Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Eur J Endocrinol ; 190(1): K8-K16, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38123488

ABSTRACT

OBJECTIVE: Somatostatin receptor ligands have come to play a pivotal role in the treatment of both ACTH- and GH-secreting pituitary adenomas. Clinical efficacy averages 30-50%, thus a considerable number of patients with Cushing's disease or acromegaly remain unresponsive to this therapeutic approach. HTL0030310 is a new somatostatin receptor ligand selective for subtype 5 over subtype 2, thus with a different receptor profile compared to clinical somatostatin receptor ligands. DESIGN: Assessment of the effect of HTL0030310 on hormone secretion in human ACTH- and GH-secreting pituitary adenomas in vitro. METHODS: Primary cultures from 3 ACTH-secreting and 5 GH-secreting pituitary adenomas were treated with 1, 10 and 100 nM HTL0030310 alone or with 10 nM CRH or GHRH, respectively. Parallel incubations with 10 nM pasireotide were also carried out. ACTH and GH secretion were assessed after 4 and 24 hour incubation; SSTR2, SSTR3, SSTR5, GH and POMC expression were evaluated after 24 hours. RESULTS: HTL0030310 reduced unchallenged ACTH and POMC levels up to 50% in 2 ACTH-secreting adenomas and blunted CRH-stimulated ACTH/POMC by 20-70% in all 3 specimens. A reduction in spontaneous GH secretion was observed in 4 GH-secreting adenomas and in 2 specimens during GHRH co-incubation. SSTRs expression was detected in all specimens. CONCLUSIONS: This first study on a novel somatostatin receptor 5-preferring ligand indicates that HTL0030310 can inhibit hormonal secretion in human ACTH- and GH-secreting pituitary adenomas. These findings suggest a potential new avenue for somatostatin ligands in the treatment of Cushing's disease and acromegaly.


Subject(s)
Acromegaly , Adenoma , Growth Hormone-Secreting Pituitary Adenoma , Pituitary ACTH Hypersecretion , Pituitary Neoplasms , Humans , Receptors, Somatostatin/metabolism , Pituitary Neoplasms/drug therapy , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Acromegaly/drug therapy , Pro-Opiomelanocortin/metabolism , Pituitary ACTH Hypersecretion/drug therapy , Ligands , Adenoma/metabolism , Adrenocorticotropic Hormone/metabolism
2.
Eur J Pharmacol ; 954: 175897, 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37394028

ABSTRACT

Capsaicin and allyl isothiocyanate (AITC) activate transient receptor potential (TRP) vanilloid-1 (TRPV1) and TRP ankyrin-1 (TRPA1), respectively. TRPV1 and TRPA1 expression have been identified in the gastrointestinal (GI) tract. GI mucosal functions remain largely undefined for TRPV1 and TRPA1 with side-dependence and regional differences in signalling unclear. Here we investigated TRPV1- and TRPA1-induced vectorial ion transport as changes in short-circuit current (ΔIsc), in defined segments of mouse colon mucosa (ascending, transverse and descending) under voltage-clamp conditions in Ussing chambers. Drugs were applied basolaterally (bl) or apically (ap). Capsaicin responses were biphasic, with primary secretory and secondary anti-secretory phases, observed with bl application only, which predominated in descending colon. AITC responses were monophasic and secretory, with ΔIsc dependent on colonic region (ascending vs. descending) and sidedness (bl vs. ap). Aprepitant (neurokinin-1 (NK1) antagonist, bl) and tetrodotoxin (Na+ channel blocker, bl) significantly inhibited capsaicin primary responses in descending colon, while GW627368 (EP4 receptor antagonist, bl) and piroxicam (cyclooxygenase inhibitor, bl) inhibited AITC responses in ascending and descending colonic mucosae. Antagonism of the calcitonin gene-related peptide (CGRP) receptor had no effect on mucosal TRPV1 signalling, while tetrodotoxin and antagonists of the 5-hydroxytryptamine-3 and 4 receptors, CGRP receptor, and EP1/2/3 receptors had no effect on mucosal TRPA1 signalling. Our data demonstrates the regional-specificity and side-dependence of colonic TRPV1 and TRPA1 signalling, with involvement of submucosal neurons and mediation by epithelial NK1 receptor activation for TRPV1, and endogenous prostaglandins and EP4 receptor activation for TRPA1 mucosal responses.


