Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(13): e2116506119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35333651

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.


Subject(s)
Diabetes Mellitus, Type 2 , Receptors, Gastrointestinal Hormone , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/therapeutic use , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Incretins/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/therapeutic use
2.
J Med Chem ; 64(6): 3439-3448, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33721487

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)NH2 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 ß-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.


Subject(s)
Allosteric Regulation/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , 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
3.
Proc Natl Acad Sci U S A ; 117(47): 29959-29967, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33177239

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 ß-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.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/pharmacology , Protein Domains/genetics , 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
4.
Biochem Biophys Res Commun ; 530(1): 246-251, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32828294

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.


Subject(s)
Exenatide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Peptide Hydrolases/metabolism , Protein Engineering/methods , 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
5.
JCI Insight ; 5(17)2020 09 03.
Article in English | MEDLINE | ID: mdl-32730231

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 ß-arrestin recruitment, coincident with a weaker ability to drive GLP-1 receptor internalization compared with GLP-1. Experiments in primary islets reveal ß-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.


Subject(s)
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 , Animals , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Knockout , beta-Arrestin 1/physiology
6.
Nat Chem Biol ; 16(10): 1105-1110, 2020 10.
Article in English | MEDLINE | ID: mdl-32690941

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 Gs. 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.


Subject(s)
Allosteric Regulation/drug effects , Glucagon-Like Peptide-1 Receptor/metabolism , Allosteric Site , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide-1 Receptor/chemistry , Models, Molecular , Molecular Structure , Protein Conformation
7.
J Biol Chem ; 291(20): 10700-15, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26975372

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.


Subject(s)
Glucagon-Like Peptide-1 Receptor/metabolism , 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
9.
Diabetes ; 64(3): 819-27, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25288673

ABSTRACT

The absence of insulin results in oscillating hyperglycemia and ketoacidosis in type 1 diabetes. Remarkably, mice genetically deficient in the glucagon receptor (Gcgr) are refractory to the pathophysiological symptoms of insulin deficiency, and therefore, studies interrogating this unique model may uncover metabolic regulatory mechanisms that are independent of insulin. A significant feature of Gcgr-null mice is the high circulating concentrations of GLP-1. Hence, the objective of this report was to investigate potential noninsulinotropic roles of GLP-1 in mice where GCGR signaling is inactivated. For these studies, pancreatic ß-cells were chemically destroyed by streptozotocin (STZ) in Gcgr(-/-):Glp-1r(-/-) mice and in Glp-1r(-/-) animals that were subsequently treated with a high-affinity GCGR antagonist antibody that recapitulates the physiological state of Gcgr ablation. Loss of GLP-1 action substantially worsened nonfasting glucose concentrations and glucose tolerance in mice deficient in, and undergoing pharmacological inhibition of, the GCGR. Further, lack of the Glp-1r in STZ-treated Gcgr(-/-) mice elevated rates of endogenous glucose production, likely accounting for the differences in glucose homeostasis. These results support the emerging hypothesis that non-ß-cell actions of GLP-1 analogs may improve metabolic control in patients with insulinopenic diabetes.


Subject(s)
Glucagon/metabolism , Glucose/metabolism , Insulin/metabolism , Receptors, Glucagon/deficiency , Animals , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Immunohistochemistry , Mice , Mice, Knockout , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Streptozocin/pharmacology
10.
PLoS One ; 9(9): e105683, 2014.
Article in English | MEDLINE | ID: mdl-25180755

ABSTRACT

Activation of the glucagon-like peptide-1 receptor (GLP-1R) in pancreatic ß-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.


