Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4687, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824166

ABSTRACT

Ligand-induced activation of G protein-coupled receptors (GPCRs) can initiate signaling through multiple distinct pathways with differing biological and physiological outcomes. There is intense interest in understanding how variation in GPCR ligand structure can be used to promote pathway selective signaling ("biased agonism") with the goal of promoting desirable responses and avoiding deleterious side effects. Here we present an approach in which a conventional peptide ligand for the type 1 parathyroid hormone receptor (PTHR1) is converted from an agonist which induces signaling through all relevant pathways to a compound that is highly selective for a single pathway. This is achieved not through variation in the core structure of the agonist, but rather by linking it to a nanobody tethering agent that binds with high affinity to a separate site on the receptor not involved in signal transduction. The resulting conjugate represents the most biased agonist of PTHR1 reported to date. This approach holds promise for facile generation of pathway selective ligands for other GPCRs.


Subject(s)
Receptor, Parathyroid Hormone, Type 1 , Receptors, G-Protein-Coupled , Signal Transduction , Single-Domain Antibodies , Ligands , Humans , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptor, Parathyroid Hormone, Type 1/agonists , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology , HEK293 Cells , Signal Transduction/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Protein Binding , Animals , Peptides/chemistry , Peptides/pharmacology , Peptides/metabolism
2.
Nature ; 621(7979): 635-641, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37524305

ABSTRACT

Class B G-protein-coupled receptors (GPCRs), including glucagon-like peptide 1 receptor (GLP1R) and parathyroid hormone 1 receptor (PTH1R), are important drug targets1-5. Injectable peptide drugs targeting these receptors have been developed, but orally available small-molecule drugs remain under development6,7. Here we report the high-resolution structure of human PTH1R in complex with the stimulatory G protein (Gs) and a small-molecule agonist, PCO371, which reveals an unexpected binding mode of PCO371 at the cytoplasmic interface of PTH1R with Gs. The PCO371-binding site is totally different from all binding sites previously reported for small molecules or peptide ligands in GPCRs. The residues that make up the PCO371-binding pocket are conserved in class B GPCRs, and a single alteration in PTH2R and two residue alterations in GLP1R convert these receptors to respond to PCO371. Functional assays reveal that PCO371 is a G-protein-biased agonist that is defective in promoting PTH1R-mediated arrestin signalling. Together, these results uncover a distinct binding site for designing small-molecule agonists for PTH1R and possibly other members of the class B GPCRs and define a receptor conformation that is specific only for G-protein activation but not arrestin signalling. These insights should facilitate the design of distinct types of class B GPCR small-molecule agonist for various therapeutic indications.


Subject(s)
Imidazolidines , Receptors, G-Protein-Coupled , Spiro Compounds , Humans , Arrestin/metabolism , Binding Sites , GTP-Binding Protein alpha Subunits, Gs/metabolism , Imidazolidines/pharmacology , Ligands , Peptides/pharmacology , Protein Conformation , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/classification , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/classification , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Spiro Compounds/pharmacology , Drug Design
3.
Endocrinology ; 162(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33242090

ABSTRACT

LA-PTH is a long-acting parathyroid hormone (PTH) peptide analogue in preclinical development for hypoparathyroidism (HP). Like native PTH, LA-PTH contains a methionine at position 8 (Met8) that is predicted to be critical for function. We assessed the impact of Met oxidation on the functional properties of LA-PTH and control PTH ligands. Oxidation of PTH(1-34) resulted in marked (~20-fold) reductions in binding affinity on the PTH receptor-1 (PTHR1) in cell membranes, similarly diminished potency for 3',5'-cyclic AMP signaling in osteoblastic cell lines (SaOS-2 and UMR106), and impaired efficacy for raising blood calcium in mice. Surprisingly, oxidation of LA-PTH resulted in little or no change in these functional responses. The signaling potency of oxidized-LA-PTH was, however, reduced approximately 40-fold compared to LA-PTH in cells expressing a PTHR1 construct that lacks the N-terminal extracellular domain (ECD). Molecular modeling revealed that while Met8 of both LA-PTH and PTH(1-34) is situated within the orthosteric ligand-binding pocket of the receptor's transmembrane domain bundle (TMD), the Met8 sidechain position is shifted for the 2 ligands so that on Met8 oxidation of PTH(1-34), steric clashes occur that are not seen with oxidized LA-PTH. The findings suggest that LA-PTH and PTH(1-34) engage the receptor differently in the Met8-interaction environment of the TMD bundle, and that this interaction environment can be allosterically influenced by the ECD component of the ligand-receptor complex. The findings should be useful for the future development of novel PTH-based peptide therapeutics for diseases of bone and mineral ion metabolism.


Subject(s)
Hypoparathyroidism/drug therapy , Parathyroid Hormone/analogs & derivatives , Receptor, Parathyroid Hormone, Type 1/agonists , Animals , Calcium/blood , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , HEK293 Cells , Humans , Methionine/metabolism , Mice , Models, Molecular , Norleucine , Oxidation-Reduction , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Rats , Receptor, Parathyroid Hormone, Type 1/metabolism
4.
J Med Chem ; 63(10): 5089-5099, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32022560

ABSTRACT

We have previously shown that the oral administration of the small molecule hPTHR1 agonist PCO371 and its lead compound, 1 (CH5447240) results in PTH-like calcemic and hypophostemic activity in thyroparathyroidectomized rats. However, 1 was converted to a reactive metabolite in a human liver microsome assay. In this article, we report on the modification path that led to an enhancement of PTHR1 agonistic activity and reduction in the formation of a reactive metabolite to result in a potent, selective, and orally active PTHR1 agonist 1-(3,5-dimethyl-4-(2-((4-oxo-2-(4-(trifluoromethoxy)phenyl)-1,3,8-triazaspiro[4.5]dec-1-en-8-yl)sulfonyl)ethyl)phenyl)-5,5-dimethylimidazolidine-2,4-dione (PCO371, 16c). This compound is currently being evaluated in a phase 1 clinical study for the treatment of hypoparathyroidism.