Subject(s)
Transient Receptor Potential Channels , Mice , Animals , TRPA1 Cation Channel , Capsaicin/pharmacology , Tetrodotoxin , Colon/metabolism , Mucous Membrane/metabolism , TRPV Cation Channels/metabolism
3.
Front Endocrinol (Lausanne) ; 14: 1139121, 2023.
Article in English | MEDLINE | ID: mdl-36967803

ABSTRACT

Introduction: The apelin receptor binds two distinct endogenous peptides, apelin and ELA, which act in an autocrine/paracrine manner to regulate the human cardiovascular system. As a class A GPCR, targeting the apelin receptor is an attractive therapeutic strategy. With improvements in imaging techniques, and the stability and brightness of dyes, fluorescent ligands are becoming increasingly useful in studying protein targets. Here, we describe the design and validation of four novel fluorescent ligands; two based on [Pyr1]apelin-13 (apelin488 and apelin647), and two based on ELA-14 (ELA488 and ELA647). Methods: Fluorescent ligands were pharmacologically assessed using radioligand and functional in vitro assays. Apelin647 was validated in high content imaging and internalisation studies, and in a clinically relevant human embryonic stem cell-derived cardiomyocyte model. Apelin488 and ELA488 were used to visualise apelin receptor binding in human renal tissue. Results: All four fluorescent ligands retained the ability to bind and activate the apelin receptor and, crucially, triggered receptor internalisation. In high content imaging studies, apelin647 bound specifically to CHO-K1 cells stably expressing apelin receptor, providing proof-of-principle for a platform that could screen novel hits targeting this GPCR. The ligand also bound specifically to endogenous apelin receptor in stem cell-derived cardiomyocytes. Apelin488 and ELA488 bound specifically to apelin receptor, localising to blood vessels and tubules of the renal cortex. Discussion: Our data indicate that the described novel fluorescent ligands expand the pharmacological toolbox for studying the apelin receptor across multiple platforms to facilitate drug discovery.


Subject(s)
Peptide Hormones , Cricetinae , Animals , Humans , Apelin Receptors/metabolism , Ligands , Peptide Hormones/metabolism , Cricetulus , Protein Binding
4.
Alzheimers Dement (N Y) ; 8(1): e12273, 2022.
Article in English | MEDLINE | ID: mdl-35229025

ABSTRACT

INTRODUCTION: This study examined the safety and pharmacodynamic effects of selective muscarinic M1 receptor orthosteric agonist HTL0018318 in 60 patients with mild-to-moderate Alzheimer's disease (AD) on background donepezil 10 mg/day. METHODS: A randomized, double-blind, placebo-controlled 4-week safety study of HTL0018318 with up-titration and maintenance phases, observing exploratory effects on electrophysiological biomarkers and cognition. RESULTS: Treatment-emergent adverse events (TEAEs) were mild and less frequently reported during maintenance versus titration. Headache was most commonly reported (7-21%); 0 to 13% reported cholinergic TEAEs (abdominal pain, diarrhea, fatigue, nausea) and two patients discontinued due to TEAEs. At 1 to 2 hours post-dose, HTL0018318-related mean maximum elevations in systolic and diastolic blood pressure of 5 to 10 mmHg above placebo were observed during up-titration but not maintenance. Postive effects of HTL0018318 were found on specific attention and memory endpoints. DISCUSSION: HTL0018318 was well tolerated in mild-to-moderate AD patients and showed positive effects on attention and episodic memory on top of therapeutic doses of donepezil.

