptide-1 Receptor agonists, Hypoglycemic Agents chemistry, Hypoglycemic Agents pharmacology
- Abstract
The identification of LSN3318839, a positive allosteric modulator of the glucagon-like peptide-1 receptor (GLP-1R), is described. LSN3318839 increases the potency and efficacy of the weak metabolite GLP-1(9-36)NH
2 to become a full agonist at the GLP-1R and modestly potentiates the activity of the highly potent full-length ligand, GLP-1(7-36)NH2 . LSN3318839 preferentially enhances G protein-coupled signaling by the GLP-1R over β-arrestin recruitment. Ex vivo experiments show that the combination of GLP-1(9-36)NH2 and LSN3318839 produces glucose-dependent insulin secretion similar to that of GLP-1(7-36)NH2 . Under nutrient-stimulated conditions that release GLP-1, LSN3318839 demonstrates robust glucose lowering in animal models alone or in treatment combination with sitagliptin. From a therapeutic perspective, the biological properties of LSN3318839 support the concept that GLP-1R potentiation is sufficient for reducing hyperglycemia.- Published
- 2021
- Full Text
- View/download PDF
7. Nuisance compounds in cellular assays.
- Author
-
Dahlin JL, Auld DS, Rothenaigner I, Haney S, Sexton JZ, Nissink JWM, Walsh J, Lee JA, Strelow JM, Willard FS, Ferrins L, Baell JB, Walters MA, Hua BK, Hadian K, and Wagner BK
- Subjects
- Artificial Intelligence, Cheminformatics, Humans, Biological Products chemistry, Pharmaceutical Preparations chemistry
- Abstract
Compounds that exhibit assay interference or undesirable mechanisms of bioactivity ("nuisance compounds") are routinely encountered in cellular assays, including phenotypic and high-content screening assays. Much is known regarding compound-dependent assay interferences in cell-free assays. However, despite the essential role of cellular assays in chemical biology and drug discovery, there is considerably less known about nuisance compounds in more complex cell-based assays. In our view, a major obstacle to realizing the full potential of chemical biology will not just be difficult-to-drug targets or even the sheer number of targets, but rather nuisance compounds, due to their ability to waste significant resources and erode scientific trust. In this review, we summarize our collective academic, government, and industry experiences regarding cellular nuisance compounds. We describe assay design strategies to mitigate the impact of nuisance compounds and suggest best practices to efficiently address these compounds in complex biological settings., Competing Interests: Declaration of interests B.K.W. is an editor of Cell Chemical Biology., (Published by Elsevier Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
8. A High-Throughput Assay for the Pancreatic Islet Beta-Cell Potassium Channel: Use in the Pharmacological Characterization of Insulin Secretagogues Identified from Phenotypic Screening.
- Author
-
Song L, Barrett DG, Cox KL, Efanov AM, Syed SK, Tomandl D, and Willard FS
- Subjects
- Cells, Cultured, Drug Evaluation, Preclinical, High-Throughput Screening Assays, Humans, Insulin Secretion drug effects, Insulin-Secreting Cells metabolism, Optical Imaging, Phenotype, Diazoxide pharmacology, Insulin-Secreting Cells drug effects, Potassium Channels, Inwardly Rectifying metabolism, Secretagogues pharmacology, Sulfonylurea Compounds pharmacology, Sulfonylurea Receptors metabolism
- Abstract
Phenotypic screening is a neoclassical approach for drug discovery. We conducted phenotypic screening for insulin secretion enhancing agents using INS-1E insulinoma cells as a model system for pancreatic beta-cells. A principal regulator of insulin secretion in beta-cells is the metabolically regulated potassium channel Kir6.2/SUR1 complex. To characterize hit compounds, we developed an assay to quantify endogenous potassium channel activity in INS-1E cells. We quantified ligand-regulated potassium channel activity in INS-1E cells using fluorescence imaging and thallium flux. Potassium channel activity was metabolically regulated and coupled to insulin secretion. The pharmacology of channel opening agents (diazoxide) and closing agents (sulfonylureas) was used to validate the applicability of the assay. A precise high-throughput assay was enabled, and phenotypic screening hits were triaged to enable a higher likelihood of discovering chemical matter with novel and useful mechanisms of action.
- Published
- 2021
- Full Text
- View/download PDF
9. A synthetic method to assay adhesion-family G-protein coupled receptors. Determination of the G-protein coupling profile of ADGRG6(GPR126).
- Author
-
Lizano E, Hayes JL, and Willard FS
- Subjects
- Amino Acid Sequence, Cell Adhesion Molecules classification, Cell Adhesion Molecules metabolism, Cyclic AMP metabolism, HEK293 Cells, Humans, Ligands, Models, Biological, Models, Molecular, Protein Binding, Receptors, G-Protein-Coupled chemistry, Receptors, G-Protein-Coupled classification, Receptors, G-Protein-Coupled genetics, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Second Messenger Systems, Signal Transduction, Synthetic Biology, GTP-Binding Proteins metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
G-protein coupled receptors (GPCRs) are the largest family of membrane-spanning receptors in metazoans and mediate diverse biological processes such as chemotaxis, vision, and neurotransmission. Adhesion GPCRs represent an understudied class of GPCRs. Adhesion GPCRs (ADGRs) are activated by an intrinsic proteolytic mechanism executed by the G-protein autoproteolysis inducing domain that defines this class of GPCRs. It is hypothesized that agonist ligands modulate the proteolyzed receptor to regulate the activity of a tethered agonist peptide that is an intramolecular activator of ADGRs. The mechanism of activation of ADGRs in physiological settings is unclear and the toolbox for interrogating ADGR physiology in cellular models is limited. Therefore, we generated a novel enterokinase-activated tethered ligand system for ADGRG6(GPR126). Enterokinase addition to cells expressing a synthetic ADGRG6 protein induced potent and efficacious signal transduction through heterotrimeric G-protein coupled second messenger pathways including cyclic nucleotide production, intracellular calcium mobilization, and GPCR-pathway linked reporter gene induction. These studies support the hypothesis that ADGRG6(GPR126) is coupled to multiple heterotrimeric G-proteins: including Gα
s , Gαq , and Gα12 . This novel assay method is robust, specific, and compatible with numerous cell pharmacology approaches. We present a new tool for determination of the biological function of ADGRs and the identification of ligands that engage these receptors., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
10. Structural basis for GLP-1 receptor activation by LY3502970, an orally active nonpeptide agonist.
- Author
-
Kawai T, Sun B, Yoshino H, Feng D, Suzuki Y, Fukazawa M, Nagao S, Wainscott DB, Showalter AD, Droz BA, Kobilka TS, Coghlan MP, Willard FS, Kawabe Y, Kobilka BK, and Sloop KW
- Subjects
- Administration, Oral, Aminopyridines pharmacology, Animals, Anti-Obesity Agents pharmacology, Benzamides pharmacology, Cryoelectron Microscopy, Glucagon-Like Peptide-1 Receptor genetics, Glucagon-Like Peptide-1 Receptor metabolism, Glucagon-Like Peptide-1 Receptor ultrastructure, HEK293 Cells, Humans, Incretins pharmacology, Macaca fascicularis, Male, Mice, Mice, Transgenic, Models, Molecular, Mutagenesis, Site-Directed, Rats, Species Specificity, Swine, Tryptophan genetics, Glucagon-Like Peptide-1 Receptor agonists, Hypoglycemic Agents pharmacology, Protein Domains genetics
- Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists are efficacious antidiabetic medications that work by enhancing glucose-dependent insulin secretion and improving energy balance. Currently approved GLP-1R agonists are peptide based, and it has proven difficult to obtain small-molecule activators possessing optimal pharmaceutical properties. We report the discovery and mechanism of action of LY3502970 (OWL833), a nonpeptide GLP-1R agonist. LY3502970 is a partial agonist, biased toward G protein activation over β-arrestin recruitment at the GLP-1R. The molecule is highly potent and selective against other class B G protein-coupled receptors (GPCRs) with a pharmacokinetic profile favorable for oral administration. A high-resolution structure of LY3502970 in complex with active-state GLP-1R revealed a unique binding pocket in the upper helical bundle where the compound is bound by the extracellular domain (ECD), extracellular loop 2, and transmembrane helices 1, 2, 3, and 7. This mechanism creates a distinct receptor conformation that may explain the partial agonism and biased signaling of the compound. Further, interaction between LY3502970 and the primate-specific Trp33 of the ECD informs species selective activity for the molecule. In efficacy studies, oral administration of LY3502970 resulted in glucose lowering in humanized GLP-1R transgenic mice and insulinotropic and hypophagic effects in nonhuman primates, demonstrating an effect size in both models comparable to injectable exenatide. Together, this work determined the molecular basis for the activity of an oral agent being developed for the treatment of type 2 diabetes mellitus, offering insights into the activation of class B GPCRs by nonpeptide ligands., Competing Interests: Competing interest statement: D.B.W., A.D.S., B.A.D., M.P.C., F.S.W., and K.W.S. may own Eli Lilly and Company stock. T.S.K. is cofounder and president of ConfometRx. B.K.K. is a cofounder of and consultant for ConfometRx., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
11. Structural insights into probe-dependent positive allosterism of the GLP-1 receptor.
- Author
-
Bueno AB, Sun B, Willard FS, Feng D, Ho JD, Wainscott DB, Showalter AD, Vieth M, Chen Q, Stutsman C, Chau B, Ficorilli J, Agejas FJ, Cumming GR, Jiménez A, Rojo I, Kobilka TS, Kobilka BK, and Sloop KW
- Subjects
- Allosteric Site, Glucagon-Like Peptide 1 analogs & derivatives, Glucagon-Like Peptide-1 Receptor chemistry, Models, Molecular, Molecular Structure, Protein Conformation, Allosteric Regulation drug effects, Glucagon-Like Peptide-1 Receptor metabolism
- Abstract
Drugs that promote the association of protein complexes are an emerging therapeutic strategy. We report discovery of a G protein-coupled receptor (GPCR) ligand that stabilizes an active state conformation by cooperatively binding both the receptor and orthosteric ligand, thereby acting as a 'molecular glue'. LSN3160440 is a positive allosteric modulator of the GLP-1R optimized to increase the affinity and efficacy of GLP-1(9-36), a proteolytic product of GLP-1(7-36). The compound enhances insulin secretion in a glucose-, ligand- and GLP-1R-dependent manner. Cryo-electron microscopy determined the structure of the GLP-1R bound to LSN3160440 in complex with GLP-1 and heterotrimeric G
s . The modulator binds high in the helical bundle at an interface between TM1 and TM2, allowing access to the peptide ligand. Pharmacological characterization showed strong probe dependence of LSN3160440 for GLP-1(9-36) versus oxyntomodulin that is driven by a single residue. Our findings expand protein-protein modulation drug discovery to uncompetitive, active state stabilizers for peptide hormone receptors.- Published