Subject(s)
Receptors, Glucagon/chemistry , Receptors, Glucagon/metabolism , 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
11.
Mol Cell Proteomics ; 13(11): 3049-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25044020

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 ß 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 ß 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 ß 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 ß cells was shown to interact with the activated GLP-1R to enhance the insulinotropic actions of GLP-1.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Membrane Proteins/metabolism , Receptors, Glucagon/metabolism , Receptors, Progesterone/metabolism , 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
12.
PLoS One ; 9(4): e93746, 2014.
Article in English | MEDLINE | ID: mdl-24695667

ABSTRACT

Class B G protein-coupled receptors (GPCRs) are important regulators of endocrine physiology, and peptide-based therapeutics targeting some of these receptors have proven effective at treating disorders such as hypercalcemia, osteoporosis, and type 2 diabetes mellitus (T2DM). As next generation efforts attempt to develop novel non-peptide, orally available molecules for these GPCRs, new animal models expressing human receptor orthologs may be required because small molecule ligands make fewer receptor contacts, and thus, the impact of amino acid differences across species may be substantially greater. The objective of this report was to generate and characterize a new mouse model of the human glucagon-like peptide-1 receptor (hGLP-1R), a class B GPCR for which established peptide therapeutics exist for the treatment of T2DM. hGLP-1R knock-in mice express the receptor from the murine Glp-1r locus. Glucose tolerance tests and gastric emptying studies show hGLP-1R mice and their wild-type littermates display similar physiological responses for glucose metabolism, insulin secretion, and gastric transit, and treatment with the GLP-1R agonist, exendin-4, elicits similar responses in both groups. Further, ex vivo assays show insulin secretion from humanized islets is glucose-dependent and enhanced by GLP-1R agonists. To enable additional utility, the targeting construct of the knock-in line was engineered to contain both flanking LoxP sites and a C-terminal FLAG epitope. Anti-FLAG affinity purification shows strong expression of hGLP-1R in islets, lung, and stomach. We crossed the hGLP-1R line with Rosa26Cre mice and generated global Glp-1r-/- animals. Immunohistochemistry of pancreas from humanized and knock-out mice identified a human GLP-1R-specific antibody that detects the GLP-1R in human pancreas as well as in the pancreas of hGLP-1r knock-in mice. This new hGLP-1R model will allow tissue-specific deletion of the GLP-1R, purification of potential GLP-1R partner proteins, and testing of novel therapeutic agents targeting the hGLP-1R.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Receptors, Glucagon/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Glucagon-Like Peptide-1 Receptor , Glucose/metabolism , Glucose Tolerance Test , Humans , Insulin/metabolism , Mice , Mice, Transgenic , Pancreas/metabolism , Receptors, Glucagon/genetics
13.
Mol Pharmacol ; 82(6): 1066-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22930710

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.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin/metabolism , Oxyntomodulin/pharmacology , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , 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
14.
Mol Pharmacol ; 82(2): 281-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22576254

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.


Subject(s)
Drug Discovery/methods , Pharmaceutical Preparations/metabolism , Receptors, G-Protein-Coupled/metabolism , 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
15.
Proc Natl Acad Sci U S A ; 108(1): 173-8, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21149718

ABSTRACT

The etiology of most pediatric hormone deficiency diseases is poorly understood. Children with combined pituitary hormone deficiency (CPHD) have insufficient levels of multiple anterior pituitary hormones causing short stature, metabolic disease, pubertal failure, and often have associated nervous system symptoms. Mutations in developmental regulatory genes required for the specification of the hormone-secreting cell types of the pituitary gland underlie severe forms of CPHD. To better understand these diseases, we have created a unique mouse model of CPHD with a targeted knockin mutation (Lhx3 W227ter), which is a model for the human LHX3 W224ter disease. The LHX3 gene encodes a LIM-homeodomain transcription factor, which has essential roles in pituitary and nervous system development in mammals. The introduced premature termination codon results in deletion of the carboxyl terminal region of the LHX3 protein, which is critical for pituitary gene activation. Mice that lack all LHX3 function do not survive beyond birth. By contrast, the homozygous Lhx3 W227ter mice survive, but display marked dwarfism, thyroid disease, and female infertility. Importantly, the Lhx3 W227ter mice have no apparent nervous system deficits. The Lhx3 W227ter mouse model provides a unique array of hormone deficits and facilitates experimental approaches that are not feasible with human patients. These experiments demonstrate that the carboxyl terminus of the LHX3 transcription factor is not required for viability. More broadly, this study reveals that the in vivo actions of a transcription factor in different tissues are molecularly separable.