6.
Cell ; 184(24): 5886-5901.e22, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34822784

ABSTRACT

Current therapies for Alzheimer's disease seek to correct for defective cholinergic transmission by preventing the breakdown of acetylcholine through inhibition of acetylcholinesterase, these however have limited clinical efficacy. An alternative approach is to directly activate cholinergic receptors responsible for learning and memory. The M1-muscarinic acetylcholine (M1) receptor is the target of choice but has been hampered by adverse effects. Here we aimed to design the drug properties needed for a well-tolerated M1-agonist with the potential to alleviate cognitive loss by taking a stepwise translational approach from atomic structure, cell/tissue-based assays, evaluation in preclinical species, clinical safety testing, and finally establishing activity in memory centers in humans. Through this approach, we rationally designed the optimal properties, including selectivity and partial agonism, into HTL9936-a potential candidate for the treatment of memory loss in Alzheimer's disease. More broadly, this demonstrates a strategy for targeting difficult GPCR targets from structure to clinic.


Subject(s)
Alzheimer Disease/drug therapy , Drug Design , Receptor, Muscarinic M1/agonists , Aged , Aged, 80 and over , Aging/pathology , Alzheimer Disease/complications , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Amino Acid Sequence , Animals , Blood Pressure/drug effects , CHO Cells , Cholinesterase Inhibitors/pharmacology , Cricetulus , Crystallization , Disease Models, Animal , Dogs , Donepezil/pharmacology , Electroencephalography , Female , HEK293 Cells , Heart Rate/drug effects , Humans , Male , Mice, Inbred C57BL , Models, Molecular , Molecular Dynamics Simulation , Nerve Degeneration/complications , Nerve Degeneration/pathology , Primates , Rats , Receptor, Muscarinic M1/chemistry , Signal Transduction , Structural Homology, Protein
7.
Nat Commun ; 12(1): 5475, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34531380

ABSTRACT

Acetylcholine release in the hippocampus plays a central role in the formation of new memory representations. An influential but largely untested theory proposes that memory formation requires acetylcholine to enhance responses in CA1 to new sensory information from entorhinal cortex whilst depressing inputs from previously encoded representations in CA3. Here, we show that excitatory inputs from entorhinal cortex and CA3 are depressed equally by synaptic release of acetylcholine in CA1. However, feedforward inhibition from entorhinal cortex exhibits greater depression than CA3 resulting in a selective enhancement of excitatory-inhibitory balance and CA1 activation by entorhinal inputs. Entorhinal and CA3 pathways engage different feedforward interneuron subpopulations and cholinergic modulation of presynaptic function is mediated differentially by muscarinic M3 and M4 receptors, respectively. Thus, our data support a role and mechanisms for acetylcholine to prioritise novel information inputs to CA1 during memory formation.


Subject(s)
Acetylcholine/metabolism , CA1 Region, Hippocampal/physiology , Entorhinal Cortex/physiology , Excitatory Postsynaptic Potentials/physiology , Feedback, Physiological/physiology , Synaptic Transmission/physiology , Animals , CA1 Region, Hippocampal/cytology , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Entorhinal Cortex/cytology , Excitatory Postsynaptic Potentials/drug effects , Feedback, Physiological/drug effects , Interneurons/metabolism , Interneurons/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Synaptic Transmission/drug effects
8.
Peptides ; 145: 170642, 2021 11.
Article in English | MEDLINE | ID: mdl-34455010

ABSTRACT

BACKGROUND: Elabela/Toddler (ELA) is a novel endogenous ligand of the apelin receptor, whose signalling has emerged as a therapeutic target, for example, in cardiovascular disease and cancer. Shorter forms of ELA-32 have been predicted, including ELA-21 and ELA-11, but metabolism and stability of ELA-32 in humans is poorly understood. We, therefore, developed an LC-MS/MS assay to identify ELA-32 metabolites in human plasma and tissues. METHOD: Human kidney homogenates or plasma were incubated at 37 °C with ELA-32 and aliquots withdrawn over 2-4 h into guanidine hydrochloride. Proteins were precipitated and supernatant solid-phase extracted. Peptides were extracted from coronary artery, brain and kidney by immunoprecipitation or solid-phase extraction following acidification. All samples were reduced and alkylated before analysis on an Orbitrap mass spectrometer in high and nano flow mode. RESULTS: The half-life of ELA-32 in plasma and kidney were 47.2 ±â€¯5.7 min and 44.2 ±â€¯3 s, respectively. Using PEAKS Studio and manual data analysis, the most important fragments of ELA-32 with potential biological activity identified were ELA-11, ELA-16, ELA-19 and ELA-20. The corresponding fragments resulting from the loss of C-terminal amino acids were also identified. Endogenous levels of these peptides could not be measured, as ELA peptides are prone to oxidation and poor chromatographic peaks. CONCLUSIONS: The relatively long ELA plasma half-life observed and identification of a potentially more stable fragment, ELA-16, may suggest that ELA could be a better tool compound and novel template for the development of new drugs acting at the apelin receptor.