- 2020
- Full Text
- View/download PDF
12. Synthetic protease-activated class B GPCRs.
- Author
-
Willard FS, Meredith TD, Showalter AD, Ma W, Ho JD, Sauder JM, and Sloop KW
- Subjects
- Animals, Cell Line, Exenatide genetics, Glucagon-Like Peptide 1 genetics, Glucagon-Like Peptide-1 Receptor genetics, HEK293 Cells, Humans, Insulin Secretion, Proteolysis, Rats, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Transfection, Exenatide metabolism, Glucagon-Like Peptide 1 metabolism, Glucagon-Like Peptide-1 Receptor metabolism, Peptide Hydrolases metabolism, Protein Engineering methods
- Abstract
G-protein coupled receptors (GPCRs) are the ligand detection machinery of a majority of extracellular signaling systems in metazoans. Novel chemical and biological tools to probe the structure-function relationships of GPCRs have impacted both basic and applied GPCR research. To better understand the structure-function of class B GPCRs, we generated receptor-ligand fusion chimeric proteins that can be activated by exogenous enzyme application. As a prototype, fusion proteins of the glucagon-like peptide-1 receptor (GLP-1R) with GLP-1(7-36) and exendin-4(1-39) peptides incorporating enterokinase-cleavable N-termini were generated. These receptors are predicted to generate fusion protein neo-epitopes upon proteolysis with enterokinase that are identical to the N-termini of GLP-1 agonists. This system was validated by measuring enterokinase-dependent GLP-1R mediated cAMP accumulation, and a structure-activity relationship for both linker length and peptide sequence was observed. Moreover, our results show this approach can be used in physiologically relevant cell systems, as GLP-1R-ligand chimeras were shown to induce glucose-dependent insulin secretion in insulinoma cells upon exposure to enterokinase. This approach suggests new strategies for understanding the structure-function of peptide-binding GPCRs., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:, (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
13. Selective Phosphodiesterase 1 Inhibitor BTTQ Reduces Blood Pressure in Spontaneously Hypertensive and Dahl Salt Sensitive Rats: Role of Peripheral Vasodilation.
- Author
-
Dey AB, Khedr S, Bean J, Porras LL, Meredith TD, Willard FS, Hass JV, Zhou X, Terashvili M, Jesudason CD, Ruley KM, Wiley MR, Kowala M, Atkinson SJ, Staruschenko A, and Rekhter MD
- Abstract
Regulation of the peripheral vascular resistance via modulating the vessel diameter has been considered as a main determinant of the arterial blood pressure. Phosphodiesterase enzymes (PDE1-11) hydrolyse cyclic nucleotides, which are key players controlling the vessel diameter and, thus, peripheral resistance. Here, we have tested and reported the effects of a novel selective PDE1 inhibitor (BTTQ) on the cardiovascular system. Normal Sprague Dawley, spontaneously hypertensive (SHR), and Dahl salt-sensitive rats were used to test in vivo the efficacy of the compound. Phosphodiesterase radiometric enzyme assay revealed that BTTQ inhibited all three isoforms of PDE1 in nanomolar concentration, while micromolar concentrations were needed to induce effective inhibition for other PDEs. The myography study conducted on mesenteric arteries revealed a potent vasodilatory effect of the drug, which was confirmed in vivo by an increase in the blood flow in the rat ear arteriols reflected by the rise in the temperature. Furthermore, BTTQ proved a high efficacy in lowering the blood pressure about 9, 36, and 24 mmHg in normal Sprague Dawley, SHR and, Dahl salt-sensitive rats, respectively, compared to the vehicle-treated group. Moreover, additional blood pressure lowering of about 22 mmHg could be achieved when BTTQ was administered on top of ACE inhibitor lisinopril, a current standard of care in the treatment of hypertension. Therefore, PDE1 inhibition induced efficient vasodilation that was accompanied by a significant reduction of blood pressure in different hypertensive rat models. Administration of BTTQ was also associated with increased heart rate in both models of hypertension as well as in the normotensive rats. Thus, PDE1 appears to be an attractive therapeutic target for the treatment of resistant hypertension, while tachycardia needs to be addressed by further compound structural optimization., (Copyright © 2020 Dey, Khedr, Bean, Porras, Meredith, Willard, Hass, Zhou, Terashvili, Jesudason, Ruley, Wiley, Kowala, Atkinson, Staruschenko and Rekhter.)
- Published
- 2020
- Full Text
- View/download PDF
14. Tirzepatide is an imbalanced and biased dual GIP and GLP-1 receptor agonist.
- Author
-
Willard FS, Douros JD, Gabe MB, Showalter AD, Wainscott DB, Suter TM, Capozzi ME, van der Velden WJ, Stutsman C, Cardona GR, Urva S, Emmerson PJ, Holst JJ, D'Alessio DA, Coghlan MP, Rosenkilde MM, Campbell JE, and Sloop KW
- Subjects
- Animals, Islets of Langerhans metabolism, Islets of Langerhans pathology, Male, Mice, Mice, Knockout, beta-Arrestin 1 physiology, Blood Glucose metabolism, Gastric Inhibitory Polypeptide pharmacology, Glucagon-Like Peptide-1 Receptor agonists, Hypoglycemic Agents pharmacology, Insulin metabolism, Islets of Langerhans drug effects, Receptors, Gastrointestinal Hormone agonists
- Abstract
Tirzepatide (LY3298176) is a dual GIP and GLP-1 receptor agonist under development for the treatment of type 2 diabetes mellitus (T2DM), obesity, and nonalcoholic steatohepatitis. Early phase trials in T2DM indicate that tirzepatide improves clinical outcomes beyond those achieved by a selective GLP-1 receptor agonist. Therefore, we hypothesized that the integrated potency and signaling properties of tirzepatide provide a unique pharmacological profile tailored for improving broad metabolic control. Here, we establish methodology for calculating occupancy of each receptor for clinically efficacious doses of the drug. This analysis reveals a greater degree of engagement of tirzepatide for the GIP receptor than the GLP-1 receptor, corroborating an imbalanced mechanism of action. Pharmacologically, signaling studies demonstrate that tirzepatide mimics the actions of native GIP at the GIP receptor but shows bias at the GLP-1 receptor to favor cAMP generation over β-arrestin recruitment, coincident with a weaker ability to drive GLP-1 receptor internalization compared with GLP-1. Experiments in primary islets reveal β-arrestin1 limits the insulin response to GLP-1, but not GIP or tirzepatide, suggesting that the biased agonism of tirzepatide enhances insulin secretion. Imbalance toward GIP receptor, combined with distinct signaling properties at the GLP-1 receptor, together may account for the promising efficacy of this investigational agent.
- Published
- 2020
- Full Text
- View/download PDF
15. Activation of the GLP-1 receptor by a non-peptidic agonist.
- Author
-
Zhao P, Liang YL, Belousoff MJ, Deganutti G, Fletcher MM, Willard FS, Bell MG, Christe ME, Sloop KW, Inoue A, Truong TT, Clydesdale L, Furness SGB, Christopoulos A, Wang MW, Miller LJ, Reynolds CA, Danev R, Sexton PM, and Wootten D
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Glucagon-Like Peptide-1 Receptor chemistry, Glucagon-Like Peptide-1 Receptor metabolism, Humans, Isoquinolines chemistry, Kinetics, Models, Molecular, Phenylalanine chemistry, Phenylalanine pharmacology, Protein Structure, Quaternary, Protein Structure, Tertiary, Pyridines chemistry, Structural Homology, Protein, Glucagon-Like Peptide-1 Receptor agonists, Isoquinolines pharmacology, Phenylalanine analogs & derivatives, Pyridines pharmacology
- Abstract
Class B G-protein-coupled receptors are major targets for the treatment of chronic diseases, including diabetes and obesity
1 . Structures of active receptors reveal peptide agonists engage deep within the receptor core, leading to an outward movement of extracellular loop 3 and the tops of transmembrane helices 6 and 7, an inward movement of transmembrane helix 1, reorganization of extracellular loop 2 and outward movement of the intracellular side of transmembrane helix 6, resulting in G-protein interaction and activation2-6 . Here we solved the structure of a non-peptide agonist, TT-OAD2, bound to the glucagon-like peptide-1 (GLP-1) receptor. Our structure identified an unpredicted non-peptide agonist-binding pocket in which reorganization of extracellular loop 3 and transmembrane helices 6 and 7 manifests independently of direct ligand interaction within the deep transmembrane domain pocket. TT-OAD2 exhibits biased agonism, and kinetics of G-protein activation and signalling that are distinct from peptide agonists. Within the structure, TT-OAD2 protrudes beyond the receptor core to interact with the lipid or detergent, providing an explanation for the distinct activation kinetics that may contribute to the clinical efficacy of this compound series. This work alters our understanding of the events that drive the activation of class B receptors.- Published
- 2020
- Full Text
- View/download PDF
16. Discovery and pharmacology of the covalent GLP-1 receptor (GLP-1R) allosteric modulator BETP: A novel tool to probe GLP-1R pharmacology.
- Author
-
Willard FS, Ho JD, and Sloop KW
- Subjects
- Allosteric Regulation drug effects, Amino Acid Sequence, Animals, Cyclic AMP metabolism, Glucagon-Like Peptide-1 Receptor chemistry, Humans, Pyrimidines chemistry, Small Molecule Libraries pharmacology, Drug Discovery, Glucagon-Like Peptide-1 Receptor agonists, Pyrimidines pharmacology
- Abstract
The glucagon-like peptide-1 receptor (GLP-1R) is a significant therapeutic target for small molecule drug discovery given the therapeutic impact of peptide agonists in the diabetes sphere. We review the discovery and subsequent characterization of the small molecule GLP-1R allosteric modulator 4-(3-(Benzyloxy)phenyl)-2-(ethylsulfinyl)-6-(trifluoromethyl)pyrimidine (BETP). BETP is a covalent modulator of the GLP-1R, and we discuss the pharmacological implications and possible structural basis of this novel mode of action. We highlight the insights into class B G-protein coupled receptor pharmacology and biology provided by studies conducted with BETP. These include the descriptions of exquisite allosteric modulator probe dependence and biased signaling in vitro and in vivo. We conclude with an analysis of the utility of BETP as a chemical probe for the GLP-1R., Competing Interests: Conflict of interest All authors are employees of Eli Lilly and Company and may own company stock or possess stock options., (© 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
17. The current state of GPCR-based drug discovery to treat metabolic disease.
- Author
-
Sloop KW, Emmerson PJ, Statnick MA, and Willard FS
- Subjects
- Animals, Humans, Metabolic Diseases metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction drug effects, Drug Discovery, Metabolic Diseases drug therapy, Receptors, G-Protein-Coupled antagonists & inhibitors
- Abstract
One approach of modern drug discovery is to identify agents that enhance or diminish signal transduction cascades in various cell types and tissues by modulating the activity of GPCRs. This strategy has resulted in the development of new medicines to treat many conditions, including cardiovascular disease, psychiatric disorders, HIV/AIDS, certain forms of cancer and Type 2 diabetes mellitus (T2DM). These successes justify further pursuit of GPCRs as disease targets and provide key learning that should help guide identifying future therapeutic agents. This report reviews the current landscape of GPCR drug discovery with emphasis on efforts aimed at developing new molecules for treating T2DM and obesity. We analyse historical efforts to generate GPCR-based drugs to treat metabolic disease in terms of causal factors leading to success and failure in this endeavour., Linked Articles: This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc., (© 2018 The British Pharmacological Society.)