Subject(s)
Disease Models, Animal , Homeodomain Proteins/metabolism , Hypopituitarism/etiology , Models, Biological , Pituitary Hormones/deficiency , Animals , Blotting, Western , Codon, Nonsense/genetics , Female , Gene Knock-In Techniques , Histological Techniques , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , LIM-Homeodomain Proteins , Male , Mice , Mice, Transgenic , Polymerase Chain Reaction , Transcription Factors
16.
Diabetes ; 59(12): 3099-107, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20823098

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.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Receptors, Glucagon/genetics , 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
17.
Amyloid ; 17(2): 43-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20462362

ABSTRACT

Leptomeningeal amyloidosis associated with mutations in transthyretin (TTR) is a rare but fatal form of amyloidosis. Dementia and intracerebral haemorrhage are prominent features of this disease for which no specific therapy is known. In previous studies, we have shown that antisense oligonucleotides (ASOs) specific for human TTR could inhibit hepatic synthesis of TTR in mice transgenic for a human amyloid-associated TTR and may offer a medical means of treating systemic TTR amyloidosis. Parenteral administration of TTR-specific ASO, however, had no effect on the expression of TTR by the choroid plexus, which is believed to be the source of the amyloid protein in patients who have leptomeningeal amyloidosis. In the present study, mice transgenic for the human TTR amyloid-associated mutation Ile84Ser were treated by administration of TTR-specific ASO (50 microg or 75 microg per day) via an osmotic pump into the cerebral ventricular system over a 4-week period. Intraventricular administration of TTR-specific ASO significantly reduced choroid human TTR mRNA levels, and these findings correlated with decreased TTR in choroid plexus epithelial cells as demonstrated by immunohistochemistry. Suppression of choroid TTR expression by intraventricular administered ASO may offer a medical means of treating leptomeningeal amyloidosis.


Subject(s)
Choroid Plexus/metabolism , Oligonucleotides, Antisense/pharmacology , Prealbumin/biosynthesis , Amyloid Neuropathies, Familial/drug therapy , Animals , Choroid Plexus/drug effects , Humans , Mice , Mice, Transgenic , Prealbumin/genetics
18.
J Biol Chem ; 282(26): 19113-21, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17478431

ABSTRACT

D-Glucose-6-phosphatase is a key regulator of endogenous glucose production, and its inhibition may improve glucose control in type 2 diabetes. Herein, 2'-O-(2-methoxy)ethyl-modified phosphorothioate antisense oligonucleotides (ASOs) specific to the glucose 6-phosphate transporter-1 (G6PT1) enabled reduction of hepatic D-Glu-6-phosphatase activity in diabetic ob/ob mice. Treatment with G6PT1 ASOs decreased G6PT1 expression, reduced G6PT1 activity, blunted glucagon-stimulated glucose production, and lowered plasma glucose concentration in a dose-dependent manner. In contrast to G6PT1 knock-out mice and patients with glycogen storage disease, excess hepatic and renal glycogen accumulation, hyperlipidemia, neutropenia, and elevations in plasma lactate and uric acid did not occur. In addition, hypoglycemia was not observed in animals during extended periods of fasting, and the ability of G6PT1 ASO-treated mice to recover from an exogenous insulin challenge was not impaired. Together, these results demonstrate that effective glucose lowering by G6PT1 inhibitors can be achieved without adversely affecting carbohydrate and lipid metabolism.