Subject(s)
Kidney/metabolism , Peptide Hormones/metabolism , Tandem Mass Spectrometry/methods , Blood Chemical Analysis/methods , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Peptide Fragments/analysis , Peptide Fragments/metabolism , Peptide Hormones/blood , Peptide Hormones/isolation & purification , Protein Isoforms/blood , Protein Isoforms/metabolism , Protein Stability
9.
Front Nutr ; 7: 568991, 2020.
Article in English | MEDLINE | ID: mdl-33195366

ABSTRACT

The short chain fatty acids (SCFAs) acetate, butyrate and propionate, are produced by fermentation of non-digestible carbohydrates by the gut microbiota and regulate appetite, adiposity, metabolism, glycemic control, and immunity. SCFAs act at two distinct G protein coupled receptors (GPCRs), FFAR2 and FFAR3 and are expressed in intestinal enteroendocrine cells (EECs), where they mediate anorectic gut hormone release. EECs also express other GPCRs that act as nutrient sensors, thus SCFAs may elicit some of their health-promoting effects by altering GPCR expression in EECs and enhance gut sensitivity to dietary molecules. Here, we identify that exposure of the murine EEC STC-1 cell line or intestinal organoids to physiological concentrations of SCFAs enhances mRNA levels of the umami taste receptors TASR1 and TASR3, without altering levels of the SCFA GPCRs, FFAR2 and FFAR3. Treatment of EECs with propionate or butyrate, but not acetate, increased levels of umami receptor transcripts, while propionate also reduced CCK expression. This was reversed by inhibiting Gαi/o signaling with pertussis toxin, suggesting that SCFAs act through FFAR2/3 to alter gene expression. Surprisingly, neither a FFAR3 nor a FFAR2 selective ligand could increase TASR1/TASR3 mRNA levels. We assessed the functional impact of increased TASR1/TASR3 expression using unique pharmacological properties of the umami taste receptor; namely, the potentiation of signaling by inosine monophosphate. Activation of umami taste receptor induced inositol-1-phosphate and calcium signaling, and butyrate pretreatment significantly enhanced such signaling. Our study reveals that SCFAs may contribute to EEC adaptation and alter EEC sensitivity to bioactive nutrients.

10.
J Med Chem ; 63(14): 7906-7920, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32558564

ABSTRACT

Structure-based drug design enabled the discovery of 8, HTL22562, a calcitonin gene-related peptide (CGRP) receptor antagonist. The structure of 8 complexed with the CGRP receptor was determined at a 1.6 Å resolution. Compound 8 is a highly potent, selective, metabolically stable, and soluble compound suitable for a range of administration routes that have the potential to provide rapid systemic exposures with resultant high levels of receptor coverage (e.g., subcutaneous). The low lipophilicity coupled with a low anticipated clinically efficacious plasma exposure for migraine also suggests a reduced potential for hepatotoxicity. These properties have led to 8 being selected as a clinical candidate for acute treatment of migraine.