- Published
- 2018
- Full Text
- View/download PDF
18. Beyond Glucagon-like Peptide-1: Is G-Protein Coupled Receptor Polypharmacology the Path Forward to Treating Metabolic Diseases?
- Author
-
Sloop KW, Briere DA, Emmerson PJ, and Willard FS
- Abstract
The glucagon-like peptide-1 receptor (GLP-1R) is a class B G-protein coupled receptor (GPCR) that has proven to be an effective target for developing medicines that treat type 2 diabetes mellitus (T2DM). GLP-1R agonists improve T2DM by enhancing glucose-stimulated insulin secretion, delaying gastric transit, decreasing glucagon levels, and reducing body weight due to anorexigenic actions. The therapeutic successes of these agents helped inspire the design of new multifunctional molecules that are GLP-1R agonists but also activate receptors linked to pathways that enhance insulin sensitization and/or energy expenditure. Herein, these agents are discussed in the context of polypharmacological approaches that may enable even further improvement in treatment outcomes. Moreover, we revisit classical polypharmaceutical GPCR approaches and how they may be utilized for treatment of T2DM. To determine optimal combination regimens, changes in drug discovery practices are likely needed because compensatory mechanisms appear to underlie progression of T2DM and limit the ability of current therapies to induce disease regression or remission., Competing Interests: The authors declare the following competing financial interest(s): All authors are employees of Eli Lilly and Company and may own company stock or possess stock options., (Copyright © 2018 American Chemical Society.)
- Published
- 2018
- Full Text
- View/download PDF
19. Regulation of Endogenous (Male) Rodent GLP-1 Secretion and Human Islet Insulin Secretion by Antagonism of Somatostatin Receptor 5.
- Author
-
Farb TB, Adeva M, Beauchamp TJ, Cabrera O, Coates DA, Meredith TD, Droz BA, Efanov A, Ficorilli JV, Gackenheimer SL, Martinez-Grau MA, Molero V, Ruano G, Statnick MA, Suter TM, Syed SK, Toledo MA, Willard FS, Zhou X, Bokvist KB, and Barrett DG
- Subjects
- Animals, CHO Cells, Cells, Cultured, Cricetinae, Cricetulus, HEK293 Cells, Humans, Insulin Secretion, Islets of Langerhans metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Rats, Rats, Sprague-Dawley, Rats, Zucker, Receptors, Somatostatin genetics, Secretory Pathway drug effects, Benzoates pharmacology, Glucagon-Like Peptide 1 metabolism, Hypoglycemic Agents pharmacology, Insulin metabolism, Islets of Langerhans drug effects, Receptors, Somatostatin antagonists & inhibitors, Spiro Compounds pharmacology
- Abstract
Incretin and insulin responses to nutrient loads are suppressed in persons with diabetes, resulting in decreased glycemic control. Agents including sulfonylureas and dipeptidyl peptidase-4 inhibitors (DPP4i) partially reverse these effects and provide therapeutic benefit; however, their modes of action limit efficacy. Because somatostatin (SST) has been shown to suppress insulin and glucagonlike peptide-1 (GLP-1) secretion through the Gi-coupled SST receptor 5 (SSTR5) isoform in vitro, antagonism of SSTR5 may improve glycemic control via intervention in both pathways. Here, we show that a potent and selective SSTR5 antagonist reverses the blunting effects of SST on insulin secretion from isolated human islets, and demonstrate that SSTR5 antagonism affords increased levels of systemic GLP-1 in vivo. Knocking out Sstr5 in mice provided a similar increase in systemic GLP-1 levels, which were not increased further by treatment with the antagonist. Treatment of mice with the SSTR5 antagonist in combination with a DPP4i resulted in increases in systemic GLP-1 levels that were more than additive and resulted in greater glycemic control compared with either agent alone. In isolated human islets, the SSTR5 antagonist completely reversed the inhibitory effect of exogenous SST-14 on insulin secretion. Taken together, these data suggest that SSTR5 antagonism should increase circulating GLP-1 levels and stimulate insulin secretion (directly and via GLP-1) in humans, improving glycemic control in patients with diabetes., (Copyright © 2017 Endocrine Society.)
- Published
- 2017
- Full Text
- View/download PDF
20. Positive Allosteric Modulation of the Glucagon-like Peptide-1 Receptor by Diverse Electrophiles.
- Author
-
Bueno AB, Showalter AD, Wainscott DB, Stutsman C, Marín A, Ficorilli J, Cabrera O, Willard FS, and Sloop KW
- Subjects
- Allosteric Regulation, Animals, Cell Line, Cyclic AMP metabolism, Glucagon-Like Peptide-1 Receptor agonists, Glucagon-Like Peptide-1 Receptor chemistry, Glucose metabolism, HEK293 Cells, Humans, Insulin metabolism, Insulin Secretion, Insulin-Secreting Cells drug effects, Insulin-Secreting Cells metabolism, Islets of Langerhans drug effects, Islets of Langerhans metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutant Proteins chemistry, Mutant Proteins metabolism, Pyrimidines chemistry, Pyrimidines pharmacology, Rats, Signal Transduction drug effects, Glucagon-Like Peptide-1 Receptor metabolism
- Abstract
Therapeutic intervention to activate the glucagon-like peptide-1 receptor (GLP-1R) enhances glucose-dependent insulin secretion and improves energy balance in patients with type 2 diabetes mellitus. Studies investigating mechanisms whereby peptide ligands activate GLP-1R have utilized mutagenesis, receptor chimeras, photo-affinity labeling, hydrogen-deuterium exchange, and crystallography of the ligand-binding ectodomain to establish receptor homology models. However, this has not enabled the design or discovery of drug-like non-peptide GLP-1R activators. Recently, studies investigating 4-(3-benzyloxyphenyl)-2-ethylsulfinyl-6-(trifluoromethyl)pyrimidine (BETP), a GLP-1R-positive allosteric modulator, determined that Cys-347 in the GLP-1R is required for positive allosteric modulator activity via covalent modification. To advance small molecule activation of the GLP-1R, we characterized the insulinotropic mechanism of BETP. In guanosine 5'-3-O-(thio)triphosphate binding and INS1 832-3 insulinoma cell cAMP assays, BETP enhanced GLP-1(9-36)-NH2-stimulated cAMP signaling. Using isolated pancreatic islets, BETP potentiated insulin secretion in a glucose-dependent manner that requires both the peptide ligand and GLP-1R. In studies of the covalent mechanism, PAGE fluorography showed labeling of GLP-1R in immunoprecipitation experiments from GLP-1R-expressing cells incubated with [(3)H]BETP. Furthermore, we investigated whether other reported GLP-1R activators and compounds identified from screening campaigns modulate GLP-1R by covalent modification. Similar to BETP, several molecules were found to enhance GLP-1R signaling in a Cys-347-dependent manner. These chemotypes are electrophiles that react with GSH, and LC/MS determined the cysteine adducts formed upon conjugation. Together, our results suggest covalent modification may be used to stabilize the GLP-1R in an active conformation. Moreover, the findings provide pharmacological guidance for the discovery and characterization of small molecule GLP-1R ligands as possible therapeutics., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
21. Novel Phenotypic Outcomes Identified for a Public Collection of Approved Drugs from a Publicly Accessible Panel of Assays.
- Author
-
Lee JA, Shinn P, Jaken S, Oliver S, Willard FS, Heidler S, Peery RB, Oler J, Chu S, Southall N, Dexheimer TS, Smallwood J, Huang R, Guha R, Jadhav A, Cox K, Austin CP, Simeonov A, Sittampalam GS, Husain S, Franklin N, Wild DJ, Yang JJ, Sutherland JJ, and Thomas CJ
- Subjects
- Cell Line, Tumor, Drug Approval, Drug Evaluation, Preclinical, High-Throughput Screening Assays, Humans, Inhibitory Concentration 50, Phenotype, Small Molecule Libraries pharmacology, Drug Repositioning
- Abstract
Phenotypic assays have a proven track record for generating leads that become first-in-class therapies. Whole cell assays that inform on a phenotype or mechanism also possess great potential in drug repositioning studies by illuminating new activities for the existing pharmacopeia. The National Center for Advancing Translational Sciences (NCATS) pharmaceutical collection (NPC) is the largest reported collection of approved small molecule therapeutics that is available for screening in a high-throughput setting. Via a wide-ranging collaborative effort, this library was analyzed in the Open Innovation Drug Discovery (OIDD) phenotypic assay modules publicly offered by Lilly. The results of these tests are publically available online at www.ncats.nih.gov/expertise/preclinical/pd2 and via the PubChem Database (https://pubchem.ncbi.nlm.nih.gov/) (AID 1117321). Phenotypic outcomes for numerous drugs were confirmed, including sulfonylureas as insulin secretagogues and the anti-angiogenesis actions of multikinase inhibitors sorafenib, axitinib and pazopanib. Several novel outcomes were also noted including the Wnt potentiating activities of rotenone and the antifolate class of drugs, and the anti-angiogenic activity of cetaben.
- Published
- 2015
- Full Text
- View/download PDF
22. Progesterone receptor membrane component 1 is a functional part of the glucagon-like peptide-1 (GLP-1) receptor complex in pancreatic β cells.
- Author
-
Zhang M, Robitaille M, Showalter AD, Huang X, Liu Y, Bhattacharjee A, Willard FS, Han J, Froese S, Wei L, Gaisano HY, Angers S, Sloop KW, Dai FF, and Wheeler MB
- Subjects
- Adenylyl Cyclase Inhibitors, Animals, CHO Cells, Cell Line, Cricetinae, Cricetulus, Cyclic AMP biosynthesis, Cyclic AMP metabolism, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Glucagon-Like Peptide-1 Receptor, Guanine Nucleotide Exchange Factors antagonists & inhibitors, Guanine Nucleotide Exchange Factors metabolism, Humans, Insulin Secretion, Mass Spectrometry, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Mice, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Rats, Receptors, Progesterone antagonists & inhibitors, Receptors, Progesterone genetics, rab5 GTP-Binding Proteins metabolism, Glucagon-Like Peptide 1 metabolism, Insulin metabolism, Insulin-Secreting Cells metabolism, Membrane Proteins metabolism, Receptors, Glucagon metabolism, Receptors, Progesterone metabolism
- Abstract
Glucagon-like peptide-1 (GLP-1) is an incretin hormone that regulates glucose homeostasis. Because of their direct stimulation of insulin secretion from pancreatic β cells, GLP-1 receptor (GLP-1R) agonists are now important therapeutic options for the treatment of type 2 diabetes. To better understand the mechanisms that control the insulinotropic actions of GLP-1, affinity purification and mass spectrometry (AP-MS) were employed to uncover potential proteins that functionally interact with the GLP-1R. AP-MS performed on Chinese hamster ovary cells or MIN6 β cells, both expressing the human GLP-1R, revealed 99 proteins potentially associated with the GLP-1R. Three novel GLP-1R interactors (PGRMC1, Rab5b, and Rab5c) were further validated through co-immunoprecipitation/immunoblotting, fluorescence resonance energy transfer, and immunofluorescence. Functional studies revealed that overexpression of PGRMC1, a novel cell surface receptor that associated with liganded GLP-1R, enhanced GLP-1-induced insulin secretion (GIIS) with the most robust effect. Knockdown of PGRMC1 in β cells decreased GIIS, indicative of positive interaction with GLP-1R. To gain insight mechanistically, we demonstrated that the cell surface PGRMC1 ligand P4-BSA increased GIIS, whereas its antagonist AG-205 decreased GIIS. It was then found that PGRMC1 increased GLP-1-induced cAMP accumulation. PGRMC1 activation and GIIS induced by P4-BSA could be blocked by inhibition of adenylyl cyclase/EPAC signaling or the EGF receptor-PI3K signal transduction pathway. These data reveal a dual mechanism for PGRMC1-increased GIIS mediated through cAMP and EGF receptor signaling. In conclusion, we identified several novel GLP-1R interacting proteins. PGRMC1 expressed on the cell surface of β cells was shown to interact with the activated GLP-1R to enhance the insulinotropic actions of GLP-1., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
23. Glucagon-like peptide-1 receptor ligand interactions: structural cross talk between ligands and the extracellular domain.