Subject(s)
Antiporters/genetics , Antiporters/metabolism , Diabetes Mellitus, Type 2/therapy , Glycogen Storage Disease/prevention & control , Liver/metabolism , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Oligoribonucleotides, Antisense/pharmacology , Acidosis, Lactic/metabolism , Acidosis, Lactic/prevention & control , Animals , Blood Glucose/biosynthesis , Blood Glucose/metabolism , Diabetes Complications/metabolism , Diabetes Complications/prevention & control , Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Glucose-6-Phosphatase/metabolism , Glycogen/metabolism , Glycogen Storage Disease/metabolism , Hyperlipidemias/metabolism , Hyperlipidemias/prevention & control , Hyperuricemia/metabolism , Hyperuricemia/prevention & control , Hypoglycemia/metabolism , Hypoglycemia/prevention & control , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , RNA, Messenger/metabolism
19.
Muscle Nerve ; 33(5): 609-18, 2006 May.
Article in English | MEDLINE | ID: mdl-16421881

ABSTRACT

Transthyretin (TTR) amyloidosis, the most common form of hereditary systemic amyloidosis, is characterized clinically by adult-onset axonal neuropathy and restrictive cardiomyopathy. More than 85 mutations in transthyretin have been found to cause this hereditary disease. Since essentially all circulating TTR is of hepatic origin, orthotopic liver transplantation has been used as the only specific form of therapy. Unfortunately, in many patients amyloid deposition continues after orthotopic liver transplantation, indicating that mutant TTR is no longer required for progression of the disease after tissue deposits have been initiated. As a first step toward medical treatment of this disease, we have employed antisense oligonucleotides (ASOs) to inhibit hepatic expression of TTR. A transgenic mouse model carrying the human TTR Ile84Ser mutation was created and shown to express high levels of human mutant transthyretin. TTR ASOs suppressed hepatic TTR mRNA levels and serum TTR levels by as much as 80%. Suppression of hepatic synthesis of transthyretin may offer a medical treatment for transthyretin systemic amyloidosis.


Subject(s)
Amyloidosis, Familial/drug therapy , Amyloidosis, Familial/genetics , Gene Silencing , Oligonucleotides, Antisense/therapeutic use , Prealbumin/antagonists & inhibitors , Alanine Transaminase/blood , Amyloidosis, Familial/blood , Animals , Aspartate Aminotransferases/blood , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression/drug effects , Humans , Immunohistochemistry/methods , Isoleucine/genetics , Liver/drug effects , Liver/enzymology , Mice , Mice, Transgenic , Mutation/physiology , Prealbumin/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Serine/genetics , Time Factors
20.
Gene ; 336(2): 263-73, 2004 Jul 21.
Article in English | MEDLINE | ID: mdl-15246537

ABSTRACT

Prophet of Pit-1 (PROP1) is a homeodomain transcription factor essential for development of the mammalian anterior pituitary gland. Studies of human patients and animal models with mutations in their Prop1 genes have established that PROP1 is required for the correct development or sustained function of the hormone-secreting cells that regulate physiological pathways controlling growth, reproduction, metabolism, and the stress response. By comparative analysis of mammalian Prop1 genes and their encoded proteins, including cloning the ovine Prop1 gene and its products, we demonstrate that two conserved basic regions (B1 and B2) of the PROP1 protein located within the homeodomain are required for nuclear localization, DNA binding, and target gene activation. Interestingly, missense mutations in the human Prop1 gene causing amino acid changes in both the B1 and B2 regions have been associated with combined pituitary hormone deficiency (CPHD) diseases, suggesting that disruption of nuclear localization may be part of the molecular basis of such diseases. The ovine Prop1 gene has three exons and two introns, a different structure compared with that of the bovine gene. Two alleles of the ovine gene were found to encode protein products with different carboxyl terminal domain sequences. We demonstrate that the two alleles are distributed in different breeds of sheep. Finally, we show for the first time that the PROP1 protein is associated with the nuclear matrix.


Subject(s)
Conserved Sequence/genetics , Homeodomain Proteins/genetics , Sheep/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Cloning, Molecular , DNA/chemistry , DNA/genetics , DNA Probes/genetics , DNA Probes/metabolism , DNA, Complementary/chemistry , DNA, Complementary/genetics , Electrophoretic Mobility Shift Assay , Exons , Genes/genetics , Green Fluorescent Proteins , Homeodomain Proteins/metabolism , Humans , Introns , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Nuclear Localization Signals/genetics , Nuclear Matrix/metabolism , Phylogeny , Pituitary Gland/metabolism , Polymorphism, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...