Subject(s)
Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Indazoles/pharmacology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Spiro Compounds/pharmacology , Animals , Binding Sites , Calcitonin Gene-Related Peptide Receptor Antagonists/chemical synthesis , Calcitonin Gene-Related Peptide Receptor Antagonists/metabolism , Calcitonin Gene-Related Peptide Receptor Antagonists/toxicity , Dogs , Drug Design , Humans , Indazoles/chemical synthesis , Indazoles/metabolism , Indazoles/toxicity , Macaca fascicularis , Migraine Disorders/drug therapy , Molecular Docking Simulation , Molecular Structure , Rats , Spiro Compounds/chemical synthesis , Spiro Compounds/metabolism , Spiro Compounds/toxicity , Structure-Activity Relationship
11.
Nat Rev Drug Discov ; 19(6): 389-413, 2020 06.
Article in English | MEDLINE | ID: mdl-32494050

ABSTRACT

Dysregulation of peptide-activated pathways causes a range of diseases, fostering the discovery and clinical development of peptide drugs. Many endogenous peptides activate G protein-coupled receptors (GPCRs) - nearly 50 GPCR peptide drugs have been approved to date, most of them for metabolic disease or oncology, and more than 10 potentially first-in-class peptide therapeutics are in the pipeline. The majority of existing peptide therapeutics are agonists, which reflects the currently dominant strategy of modifying the endogenous peptide sequence of ligands for peptide-binding GPCRs. Increasingly, novel strategies are being employed to develop both agonists and antagonists, to both introduce chemical novelty and improve drug-like properties. Pharmacodynamic improvements are evolving to allow biasing ligands to activate specific downstream signalling pathways, in order to optimize efficacy and reduce side effects. In pharmacokinetics, modifications that increase plasma half-life have been revolutionary. Here, we discuss the current status of the peptide drugs targeting GPCRs, with a focus on evolving strategies to improve pharmacokinetic and pharmacodynamic properties.


Subject(s)
Drug Design , Peptides , Receptors, G-Protein-Coupled/metabolism , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Targeted Therapy , Peptide Library , Peptides/pharmacokinetics , Peptides/pharmacology , Peptides/therapeutic use , Protein Binding , Signal Transduction
12.
Drug Discov Today ; 24(12): 2307-2314, 2019 12.
Article in English | MEDLINE | ID: mdl-31499186

ABSTRACT

Cholinergic dysfunction is involved in a range of neurological and psychiatric disorders, including schizophrenia, dementia and Lewy body disease (LBD), leading to widespread use of cholinergic therapies. However, such drugs have focused on increasing the availability of acetylcholine (ACh) generally, with relatively little work done on the muscarinic system and specific muscarinic receptor subtypes. In this review, we provide an overview of the major cholinergic pathways and cholinergic muscarinic receptors in the human brain and evidence for their dysfunction in several neurological and psychiatric disorders. We discuss how the selectivity of cholinergic system dysfunction suggests that targeted cholinergic therapeutics to the muscarinic receptor subtypes will be vital in treating several disorders associated with cognitive dysfunction and behavioural and psychological symptoms.


Subject(s)
Mental Disorders/physiopathology , Nervous System Diseases/physiopathology , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M4/metabolism , Acetylcholine/metabolism , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Drug Development , Humans , Mental Disorders/drug therapy , Molecular Targeted Therapy , Nervous System Diseases/drug therapy
13.
Bioorg Med Chem Lett ; 29(20): 126611, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31447084

ABSTRACT

A series of novel allosteric antagonists of the GLP-1 receptor (GLP-1R), exemplified by HTL26119, are described. SBDD approaches were employed to identify HTL26119, exploiting structural understanding of the allosteric binding site of the closely related Glucagon receptor (GCGR) (Jazayeri et al., 2016) and the homology relationships between GCGR and GLP-1R. The region around residue C3476.36b of the GLP-1R receptor represents a key difference from GCGR and was targeted for selectivity for GLP-1R.


Subject(s)
Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Heterocyclic Compounds/chemistry , Allosteric Regulation/drug effects , Allosteric Site , Amino Acid Sequence , Drug Design , Molecular Docking Simulation , Molecular Structure , Protein Binding , Receptors, Glucagon/antagonists & inhibitors , Signal Transduction , Structure-Activity Relationship
14.
Biochem Pharmacol ; 147: 38-54, 2018 01.
Article in English | MEDLINE | ID: mdl-29102678