- Author
-
West GM, Willard FS, Sloop KW, Showalter AD, Pascal BD, and Griffin PR
- Subjects
- Amino Acid Sequence, Crystallography, X-Ray, Deuterium Exchange Measurement, Exenatide, Glucagon-Like Peptide-1 Receptor, Ligands, Molecular Sequence Data, Peptides chemistry, Peptides metabolism, Protein Structure, Tertiary, Small Molecule Libraries chemistry, Small Molecule Libraries metabolism, Venoms chemistry, Venoms metabolism, Receptors, Glucagon chemistry, Receptors, Glucagon metabolism
- Abstract
Activation of the glucagon-like peptide-1 receptor (GLP-1R) in pancreatic β-cells potentiates insulin production and is a current therapeutic target for the treatment of type 2 diabetes mellitus (T2DM). Like other class B G protein-coupled receptors (GPCRs), the GLP-1R contains an N-terminal extracellular ligand binding domain. N-terminal truncations on the peptide agonist generate antagonists capable of binding to the extracellular domain, but not capable of activating full length receptor. The main objective of this study was to use Hydrogen/deuterium exchange (HDX) to identify how the amide hydrogen bonding network of peptide ligands and the extracellular domain of GLP-1R (nGLP-1R) were altered by binding interactions and to then use this platform to validate direct binding events for putative GLP-1R small molecule ligands. The HDX studies presented here for two glucagon-like peptide-1 receptor (GLP-1R) peptide ligands indicates that the antagonist exendin-4[9-39] is significantly destabilized in the presence of nonionic detergents as compared to the agonist exendin-4. Furthermore, HDX can detect stabilization of exendin-4 and exendin-4[9-39] hydrogen bonding networks at the N-terminal helix [Val19 to Lys27] upon binding to the N-terminal extracellular domain of GLP-1R (nGLP-1R). In addition we show hydrogen bonding network stabilization on nGLP-1R in response to ligand binding, and validate direct binding events with the extracellular domain of the receptor for putative GLP-1R small molecule ligands.
- Published
- 2014
- Full Text
- View/download PDF
24. RGS21, a regulator of taste and mucociliary clearance?
- Author
-
Kimple AJ, Garland AL, Cohen SP, Setola V, Willard FS, Zielinski T, Lowery RG, Tarran R, and Siderovski DP
- Subjects
- Animals, Mice, Mice, Transgenic, Models, Animal, Pilot Projects, Random Allocation, Real-Time Polymerase Chain Reaction methods, Sensitivity and Specificity, Signal Transduction genetics, Gene Expression Regulation, Mucociliary Clearance genetics, RGS Proteins genetics, Taste genetics
- Abstract
Objectives/hypothesis: Motile cilia of airway epithelial cells help to expel harmful inhaled material. Activation of bitterant-responsive G protein-coupled receptors (GPCRs) is believed to potentiate cilia beat frequency and mucociliary clearance. In this study, we investigated whether regulator of G protein signaling-21 (RGS21) has the potential to modulate signaling pathways connected to airway mucociliary clearance, given that RGS proteins modulate GPCR signaling by acting as GTPase-accelerating proteins (GAPs) for the Gα subunits of heterotrimeric G proteins., Study Design: This is a pilot investigation to determine if RGS21, a potential tastant specific RGS gene, is expressed in sinonasal mucosa, and to determine its specific Gα substrate using in vitro biochemical assays with purified proteins., Methods: Rgs21 expression in sinonasal mucosa was determined using quantitative, real-time PCR and a transgenic mouse expressing RFP from the Rgs21 promoter. Rgs21 was cloned, over-expressed, and purified using multistep protein chromatography. Biochemical and biophysical assays were used to determine if RGS21 could bind and accelerate the hydrolysis of GTP on heterotrimeric Gα subunits., Results: Rgs21 was expressed in sinonasal mucosa and lingual epithelium. Purified recombinant protein directly bound and accelerated GTP hydrolysis on Gα subunits., Conclusions: Rgs21 is expressed in sinonasal mucosa, is amenable to purification as a recombinant protein, and can bind to Gα(i/o/q) subunits. Furthermore, RGS21 can accelerate the hydrolysis rate of GTP on Gαi subunits. This provides evidence that RGS21 may be a negative regulator of bitterant responses. Future studies will be needed to determine the physiological role of this protein in mucociliary clearance., (© 2013 The American Laryngological, Rhinological and Otological Society, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
25. The insulin secretory action of novel polycyclic guanidines: discovery through open innovation phenotypic screening, and exploration of structure-activity relationships.
- Author
-
Shaghafi MB, Barrett DG, Willard FS, and Overman LE
- Subjects
- Animals, Dose-Response Relationship, Drug, Glucose pharmacology, Guanidines chemical synthesis, Guanidines chemistry, Islets of Langerhans drug effects, Islets of Langerhans metabolism, Molecular Structure, Phenotype, Polycyclic Compounds chemical synthesis, Polycyclic Compounds chemistry, Rats, Structure-Activity Relationship, Drug Discovery, Guanidines pharmacology, Insulin metabolism, Polycyclic Compounds pharmacology
- Abstract
We report the discovery of the glucose-dependent insulin secretogogue activity of a novel class of polycyclic guanidines through phenotypic screening as part of the Lilly Open Innovation Drug Discovery platform. Three compounds from the University of California, Irvine, 1-3, having the 3-arylhexahydropyrrolo[1,2-c]pyrimidin-1-amine scaffold acted as insulin secretagogues under high, but not low, glucose conditions. Exploration of the structure-activity relationship around the scaffold demonstrated the key role of the guanidine moiety, as well as the importance of two lipophilic regions, and led to the identification of 9h, which stimulated insulin secretion in isolated rat pancreatic islets in a glucose-dependent manner., (Copyright © 2014 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
26. Differential activation and modulation of the glucagon-like peptide-1 receptor by small molecule ligands.
- Author
-
Wootten D, Savage EE, Willard FS, Bueno AB, Sloop KW, Christopoulos A, and Sexton PM
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Cyclobutanes pharmacology, Glucagon-Like Peptide-1 Receptor, Humans, Ligands, Signal Transduction drug effects, Signal Transduction physiology, Cyclobutanes metabolism, Receptors, Glucagon agonists, Receptors, Glucagon metabolism
- Abstract
The glucagon-like peptide-1 receptor (GLP-1R) is a major therapeutic target for the treatment of type 2 diabetes due to its role in glucose homeostasis. Despite the availability of peptide-based GLP-1R drugs for treatment of this disease, there is great interest in developing small molecules that can be administered orally. The GLP-1R system is complex, with multiple endogenous and clinically used peptide ligands that exhibit different signaling biases at this receptor. This study revealed that small molecule ligands acting at this receptor are differentially biased to peptide ligands and also from each other with respect to the signaling pathways that they activate. Furthermore, allosteric small molecule ligands were also able to induce bias in signaling mediated by orthosteric ligands. This was dependent on both the orthosteric and allosteric ligand as no two allosteric-orthosteric ligand pairs could induce the same signaling profile. We highlight the need to profile compounds across multiple signaling pathways and in combination with multiple orthosteric ligands in systems such as the GLP-1R where more than one endogenous ligand exists. In the context of pleiotropical coupling of receptors and the interplay of multiple pathways leading to physiologic responses, profiling of small molecules in this manner may lead to a better understanding of the physiologic consequences of biased signaling at this receptor. This could enable the design and development of improved therapeutics that have the ability to fine-tune receptor signaling, leading to beneficial therapeutic outcomes while reducing side effect profiles.
- Published
- 2013
- Full Text
- View/download PDF
27. Structural determinants of RGS-RhoGEF signaling critical to Entamoeba histolytica pathogenesis.
- Author
-
Bosch DE, Kimple AJ, Manning AJ, Muller RE, Willard FS, Machius M, Rogers SL, and Siderovski DP
- Subjects
- Amino Acid Substitution, Animals, Binding Sites, Cell Adhesion, Cell Line, Cell Shape, Cell Survival, Chemotaxis, Crystallography, X-Ray, Drosophila melanogaster, Entamoeba histolytica metabolism, GTP-Binding Protein alpha Subunits chemistry, Guanine Nucleotide Exchange Factors genetics, Guanine Nucleotide Exchange Factors metabolism, Guanosine Triphosphate chemistry, Host-Parasite Interactions, Hydrolysis, Models, Molecular, Mutagenesis, Site-Directed, Protein Binding, Protein Interaction Domains and Motifs, Protein Structure, Secondary, Protozoan Proteins genetics, Protozoan Proteins metabolism, Rho Guanine Nucleotide Exchange Factors, Trophozoites metabolism, Entamoeba histolytica physiology, Guanine Nucleotide Exchange Factors chemistry, Protozoan Proteins chemistry, Signal Transduction, Trophozoites physiology
- Abstract
G protein signaling pathways, as key components of physiologic responsiveness and timing, are frequent targets for pharmacologic intervention. Here, we identify an effector for heterotrimeric G protein α subunit (EhGα1) signaling from Entamoeba histolytica, the causative agent of amoebic colitis. EhGα1 interacts with this effector and guanosine triphosphatase-accelerating protein, EhRGS-RhoGEF, in a nucleotide state-selective fashion. Coexpression of EhRGS-RhoGEF with constitutively active EhGα1 and EhRacC leads to Rac-dependent spreading in Drosophila S2 cells. EhRGS-RhoGEF overexpression in E. histolytica trophozoites leads to reduced migration toward serum and lower cysteine protease activity, as well as reduced attachment to, and killing of, host cells. A 2.3 Å crystal structure of the full-length EhRGS-RhoGEF reveals a putative inhibitory helix engaging the Dbl homology domain Rho-binding surface and the pleckstrin homology domain. Mutational analysis of the EhGα1/EhRGS-RhoGEF interface confirms a canonical "regulator of G protein signaling" domain rather than a RhoGEF-RGS ("rgRGS") domain, suggesting a convergent evolution toward heterotrimeric and small G protein cross-talk., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
28. Regulator of G-protein signaling-21 (RGS21) is an inhibitor of bitter gustatory signaling found in lingual and airway epithelia.