ABSTRACT

Recent interest has focused on antibodies that can discriminate between different receptor conformations. Here we have characterised the effect of a monoclonal antibody (mAb3), raised against a purified thermo-stabilised turkey ß1-adrenoceptor (ß1AR-m23 StaR), on ß1-ARs expressed in CHO-K1 or HEK 293 cells. Immunohistochemical and radioligand-binding studies demonstrated that mAb3 was able to bind to ECL2 of the tß1-AR, but not its human homologue. Specific binding of mAb3 to tß1-AR was inhibited by a peptide based on the turkey, but not the human, ECL2 sequence. Studies with [3H]-CGP 12177 demonstrated that mAb3 prevented the binding of orthosteric ligands to a subset (circa 40%) of turkey ß1-receptors expressed in both CHO K1 and HEK 293 cells. MAb3 significantly reduced the maximum specific binding capacity of [3H]-CGP-12177 without influencing its binding affinity. Substitution of ECL2 of tß1-AR with its human equivalent, or mutation of residues D186S, P187D, Q188E prevented the inhibition of [3H]-CGP 12177 binding by mAb3. MAb3 also elicited a negative allosteric effect on agonist-stimulated cAMP responses. The identity of the subset of turkey ß1-adrenoceptors influenced by mAb3 remains to be established but mAb3 should become an important tool to investigate the nature of ß1-AR conformational states and oligomeric complexes.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/metabolism , Adrenergic beta-Agonists/metabolism , Allosteric Regulation/physiology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Propanolamines/metabolism , Protein Binding/physiology , Protein Stability , Protein Structure, Secondary , Receptors, Adrenergic, beta-1/genetics , Turkey
16.
Nature ; 546(7657): 254-258, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28562585

ABSTRACT

Glucagon-like peptide 1 (GLP-1) regulates glucose homeostasis through the control of insulin release from the pancreas. GLP-1 peptide agonists are efficacious drugs for the treatment of diabetes. To gain insight into the molecular mechanism of action of GLP-1 peptides, here we report the crystal structure of the full-length GLP-1 receptor bound to a truncated peptide agonist. The peptide agonist retains an α-helical conformation as it sits deep within the receptor-binding pocket. The arrangement of the transmembrane helices reveals hallmarks of an active conformation similar to that observed in class A receptors. Guided by this structural information, we design peptide agonists with potent in vivo activity in a mouse model of diabetes.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/chemistry , Peptides/chemistry , Peptides/pharmacology , Animals , Binding Sites , Crystallography, X-Ray , Dose-Response Relationship, Drug , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Male , Mice , Models, Molecular , Peptides/metabolism , Protein Conformation , Rats , Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, Glucagon/chemistry
17.
J Med Chem ; 60(7): 3187-3197, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28374589

ABSTRACT

GPR120 agonists have therapeutic potential for the treatment of diabetes, but few selective agonists have been reported. We identified an indazole-6-phenylcyclopropylcarboxylic acid series of GPR120 agonists and conducted SAR studies to optimize GPR120 potency. Furthermore, we identified a (S,S)-cyclopropylcarboxylic acid structural motif which gave selectivity against GPR40. Good oral exposure was obtained with some compounds displaying unexpected high CNS penetration. Increased MDCK efflux was utilized to identify compounds such as 33 with lower CNS penetration, and activity in oral glucose tolerance studies was demonstrated. Differential activity was observed in GPR120 null and wild-type mice indicating that this effect operates through a mechanism involving GPR120 agonism.


Subject(s)
Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Indazoles/chemistry , Indazoles/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Blood Glucose/analysis , Carboxylic Acids/pharmacokinetics , Humans , Indazoles/pharmacokinetics , Mice , Mice, Inbred C57BL , Models, Molecular
18.
Pharmacol Rev ; 68(4): 954-1013, 2016 10.
Article in English | MEDLINE | ID: mdl-27630114