- Author
-
Cohen SP, Buckley BK, Kosloff M, Garland AL, Bosch DE, Cheng G Jr, Radhakrishna H, Brown MD, Willard FS, Arshavsky VY, Tarran R, Siderovski DP, and Kimple AJ
- Subjects
- Animals, COS Cells, Calcium metabolism, Chlorocebus aethiops, Cyclic AMP genetics, GTP-Binding Protein Regulators genetics, Humans, Mice, Mice, Transgenic, RGS Proteins, Respiratory Mucosa cytology, Reverse Transcriptase Polymerase Chain Reaction, Taste Buds cytology, Calcium Signaling physiology, Cyclic AMP metabolism, GTP-Binding Protein Regulators biosynthesis, Respiratory Mucosa metabolism, Taste physiology, Taste Buds metabolism
- Abstract
The gustatory system detects tastants and transmits signals to the brain regarding ingested substances and nutrients. Although tastant receptors and taste signaling pathways have been identified, little is known about their regulation. Because bitter, sweet, and umami taste receptors are G protein-coupled receptors (GPCRs), we hypothesized that regulators of G protein signaling (RGS) proteins may be involved. The recent cloning of RGS21 from taste bud cells has implicated this protein in the regulation of taste signaling; however, the exact role of RGS21 has not been precisely defined. Here, we sought to determine the role of RGS21 in tastant responsiveness. Biochemical analyses confirmed in silico predictions that RGS21 acts as a GTPase-accelerating protein (GAP) for multiple G protein α subunits, including adenylyl cyclase-inhibitory (Gα(i)) subunits and those thought to be involved in tastant signal transduction. Using a combination of in situ hybridization, RT-PCR, immunohistochemistry, and immunofluorescence, we demonstrate that RGS21 is not only endogenously expressed in mouse taste buds but also in lung airway epithelial cells, which have previously been shown to express components of the taste signaling cascade. Furthermore, as shown by reverse transcription-PCR, the immortalized human airway cell line 16HBE was found to express transcripts for tastant receptors, RGS21, and downstream taste signaling components. Over- and underexpression of RGS21 in 16HBE cells confirmed that RGS21 acts to oppose bitter tastant signaling to cAMP and calcium second messenger changes. Our data collectively suggests that RGS21 modulates bitter taste signal transduction.
- Published
- 2012
- Full Text
- View/download PDF
29. Small molecule allosteric modulation of the glucagon-like Peptide-1 receptor enhances the insulinotropic effect of oxyntomodulin.
- Author
-
Willard FS, Wootten D, Showalter AD, Savage EE, Ficorilli J, Farb TB, Bokvist K, Alsina-Fernandez J, Furness SG, Christopoulos A, Sexton PM, and Sloop KW
- Subjects
- Animals, CHO Cells, Cell Line, Cricetinae, Cyclic AMP metabolism, Diabetes Mellitus, Type 2 drug therapy, Diabetes Mellitus, Type 2 metabolism, Drug Synergism, GTP-Binding Proteins metabolism, Glucagon-Like Peptide 1 metabolism, Glucagon-Like Peptide-1 Receptor, HEK293 Cells, Humans, Signal Transduction drug effects, Hypoglycemic Agents pharmacology, Insulin metabolism, Oxyntomodulin pharmacology, Receptors, Glucagon agonists, Receptors, Glucagon metabolism
- Abstract
Identifying novel mechanisms to enhance glucagon-like peptide-1 (GLP-1) receptor signaling may enable nascent medicinal chemistry strategies with the aim of developing new orally available therapeutic agents for the treatment of type 2 diabetes mellitus. Therefore, we tested the hypothesis that selectively modulating the low-affinity GLP-1 receptor agonist, oxyntomodulin, would improve the insulin secretory properties of this naturally occurring hormone to provide a rationale for pursuing an unexplored therapeutic approach. Signal transduction and competition binding studies were used to investigate oxyntomodulin activity on the GLP-1 receptor in the presence of the small molecule GLP-1 receptor modulator, 4-(3-benzyloxyphenyl)-2-ethylsulfinyl-6-(trifluoromethyl)pyrimidine (BETP). In vivo, the intravenous glucose tolerance test characterized oxyntomodulin-induced insulin secretion in animals administered the small molecule. BETP increased oxyntomodulin binding affinity for the GLP-1 receptor and enhanced oxyntomodulin-mediated GLP-1 receptor signaling as measured by activation of the α subunit of heterotrimeric G protein and cAMP accumulation. In addition, oxyntomodulin-induced insulin secretion was enhanced in the presence of the compound. BETP was pharmacologically characterized to induce biased signaling by oxyntomodulin. These studies demonstrate that small molecules targeting the GLP-1 receptor can increase binding and receptor activation of the endogenous peptide oxyntomodulin. The biased signaling engendered by BETP suggests that GLP-1 receptor mobilization of cAMP is the critical insulinotropic signaling event. Because of the unique metabolic properties of oxyntomodulin, identifying molecules that enhance its activity should be pursued to assess the efficacy and safety of this novel mechanism.
- Published
- 2012
- Full Text
- View/download PDF
30. Crystal structure of the human PRMT5:MEP50 complex.
- Author
-
Antonysamy S, Bonday Z, Campbell RM, Doyle B, Druzina Z, Gheyi T, Han B, Jungheim LN, Qian Y, Rauch C, Russell M, Sauder JM, Wasserman SR, Weichert K, Willard FS, Zhang A, and Emtage S
- Subjects
- Catalytic Domain, Crystallography, X-Ray, Dimerization, Humans, Models, Molecular, Protein Conformation, Adaptor Proteins, Signal Transducing chemistry, Protein-Arginine N-Methyltransferases chemistry
- Abstract
Protein arginine methyltransferases (PRMTs) play important roles in several cellular processes, including signaling, gene regulation, and transport of proteins and nucleic acids, to impact growth, differentiation, proliferation, and development. PRMT5 symmetrically di-methylates the two-terminal ω-guanidino nitrogens of arginine residues on substrate proteins. PRMT5 acts as part of a multimeric complex in concert with a variety of partner proteins that regulate its function and specificity. A core component of these complexes is the WD40 protein MEP50/WDR77/p44, which mediates interactions with binding partners and substrates. We have determined the crystal structure of human PRMT5 in complex with MEP50 (methylosome protein 50), bound to an S-adenosylmethionine analog and a peptide substrate derived from histone H4. The structure of the surprising hetero-octameric complex reveals the close interaction between the seven-bladed β-propeller MEP50 and the N-terminal domain of PRMT5, and delineates the structural elements of substrate recognition.
- Published
- 2012
- Full Text
- View/download PDF
31. Allosteric modulation of endogenous metabolites as an avenue for drug discovery.
- Author
-
Wootten D, Savage EE, Valant C, May LT, Sloop KW, Ficorilli J, Showalter AD, Willard FS, Christopoulos A, and Sexton PM
- Subjects
- Allosteric Regulation physiology, Animals, CHO Cells, Cricetinae, Cricetulus, Drug Discovery trends, Islets of Langerhans drug effects, Islets of Langerhans metabolism, Male, Neurotransmitter Agents metabolism, Peptide Hormones metabolism, Pharmaceutical Preparations administration & dosage, Protein Binding physiology, Rats, Rats, Sprague-Dawley, Rats, Wistar, Drug Discovery methods, Pharmaceutical Preparations metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and a key drug target class. Recently, allosteric drugs that can co-bind with and modulate the activity of the endogenous ligand(s) for the receptor have become a major focus of the pharmaceutical and biotechnology industry for the development of novel GPCR therapeutic agents. This class of drugs has distinct properties compared with drugs targeting the endogenous (orthosteric) ligand-binding site that include the ability to sculpt cellular signaling and to respond differently in the presence of discrete orthosteric ligands, a behavior termed "probe dependence." Here, using cell signaling assays combined with ex vivo and in vivo studies of insulin secretion, we demonstrate that allosteric ligands can cause marked potentiation of previously "inert" metabolic products of neurotransmitters and peptide hormones, a novel consequence of the phenomenon of probe dependence. Indeed, at the muscarinic M(2) receptor and glucagon-like peptide 1 (GLP-1) receptor, allosteric potentiation of the metabolites, choline and GLP-1(9-36)NH(2), respectively, was ~100-fold and up to 200-fold greater than that seen with the physiological signaling molecules acetylcholine and GLP-1(7-36)NH(2). Modulation of GLP-1(9-36)NH(2) was also demonstrated in ex vivo and in vivo assays of insulin secretion. This work opens up new avenues for allosteric drug discovery by directly targeting modulation of metabolites, but it also identifies a behavior that could contribute to unexpected clinical outcomes if interaction of allosteric drugs with metabolites is not part of their preclinical assessment.
- Published
- 2012
- Full Text
- View/download PDF
32. A P-loop mutation in Gα subunits prevents transition to the active state: implications for G-protein signaling in fungal pathogenesis.
- Author
-
Bosch DE, Willard FS, Ramanujam R, Kimple AJ, Willard MD, Naqvi NI, and Siderovski DP
- Subjects
- Amino Acid Substitution physiology, Catalytic Domain genetics, Crystallography, X-Ray, GTP-Binding Protein alpha Subunits chemistry, GTP-Binding Protein alpha Subunits metabolism, Hordeum microbiology, Magnaporthe genetics, Magnaporthe metabolism, Models, Molecular, Mutant Proteins chemistry, Mutant Proteins genetics, Mutant Proteins metabolism, Mutant Proteins physiology, Plant Diseases genetics, Plant Diseases microbiology, Plant Leaves microbiology, Protein Structure, Tertiary genetics, Signal Transduction genetics, GTP-Binding Protein alpha Subunits genetics, GTP-Binding Protein alpha Subunits physiology, Magnaporthe pathogenicity, Mycoses genetics, Point Mutation physiology, Protein Folding
- Abstract
Heterotrimeric G-proteins are molecular switches integral to a panoply of different physiological responses that many organisms make to environmental cues. The switch from inactive to active Gαβγ heterotrimer relies on nucleotide cycling by the Gα subunit: exchange of GTP for GDP activates Gα, whereas its intrinsic enzymatic activity catalyzes GTP hydrolysis to GDP and inorganic phosphate, thereby reverting Gα to its inactive state. In several genetic studies of filamentous fungi, such as the rice blast fungus Magnaporthe oryzae, a G42R mutation in the phosphate-binding loop of Gα subunits is assumed to be GTPase-deficient and thus constitutively active. Here, we demonstrate that Gα(G42R) mutants are not GTPase deficient, but rather incapable of achieving the activated conformation. Two crystal structure models suggest that Arg-42 prevents a typical switch region conformational change upon Gα(i1)(G42R) binding to GDP·AlF(4)(-) or GTP, but rotameric flexibility at this locus allows for unperturbed GTP hydrolysis. Gα(G42R) mutants do not engage the active state-selective peptide KB-1753 nor RGS domains with high affinity, but instead favor interaction with Gβγ and GoLoco motifs in any nucleotide state. The corresponding Gα(q)(G48R) mutant is not constitutively active in cells and responds poorly to aluminum tetrafluoride activation. Comparative analyses of M. oryzae strains harboring either G42R or GTPase-deficient Q/L mutations in the Gα subunits MagA or MagB illustrate functional differences in environmental cue processing and intracellular signaling outcomes between these two Gα mutants, thus demonstrating the in vivo functional divergence of G42R and activating G-protein mutants.
- Published
- 2012
- Full Text
- View/download PDF
33. Physiology and emerging biochemistry of the glucagon-like peptide-1 receptor.
- Author
-
Willard FS and Sloop KW
- Subjects
- Allosteric Site, Biochemistry methods, Crystallography, X-Ray methods, Cyclic AMP metabolism, Diabetes Mellitus, Type 2 genetics, Exenatide, Glucagon-Like Peptide 1 analogs & derivatives, Glucagon-Like Peptide 1 pharmacology, Glucagon-Like Peptide-1 Receptor, Glucose metabolism, Homeostasis, Humans, Ligands, Liraglutide, Models, Biological, Peptides chemistry, Peptides metabolism, Peptides pharmacology, Protein Binding, Receptors, Glucagon chemistry, Signal Transduction, Venoms metabolism, Venoms pharmacology, Diabetes Mellitus, Type 2 blood, Receptors, Glucagon physiology
- Abstract
The glucagon-like peptide-1 (GLP-1) receptor is one of the best validated therapeutic targets for the treatment of type 2 diabetes mellitus (T2DM). Over several years, the accumulation of basic, translational, and clinical research helped define the physiologic roles of GLP-1 and its receptor in regulating glucose homeostasis and energy metabolism. These efforts provided much of the foundation for pharmaceutical development of the GLP-1 receptor peptide agonists, exenatide and liraglutide, as novel medicines for patients suffering from T2DM. Now, much attention is focused on better understanding the molecular mechanisms involved in ligand induced signaling of the GLP-1 receptor. For example, advancements in biophysical and structural biology techniques are being applied in attempts to more precisely determine ligand binding and receptor occupancy characteristics at the atomic level. These efforts should better inform three-dimensional modeling of the GLP-1 receptor that will help inspire more rational approaches to identify and optimize small molecule agonists or allosteric modulators targeting the GLP-1 receptor. This article reviews GLP-1 receptor physiology with an emphasis on GLP-1 induced signaling mechanisms in order to highlight new molecular strategies that help determine desired pharmacologic characteristics for guiding development of future nonpeptide GLP-1 receptor activators.
- Published
- 2012
- Full Text
- View/download PDF
34. Small molecule drug discovery at the glucagon-like peptide-1 receptor.
- Author
-
Willard FS, Bueno AB, and Sloop KW
- Subjects
- Glucagon-Like Peptide-1 Receptor, Humans, Ligands, Receptors, Glucagon metabolism, Diabetes Mellitus, Type 2 drug therapy, Drug Discovery, Receptors, Glucagon agonists
- Abstract
The therapeutic success of peptide glucagon-like peptide-1 (GLP-1) receptor agonists for the treatment of type 2 diabetes mellitus has inspired discovery efforts aimed at developing orally available small molecule GLP-1 receptor agonists. Although the GLP-1 receptor is a member of the structurally complex class B1 family of GPCRs, in recent years, a diverse array of orthosteric and allosteric nonpeptide ligands has been reported. These compounds include antagonists, agonists, and positive allosteric modulators with intrinsic efficacy. In this paper, a comprehensive review of currently disclosed small molecule GLP-1 receptor ligands is presented. In addition, examples of "ligand bias" and "probe dependency" for the GLP-1 receptor are discussed; these emerging concepts may influence further optimization of known molecules or persuade designs of expanded screening strategies to identify novel chemical starting points for GLP-1 receptor drug discovery.
- Published
- 2012
- Full Text
- View/download PDF
35. Heterotrimeric G-protein signaling is critical to pathogenic processes in Entamoeba histolytica.
- Author
-
Bosch DE, Kimple AJ, Muller RE, Giguère PM, Machius M, Willard FS, Temple BR, and Siderovski DP
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Crystallography, X-Ray, Entamoeba histolytica enzymology, Entamoeba histolytica genetics, Entamoebiasis enzymology, Entamoebiasis genetics, GTP-Binding Protein alpha Subunits chemistry, GTP-Binding Protein alpha Subunits genetics, Gene Expression Regulation immunology, Guanine Nucleotide Exchange Factors chemistry, Guanine Nucleotide Exchange Factors genetics, Guanine Nucleotide Exchange Factors immunology, Humans, Jurkat Cells, Protein Structure, Tertiary, Protozoan Proteins chemistry, Protozoan Proteins genetics, Rho Guanine Nucleotide Exchange Factors, Transcription, Genetic immunology, Virulence Factors biosynthesis, Virulence Factors chemistry, Entamoeba histolytica immunology, Entamoebiasis immunology, GTP-Binding Protein alpha Subunits immunology, Protozoan Proteins immunology, Virulence Factors immunology
- Abstract
Heterotrimeric G-protein signaling pathways are vital components of physiology, and many are amenable to pharmacologic manipulation. Here, we identify functional heterotrimeric G-protein subunits in Entamoeba histolytica, the causative agent of amoebic colitis. The E. histolytica Gα subunit EhGα1 exhibits conventional nucleotide cycling properties and is seen to interact with EhGβγ dimers and a candidate effector, EhRGS-RhoGEF, in typical, nucleotide-state-selective fashions. In contrast, a crystal structure of EhGα1 highlights unique features and classification outside of conventional mammalian Gα subfamilies. E. histolytica trophozoites overexpressing wildtype EhGα1 in an inducible manner exhibit an enhanced ability to kill host cells that may be wholly or partially due to enhanced host cell attachment. EhGα1-overexpressing trophozoites also display enhanced transmigration across a Matrigel barrier, an effect that may result from altered baseline migration. Inducible expression of a dominant negative EhGα1 variant engenders the converse phenotypes. Transcriptomic studies reveal that modulation of pathogenesis-related trophozoite behaviors by perturbed heterotrimeric G-protein expression includes transcriptional regulation of virulence factors and altered trafficking of cysteine proteases. Collectively, our studies suggest that E. histolytica possesses a divergent heterotrimeric G-protein signaling axis that modulates key aspects of cellular processes related to the pathogenesis of this infectious organism.
- Published
- 2012
- Full Text
- View/download PDF
36. Open innovation for phenotypic drug discovery: The PD2 assay panel.
- Author
-
Lee JA, Chu S, Willard FS, Cox KL, Sells Galvin RJ, Peery RB, Oliver SE, Oler J, Meredith TD, Heidler SA, Gough WH, Husain S, Palkowitz AD, and Moxham CM
- Subjects
- Animals, Apolipoproteins E metabolism, Cell Cycle drug effects, Cell Differentiation drug effects, Cell Line, Drug Evaluation, Preclinical, HeLa Cells, Humans, Insulin metabolism, Insulin Secretion, Mice, Neovascularization, Physiologic drug effects, Nocodazole pharmacology, Osteoblasts cytology, Osteoblasts metabolism, Protein Kinase Inhibitors pharmacology, Rats, Reproducibility of Results, Signal Transduction drug effects, Tubulin Modulators pharmacology, Wnt Proteins metabolism, Drug Discovery, Phenotype
- Abstract
Phenotypic lead generation strategies seek to identify compounds that modulate complex, physiologically relevant systems, an approach that is complementary to traditional, target-directed strategies. Unlike gene-specific assays, phenotypic assays interrogate multiple molecular targets and signaling pathways in a target "agnostic" fashion, which may reveal novel functions for well-studied proteins and discover new pathways of therapeutic value. Significantly, existing compound libraries may not have sufficient chemical diversity to fully leverage a phenotypic strategy. To address this issue, Eli Lilly and Company launched the Phenotypic Drug Discovery Initiative (PD(2)), a model of open innovation whereby external research groups can submit compounds for testing in a panel of Lilly phenotypic assays. This communication describes the statistical validation, operations, and initial screening results from the first PD(2) assay panel. Analysis of PD(2) submissions indicates that chemical diversity from open source collaborations complements internal sources. Screening results for the first 4691 compounds submitted to PD(2) have confirmed hit rates from 1.6% to 10%, with the majority of active compounds exhibiting acceptable potency and selectivity. Phenotypic lead generation strategies, in conjunction with novel chemical diversity obtained via open-source initiatives such as PD(2), may provide a means to identify compounds that modulate biology by novel mechanisms and expand the innovation potential of drug discovery.
- Published
- 2011
- Full Text
- View/download PDF
37. Structural determinants of affinity enhancement between GoLoco motifs and G-protein alpha subunit mutants.
- Author
-
Bosch DE, Kimple AJ, Sammond DW, Muller RE, Miley MJ, Machius M, Kuhlman B, Willard FS, and Siderovski DP
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Computer Simulation, Crystallography, X-Ray, GTP-Binding Protein alpha Subunits chemistry, GTP-Binding Protein alpha Subunits genetics, Humans, Peptides chemical synthesis, Protein Binding genetics, Protein Conformation, Thermodynamics, GTP-Binding Protein alpha Subunits metabolism, Molecular Dynamics Simulation, Peptides metabolism
- Abstract
GoLoco motif proteins bind to the inhibitory G(i) subclass of G-protein α subunits and slow the release of bound GDP; this interaction is considered critical to asymmetric cell division and neuro-epithelium and epithelial progenitor differentiation. To provide protein tools for interrogating the precise cellular role(s) of GoLoco motif/Gα(i) complexes, we have employed structure-based protein design strategies to predict gain-of-function mutations that increase GoLoco motif binding affinity. Here, we describe fluorescence polarization and isothermal titration calorimetry measurements showing three predicted Gα(i1) point mutations, E116L, Q147L, and E245L; each increases affinity for multiple GoLoco motifs. A component of this affinity enhancement results from a decreased rate of dissociation between the Gα mutants and GoLoco motifs. For Gα(i1)(Q147L), affinity enhancement was seen to be driven by favorable changes in binding enthalpy, despite reduced contributions from binding entropy. The crystal structure of Gα(i1)(Q147L) bound to the RGS14 GoLoco motif revealed disorder among three peptide residues surrounding a well defined Leu-147 side chain. Monte Carlo simulations of the peptide in this region showed a sampling of multiple backbone conformations in contrast to the wild-type complex. We conclude that mutation of Glu-147 to leucine creates a hydrophobic surface favorably buried upon GoLoco peptide binding, yet the hydrophobic Leu-147 also promotes flexibility among residues 511-513 of the RGS14 GoLoco peptide.
- Published
- 2011
- Full Text
- View/download PDF
38. Novel small molecule glucagon-like peptide-1 receptor agonist stimulates insulin secretion in rodents and from human islets.