ABSTRACT

The glucagon-like peptide (GLP)-1 receptor (GLP-1R) is a class B G protein-coupled receptor (GPCR) that mediates the action of GLP-1, a peptide hormone secreted from three major tissues in humans, enteroendocrine L cells in the distal intestine, α cells in the pancreas, and the central nervous system, which exerts important actions useful in the management of type 2 diabetes mellitus and obesity, including glucose homeostasis and regulation of gastric motility and food intake. Peptidic analogs of GLP-1 have been successfully developed with enhanced bioavailability and pharmacological activity. Physiologic and biochemical studies with truncated, chimeric, and mutated peptides and GLP-1R variants, together with ligand-bound crystal structures of the extracellular domain and the first three-dimensional structures of the 7-helical transmembrane domain of class B GPCRs, have provided the basis for a two-domain-binding mechanism of GLP-1 with its cognate receptor. Although efforts in discovering therapeutically viable nonpeptidic GLP-1R agonists have been hampered, small-molecule modulators offer complementary chemical tools to peptide analogs to investigate ligand-directed biased cellular signaling of GLP-1R. The integrated pharmacological and structural information of different GLP-1 analogs and homologous receptors give new insights into the molecular determinants of GLP-1R ligand selectivity and functional activity, thereby providing novel opportunities in the design and development of more efficacious agents to treat metabolic disorders.


Subject(s)
Glucagon-Like Peptide 1 , Receptors, G-Protein-Coupled , Animals , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism
19.
Nature ; 533(7602): 274-7, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27111510

ABSTRACT

Glucagon is a 29-amino-acid peptide released from the α-cells of the islet of Langerhans, which has a key role in glucose homeostasis. Glucagon action is transduced by the class B G-protein-coupled glucagon receptor (GCGR), which is located on liver, kidney, intestinal smooth muscle, brain, adipose tissue, heart and pancreas cells, and this receptor has been considered an important drug target in the treatment of diabetes. Administration of recently identified small-molecule GCGR antagonists in patients with type 2 diabetes results in a substantial reduction of fasting and postprandial glucose concentrations. Although an X-ray structure of the transmembrane domain of the GCGR has previously been solved, the ligand (NNC0640) was not resolved. Here we report the 2.5 Å structure of human GCGR in complex with the antagonist MK-0893 (ref. 4), which is found to bind to an allosteric site outside the seven transmembrane (7TM) helical bundle in a position between TM6 and TM7 extending into the lipid bilayer. Mutagenesis of key residues identified in the X-ray structure confirms their role in the binding of MK-0893 to the receptor. The unexpected position of the binding site for MK-0893, which is structurally similar to other GCGR antagonists, suggests that glucagon activation of the receptor is prevented by restriction of the outward helical movement of TM6 required for G-protein coupling. Structural knowledge of class B receptors is limited, with only one other ligand-binding site defined--for the corticotropin-releasing hormone receptor 1 (CRF1R)--which was located deep within the 7TM bundle. We describe a completely novel allosteric binding site for class B receptors, providing an opportunity for structure-based drug design for this receptor class and furthering our understanding of the mechanisms of activation of these receptors.


Subject(s)
Pyrazoles/metabolism , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/chemistry , beta-Alanine/analogs & derivatives , Allosteric Site/drug effects , Crystallography, X-Ray , Glucagon/metabolism , Glucagon/pharmacology , Humans , Ligands , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Models, Molecular , Protein Conformation/drug effects , Pyrazoles/chemistry , Pyrazoles/pharmacology , Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Glucagon/classification , Receptors, Glucagon/metabolism , beta-Alanine/chemistry , beta-Alanine/metabolism , beta-Alanine/pharmacology
20.
Drug Discov Today ; 20(11): 1355-64, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26303408

ABSTRACT

X-ray structures of G protein-coupled receptors (GPCRs) have now been reported for more than 60 ligands and 20 receptors, including examples from GPCR classes A, B, C and F. The new structures show previously unobtainable details of interactions between GPCRs and ligands, including the roles of lipophilic regions and water molecules as key drivers of binding. In addition, the structures have revealed several surprising ligand-binding modes, including sites outside the orthosteric pocket. This new information is dramatically changing the way we approach GPCR drug discovery.


Subject(s)
Drug Design , Drug Discovery/methods , Receptors, G-Protein-Coupled/chemistry , Binding Sites , Humans , Ligands , Protein Binding , Receptors, G-Protein-Coupled/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...