- Author
-
Sloop KW, Willard FS, Brenner MB, Ficorilli J, Valasek K, Showalter AD, Farb TB, Cao JX, Cox AL, Michael MD, Gutierrez Sanfeliciano SM, Tebbe MJ, and Coghlan MJ
- Subjects
- Animals, Cyclic AMP metabolism, Gastric Inhibitory Polypeptide pharmacology, Genes, Reporter, Glucagon pharmacology, Glucagon-Like Peptide 1 physiology, Glucagon-Like Peptide-1 Receptor, Glucose Tolerance Test, Humans, Insulin Secretion, Islets of Langerhans cytology, Islets of Langerhans drug effects, Luciferases genetics, Male, Parathyroid Hormone pharmacology, Rats, Rats, Sprague-Dawley, Receptors, Glucagon agonists, Vasoactive Intestinal Peptide pharmacology, Insulin metabolism, Islets of Langerhans metabolism, Receptors, Glucagon genetics
- Abstract
Objective: The clinical effectiveness of parenterally-administered glucagon-like peptide-1 (GLP-1) mimetics to improve glucose control in patients suffering from type 2 diabetes strongly supports discovery pursuits aimed at identifying and developing orally active, small molecule GLP-1 receptor agonists. The purpose of these studies was to identify and characterize novel nonpeptide agonists of the GLP-1 receptor., Research Design and Methods: Screening using cells expressing the GLP-1 receptor and insulin secretion assays with rodent and human islets were used to identify novel molecules. The intravenous glucose tolerance test (IVGTT) and hyperglycemic clamp characterized the insulinotropic effects of compounds in vivo., Results: Novel low molecular weight pyrimidine-based compounds that activate the GLP-1 receptor and stimulate glucose-dependent insulin secretion are described. These molecules induce GLP-1 receptor-mediated cAMP signaling in HEK293 cells expressing the GLP-1 receptor and increase insulin secretion from rodent islets in a dose-dependent manner. The compounds activate GLP-1 receptor signaling, both alone or in an additive fashion when combined with the endogenous GLP-1 peptide; however, these agonists do not compete with radiolabeled GLP-1 in receptor-binding assays. In vivo studies using the IVGTT and the hyperglycemic clamp in Sprague Dawley rats demonstrate increased insulin secretion in compound-treated animals. Further, perifusion assays with human islets isolated from a donor with type 2 diabetes show near-normalization of insulin secretion upon compound treatment., Conclusions: These studies characterize the insulinotropic effects of an early-stage, small molecule GLP-1 receptor agonist and provide compelling evidence to support pharmaceutical optimization.
- Published
- 2010
- Full Text
- View/download PDF
39. Regulators of G-protein signaling accelerate GPCR signaling kinetics and govern sensitivity solely by accelerating GTPase activity.
- Author
-
Lambert NA, Johnston CA, Cappell SD, Kuravi S, Kimple AJ, Willard FS, and Siderovski DP
- Subjects
- Alanine chemistry, Dose-Response Relationship, Drug, Glycine chemistry, Humans, Hydrolysis, Kinetics, Luminescence, Models, Molecular, Mutation, Pheromones metabolism, Saccharomyces cerevisiae metabolism, Signal Transduction, GTP Phosphohydrolases chemistry, GTP-Binding Proteins metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
G-protein heterotrimers, composed of a guanine nucleotide-binding G alpha subunit and an obligate G betagamma dimer, regulate signal transduction pathways by cycling between GDP- and GTP-bound states. Signal deactivation is achieved by G alpha-mediated GTP hydrolysis (GTPase activity) which is enhanced by the GTPase-accelerating protein (GAP) activity of "regulator of G-protein signaling" (RGS) proteins. In a cellular context, RGS proteins have also been shown to speed up the onset of signaling, and to accelerate deactivation without changing amplitude or sensitivity of the signal. This latter paradoxical activity has been variably attributed to GAP/enzymatic or non-GAP/scaffolding functions of these proteins. Here, we validated and exploited a G alpha switch-region point mutation, known to engender increased GTPase activity, to mimic in cis the GAP function of RGS proteins. While the transition-state, GDP x AlF(4)(-)-bound conformation of the G202A mutant was found to be nearly identical to wild-type, G alpha(i1)(G202A) x GDP assumed a divergent conformation more closely resembling the GDP x AlF(4)(-)-bound state. When placed within Saccharomyces cerevisiae G alpha subunit Gpa1, the fast-hydrolysis mutation restored appropriate dose-response behaviors to pheromone signaling in the absence of RGS-mediated GAP activity. A bioluminescence resonance energy transfer (BRET) readout of heterotrimer activation with high temporal resolution revealed that fast intrinsic GTPase activity could recapitulate in cis the kinetic sharpening (increased onset and deactivation rates) and blunting of sensitivity also engendered by RGS protein action in trans. Thus G alpha-directed GAP activity, the first biochemical function ascribed to RGS proteins, is sufficient to explain the activation kinetics and agonist sensitivity observed from G-protein-coupled receptor (GPCR) signaling in a cellular context.
- Published
- 2010
- Full Text
- View/download PDF
40. A capture coupling method for the covalent immobilization of hexahistidine tagged proteins for surface plasmon resonance.
- Author
-
Kimple AJ, Muller RE, Siderovski DP, and Willard FS
- Subjects
- Amines chemistry, Buffers, GTP-Binding Proteins analysis, GTP-Binding Proteins chemistry, GTP-Binding Proteins metabolism, Immobilized Proteins metabolism, Protein Multimerization, Protein Structure, Quaternary, Reproducibility of Results, Histidine metabolism, Immobilized Proteins analysis, Immobilized Proteins chemistry, Oligopeptides metabolism, Surface Plasmon Resonance methods
- Abstract
Surface plasmon resonance (SPR) is a robust method to detect and quantify macromolecular interactions; however, to measure binding interactions, one component must be immobilized on a sensor surface. This is typically achieved using covalent immobilization via free amines or thiols, or noncovalent immobilization using high-affinity interactions such as biotin/streptavidin or antibody/antigen. In this chapter we describe a robust method to covalently immobilize His(6) fusion proteins on the sensor surface for SPR analysis.
- Published
- 2010
- Full Text
- View/download PDF
41. High-affinity immobilization of proteins using biotin- and GST-based coupling strategies.
- Author
-
Hutsell SQ, Kimple RJ, Siderovski DP, Willard FS, and Kimple AJ
- Subjects
- Amino Acid Sequence, Antibodies, Immobilized chemistry, Antibodies, Immobilized immunology, Antibodies, Immobilized metabolism, Biotin chemistry, Biotinylation, Glutathione Transferase metabolism, Immobilized Proteins chemistry, Kinetics, Molecular Sequence Data, Peptides chemistry, Peptides metabolism, Recombinant Fusion Proteins immunology, Streptavidin chemistry, Streptavidin metabolism, Biotin metabolism, Glutathione Transferase immunology, Immobilized Proteins immunology, Immobilized Proteins metabolism, Surface Plasmon Resonance methods
- Abstract
Surface plasmon resonance (SPR) is a highly sensitive method for the detection of molecular interactions. One interacting partner is immobilized on the sensor chip surface while the other is injected across the sensor surface. This chapter focuses on high-affinity immobilization of protein substrates for affinity and kinetic analyses using biotin/streptavidin interaction and GST/anti-GST-antibody interaction.
- Published
- 2010
- Full Text
- View/download PDF
42. Structural determinants of G-protein alpha subunit selectivity by regulator of G-protein signaling 2 (RGS2).
- Author
-
Kimple AJ, Soundararajan M, Hutsell SQ, Roos AK, Urban DJ, Setola V, Temple BR, Roth BL, Knapp S, Willard FS, and Siderovski DP
- Subjects
- Binding Sites genetics, Cell Line, Evolution, Molecular, Fluorescence Resonance Energy Transfer, GTP-Binding Protein alpha Subunits genetics, Guanosine Triphosphate metabolism, Humans, Hydrolysis, Luminescent Proteins genetics, Luminescent Proteins metabolism, Models, Molecular, Point Mutation, Protein Binding, Protein Structure, Tertiary, RGS Proteins chemistry, RGS Proteins genetics, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Surface Plasmon Resonance, Transfection, GTP-Binding Protein alpha Subunits metabolism, Protein Interaction Domains and Motifs, RGS Proteins metabolism
- Abstract
"Regulator of G-protein signaling" (RGS) proteins facilitate the termination of G protein-coupled receptor (GPCR) signaling via their ability to increase the intrinsic GTP hydrolysis rate of Galpha subunits (known as GTPase-accelerating protein or "GAP" activity). RGS2 is unique in its in vitro potency and selectivity as a GAP for Galpha(q) subunits. As many vasoconstrictive hormones signal via G(q) heterotrimer-coupled receptors, it is perhaps not surprising that RGS2-deficient mice exhibit constitutive hypertension. However, to date the particular structural features within RGS2 determining its selectivity for Galpha(q) over Galpha(i/o) substrates have not been completely characterized. Here, we examine a trio of point mutations to RGS2 that elicits Galpha(i)-directed binding and GAP activities without perturbing its association with Galpha(q). Using x-ray crystallography, we determined a model of the triple mutant RGS2 in complex with a transition state mimetic form of Galpha(i) at 2.8-A resolution. Structural comparison with unliganded, wild type RGS2 and of other RGS domain/Galpha complexes highlighted the roles of these residues in wild type RGS2 that weaken Galpha(i) subunit association. Moreover, these three amino acids are seen to be evolutionarily conserved among organisms with modern cardiovascular systems, suggesting that RGS2 arose from the R4-subfamily of RGS proteins to have specialized activity as a potent and selective Galpha(q) GAP that modulates cardiovascular function.
- Published
- 2009
- Full Text
- View/download PDF
43. Regulator of G-protein signaling 14 (RGS14) is a selective H-Ras effector.
- Author
-
Willard FS, Willard MD, Kimple AJ, Soundararajan M, Oestreich EA, Li X, Sowa NA, Kimple RJ, Doyle DA, Der CJ, Zylka MJ, Snider WD, and Siderovski DP
- Subjects
- Animals, Binding Sites, Cell Differentiation, Extracellular Signal-Regulated MAP Kinases, Fibroblast Growth Factor 2 physiology, Humans, Mice, Mitogen-Activated Protein Kinases, Multiprotein Complexes, Nerve Growth Factor physiology, Neurites, PC12 Cells, Protein Binding, Rats, raf Kinases, Fibroblast Growth Factor 2 antagonists & inhibitors, Nerve Growth Factor antagonists & inhibitors, RGS Proteins physiology, ras Proteins metabolism
- Abstract
Background: Regulator of G-protein signaling (RGS) proteins have been well-described as accelerators of Galpha-mediated GTP hydrolysis ("GTPase-accelerating proteins" or GAPs). However, RGS proteins with complex domain architectures are now known to regulate much more than Galpha GTPase activity. RGS14 contains tandem Ras-binding domains that have been reported to bind to Rap- but not Ras GTPases in vitro, leading to the suggestion that RGS14 is a Rap-specific effector. However, more recent data from mammals and Drosophila imply that, in vivo, RGS14 may instead be an effector of Ras., Methodology/principal Findings: Full-length and truncated forms of purified RGS14 protein were found to bind indiscriminately in vitro to both Rap- and Ras-family GTPases, consistent with prior literature reports. In stark contrast, however, we found that in a cellular context RGS14 selectively binds to activated H-Ras and not to Rap isoforms. Co-transfection / co-immunoprecipitation experiments demonstrated the ability of full-length RGS14 to assemble a multiprotein complex with components of the ERK MAPK pathway in a manner dependent on activated H-Ras. Small interfering RNA-mediated knockdown of RGS14 inhibited both nerve growth factor- and basic fibrobast growth factor-mediated neuronal differentiation of PC12 cells, a process which is known to be dependent on Ras-ERK signaling., Conclusions/significance: In cells, RGS14 facilitates the formation of a selective Ras.GTP-Raf-MEK-ERK multiprotein complex to promote sustained ERK activation and regulate H-Ras-dependent neuritogenesis. This cellular function for RGS14 is similar but distinct from that recently described for its closely-related paralogue, RGS12, which shares the tandem Ras-binding domain architecture with RGS14.
- Published
- 2009
- Full Text
- View/download PDF
44. A point mutation to Galphai selectively blocks GoLoco motif binding: direct evidence for Galpha.GoLoco complexes in mitotic spindle dynamics.
- Author
-
Willard FS, Zheng Z, Guo J, Digby GJ, Kimple AJ, Conley JM, Johnston CA, Bosch D, Willard MD, Watts VJ, Lambert NA, Ikeda SR, Du Q, and Siderovski DP
- Subjects
- 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, GTP-Binding Protein alpha Subunits metabolism, Spindle Apparatus
- 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.
- Published
- 2008
- Full Text
- View/download PDF
45. A sweet cycle for Arabidopsis G-proteins: Recent discoveries and controversies in plant G-protein signal transduction.
- Author
-
Johnston CA, Willard MD, Kimple AJ, Siderovski DP, and Willard FS
- Abstract
Heterotrimeric G-proteins are a class of signal transduction proteins highly conserved throughout evolution that serve as dynamic molecular switches regulating the intracellular communication initiated by extracellular signals including sensory information. This property is achieved by a guanine nucleotide cycle wherein the inactive, signaling-incompetent Galpha subunit is normally bound to GDP; activation to signaling-competent Galpha occurs through the exchange of GDP for GTP (typically catalyzed via seven-transmembrane domain G-protein coupled receptors [GPCRs]), which dissociates the Gbetagamma dimer from Galpha-GTP and initiates signal transduction. The hydrolysis of GTP, greatly accelerated by "Regulator of G-protein Signaling" (RGS) proteins, returns Galpha to its inactive GDP-bound form and terminates signaling. Through extensive characterization of mammalian Galpha isoforms, the rate-limiting step in this cycle is currently considered to be the GDP/GTP exchange rate, which can be orders of magnitude slower than the GTP hydrolysis rate. However, we have recently demonstrated that, in Arabidopsis, the guanine nucleotide cycle appears to be limited by the rate of GTP hydrolysis rather than nucleotide exchange. This finding has important implications for the mechanism of sugar sensing in Arabidopsis. We also discuss these data on Arabidopsis G-protein nucleotide cycling in relation to recent reports of putative plant GPCRs and heterotrimeric G-protein effectors in Arabidopsis.
- Published
- 2008
- Full Text
- View/download PDF
46. Structural determinants underlying the temperature-sensitive nature of a Galpha mutant in asymmetric cell division of Caenorhabditis elegans.
- Author
-
Johnston CA, Afshar K, Snyder JT, Tall GG, Gönczy P, Siderovski DP, and Willard FS
- Subjects
- Animals, Cell Division, Circular Dichroism, Crystallography, X-Ray methods, Guanosine Triphosphate chemistry, Humans, Models, Biological, Mutation, Point Mutation, Protein Conformation, Surface Plasmon Resonance, Temperature, Caenorhabditis elegans metabolism, GTP-Binding Protein alpha Subunits, Gi-Go genetics, Gene Expression Regulation
- Abstract
Heterotrimeric G-proteins are integral to a conserved regulatory module that influences metazoan asymmetric cell division (ACD). In the Caenorhabditis elegans zygote, GOA-1 (Galpha(o)) and GPA-16 (Galpha(i)) are involved in generating forces that pull on astral microtubules and position the spindle asymmetrically. GPA-16 function has been analyzed in vivo owing notably to a temperature-sensitive allele gpa-16(it143), which, at the restrictive temperature, results in spindle orientation defects in early embryos. Here we identify the structural basis of gpa-16(it143), which encodes a point mutation (G202D) in the switch II region of GPA-16. Using Galpha(i1)(G202D) as a model in biochemical analyses, we demonstrate that high temperature induces instability of the mutant Galpha. At the permissive temperature, the mutant Galpha was stable upon GTP binding, but switch II rearrangement was compromised, as were activation state-selective interactions with regulators involved in ACD, including GoLoco motifs, RGS proteins, and RIC-8. We solved the crystal structure of the mutant Galpha bound to GDP, which indicates a unique switch II conformation as well as steric constraints that suggest activated GPA-16(it143) is destabilized relative to wild type. Spindle severing in gpa-16(it143) embryos revealed that pulling forces are symmetric and markedly diminished at the restrictive temperature. Interestingly, pulling forces are asymmetric and generally similar in magnitude to wild type at the permissive temperature despite defects in the structure of GPA-16(it143). These normal pulling forces in gpa-16(it143) embryos at the permissive temperature were attributable to GOA-1 function, underscoring a complex interplay of Galpha subunit function in ACD.
- Published
- 2008
- Full Text
- View/download PDF
47. A high throughput fluorescence polarization assay for inhibitors of the GoLoco motif/G-alpha interaction.
- Author
-
Kimple AJ, Yasgar A, Hughes M, Jadhav A, Willard FS, Muller RE, Austin CP, Inglese J, Ibeanu GC, Siderovski DP, and Simeonov A
- Subjects
- Amino Acid Motifs, GTP Phosphohydrolase Activators metabolism, GTP-Binding Protein alpha Subunits, Gi-Go analysis, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Humans, Models, Molecular, Molecular Sequence Data, RGS Proteins metabolism, Staining and Labeling, Biological Assay methods, Fluorescence Polarization methods, GTP-Binding Protein alpha Subunits, Gi-Go antagonists & inhibitors
- Abstract
The GoLoco motif is a short Galpha-binding polypeptide sequence. It is often found in proteins that regulate cell-surface receptor signaling, such as RGS12, as well as in proteins that regulate mitotic spindle orientation and force generation during cell division, such as GPSM2/LGN. Here, we describe a high throughput fluorescence polarization (FP) assay using fluorophore-labeled GoLoco motif peptides for identifying inhibitors of the GoLoco motif interaction with the G-protein alpha subunit Galpha (i1). The assay exhibits considerable stability over time and is tolerant to DMSO up to 5%. The Z'-factors for robustness of the GPSM2 and RGS12 GoLoco motif assays in a 96-well plate format were determined to be 0.81 and 0.84, respectively; the latter assay was run in a 384-well plate format and produced a Z'-factor of 0.80. To determine the screening factor window (Z-factor) of the RGS12 GoLoco motif screen using a small molecule library, the NCI Diversity Set was screened. The Z-factor was determined to be 0.66, suggesting that this FP assay would perform well when developed for 1,536-well format and scaled up to larger libraries. We then miniaturized to a 4 microL final volume a pair of FP assays utilizing fluorescein- (green) and rhodamine- (red) labeled RGS12 GoLoco motif peptides. In a fully-automated run, the Sigma-Aldrich LOPAC(1280) collection was screened three times with every library compound being tested over a range of concentrations following the quantitative high throughput screening (qHTS) paradigm; excellent assay performance was noted with average Z-factors of 0.84 and 0.66 for the green- and red-label assays, respectively.
- Published
- 2008
- Full Text
- View/download PDF
48. GPCR high throughput screening (part 1).
- Author
-
Willard FS and Siderovski DP
- Subjects
- Biological Assay, Drug Design, Drug Evaluation, Preclinical, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled metabolism
- Published
- 2008
- Full Text
- View/download PDF
49. State-selective binding peptides for heterotrimeric G-protein subunits: novel tools for investigating G-protein signaling dynamics.
- Author
-
Johnston CA, Willard FS, Ramer JK, Blaesius R, Roques CN, and Siderovski DP
- Subjects
- Amino Acid Sequence, Drug Evaluation, Preclinical, Heterotrimeric GTP-Binding Proteins genetics, Molecular Sequence Data, Sensitivity and Specificity, Heterotrimeric GTP-Binding Proteins chemistry, Heterotrimeric GTP-Binding Proteins metabolism, Peptides chemistry, Peptides metabolism, Signal Transduction
- Abstract
Heterotrimeric G-proteins, comprising Galpha, Gbeta, and Ggamma subunits, are molecular switches that regulate numerous signaling pathways involved in cellular physiology. This characteristic is achieved by the adoption of two principal states: an inactive state in which GDP-bound Galpha is complexed with the Gbetagamma dimer, and an active state in which GTP-bound Galpha is freed of its Gbetagamma binding partner. Structural studies have illustrated the basis for the distinct conformations of these states which are regulated by alterations in three precise 'switch regions' of the Galpha subunit. Discrete differences in conformation between GDP- and GTP-bound Galpha underlie its nucleotide-dependent protein-protein interactions (e.g., with Gbetagamma/receptor and effectors, respectively) that are critical for maintaining their proper nucleotide cycling and signaling properties. Recently, several screening approaches have been used to identify peptide sequences capable of interacting with Galpha (and free Gbetagamma) in nucleotide-dependent fashions. These peptides have demonstrated applications in direct modulation of the nucleotide cycle, assessing the structural basis for aspects of Galpha and Gbetagamma signaling, and serving as biosensor tools in assays for Galpha activation including high throughput drug screening. In this review, we highlight some of the methods used for such discoveries and discuss the insights that can be gleaned from application of these identified peptides.
- Published
- 2008
- Full Text
- View/download PDF
50. Structural diversity in the RGS domain and its interaction with heterotrimeric G protein alpha-subunits.
- Author
-
Soundararajan M, Willard FS, Kimple AJ, Turnbull AP, Ball LJ, Schoch GA, Gileadi C, Fedorov OY, Dowler EF, Higman VA, Hutsell SQ, Sundström M, Doyle DA, and Siderovski DP
- Subjects
- Apoproteins metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 chemistry, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Humans, Models, Molecular, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Protein Subunits chemistry, Protein Subunits metabolism, GTP-Binding Protein alpha Subunits metabolism, RGS Proteins chemistry, RGS Proteins metabolism
- Abstract
Regulator of G protein signaling (RGS) proteins accelerate GTP hydrolysis by Galpha subunits and thus facilitate termination of signaling initiated by G protein-coupled receptors (GPCRs). RGS proteins hold great promise as disease intervention points, given their signature role as negative regulators of GPCRs-receptors to which the largest fraction of approved medications are currently directed. RGS proteins share a hallmark RGS domain that interacts most avidly with Galpha when in its transition state for GTP hydrolysis; by binding and stabilizing switch regions I and II of Galpha, RGS domain binding consequently accelerates Galpha-mediated GTP hydrolysis. The human genome encodes more than three dozen RGS domain-containing proteins with varied Galpha substrate specificities. To facilitate their exploitation as drug-discovery targets, we have taken a systematic structural biology approach toward cataloging the structural diversity present among RGS domains and identifying molecular determinants of their differential Galpha selectivities. Here, we determined 14 structures derived from NMR and x-ray crystallography of members of the R4, R7, R12, and RZ subfamilies of RGS proteins, including 10 uncomplexed RGS domains and 4 RGS domain/Galpha complexes. Heterogeneity observed in the structural architecture of the RGS domain, as well as in engagement of switch III and the all-helical domain of the Galpha substrate, suggests that unique structural determinants specific to particular RGS protein/Galpha pairings exist and could be used to achieve selective inhibition by small molecules.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.