Subject(s)
Imidazolidines/administration & dosage , Imidazolidines/metabolism , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/metabolism , Spiro Compounds/administration & dosage , Spiro Compounds/metabolism , Administration, Oral , Animals , Female , Humans , Hypoparathyroidism/drug therapy , Hypoparathyroidism/metabolism , Imidazolidines/chemistry , LLC-PK1 Cells , Rats , Rats, Sprague-Dawley , Spiro Compounds/chemistry , Swine
5.
J Am Chem Soc ; 141(37): 14486-14490, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31496241

ABSTRACT

The type-1 parathyroid hormone receptor (PTHR1), which regulates calcium homeostasis and tissue development, has two native agonists, parathyroid hormone (PTH) and PTH-related protein (PTHrP). PTH forms a complex with the PTHR1 that is rapidly internalized and induces prolonged cAMP production from endosomes. In contrast, PTHrP induces only transient cAMP production, which primarily arises from receptors on the cell surface. We show that backbone modification of PTH(1-34)-NH2 and abaloparatide (a PTHrP derivative) with a single homologous ß-amino acid residue can generate biased agonists that induce prolonged cAMP production from receptors at the cell surface. This unique spatiotemporal profile could be useful for distinguishing effects associated with the duration of cAMP production from effects associated with the site of cAMP production.


Subject(s)
Receptor, Parathyroid Hormone, Type 1/chemistry , Amino Acid Sequence , Cyclic AMP/biosynthesis , HEK293 Cells , Humans , Parathyroid Hormone-Related Protein/pharmacology , Receptor, Parathyroid Hormone, Type 1/agonists , Sequence Homology, Amino Acid , Signal Transduction
6.
Future Med Chem ; 11(8): 817-831, 2019 04.
Article in English | MEDLINE | ID: mdl-30998079

ABSTRACT

Aim: Parathyroid hormone-1 receptor (PTH1R) is a member of B G protein-coupled receptors. The agonistic activation of the PTH1R results in the production and secretion of osteoclast-stimulating cytokines while antagonists may be used to treat bone metastases, hypercalcemia, cachexia and hyperparathyroidism. Results: We built pharmacophore models and investigated the characteristics of PTH1R agonists and antagonists. The agonist model consists of three hydrophobic points, one hydrogen bond acceptor and one positive ionizable point. The antagonist model consists of one hydrogen bond donor and three hydrophobic points. Conclusion: The features of the two models are similar, but the hydrogen bond acceptor, which is the main difference between PTH1R agonists and antagonists, suggests it may be essential for the agonist.


Subject(s)
Drug Design , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/antagonists & inhibitors , Amino Acid Sequence , Animals , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptor, Parathyroid Hormone, Type 1/metabolism
7.
Science ; 364(6436): 148-153, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30975883

ABSTRACT

The parathyroid hormone receptor-1 (PTH1R) is a class B G protein-coupled receptor central to calcium homeostasis and a therapeutic target for osteoporosis and hypoparathyroidism. Here we report the cryo-electron microscopy structure of human PTH1R bound to a long-acting PTH analog and the stimulatory G protein. The bound peptide adopts an extended helix with its amino terminus inserted deeply into the receptor transmembrane domain (TMD), which leads to partial unwinding of the carboxyl terminus of transmembrane helix 6 and induces a sharp kink at the middle of this helix to allow the receptor to couple with G protein. In contrast to a single TMD structure state, the extracellular domain adopts multiple conformations. These results provide insights into the structural basis and dynamics of PTH binding and receptor activation.


Subject(s)
Parathyroid Hormone/chemistry , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/chemistry , Amino Acid Motifs , Cryoelectron Microscopy , Humans , Parathyroid Hormone/pharmacology , Parathyroid Hormone/physiology , Protein Binding , Protein Domains , Receptor, Parathyroid Hormone, Type 1/ultrastructure
8.
J Orthop Res ; 37(4): 812-820, 2019 04.
Article in English | MEDLINE | ID: mdl-30790359

ABSTRACT

Fractures typically heal via endochondral and intramembranous bone formation, which together form a callus that achieves union and biomechanical recovery. PTHrP, a PTH receptor agonist, plays an important physiological role in fracture healing as an endogenous stimulator of endochondral and intramembranous bone formation. Abaloparatide, a novel systemically-administered osteoanabolic PTH receptor agonist that reduces fracture risk in women with postmenopausal osteoporosis, has 76% homology to PTHrP, suggesting it may have potential to improve fracture healing. To test this hypothesis, ninety-six 12-week-old male rats underwent unilateral internally-stabilized closed mid-diaphyseal femoral fractures and were treated starting the next day with daily s.c. saline (Vehicle) or abaloparatide at 5 or 20 µg/kg/d for 4 or 6 weeks (16 rats/group/time point). Histomorphometry and histology analyses indicated that fracture calluses from the abaloparatide groups exhibited significantly greater total area, higher fluorescence scores indicating more newly-formed bone, and higher fracture bridging scores versus Vehicle controls. Callus bridging score best correlated with callus cartilage score (r = 0.64) and fluorescence score (r = 0.67) at week 4, and callus area correlated with cartilage score (r = 0.60) and fluorescence score (r = 0.89) at Week 6. By micro-CT, calluses from one or both abaloparatide groups had greater bone volume, bone volume fraction, bone mineral content, bone mineral density, and cross-sectional area at both time points versus Vehicle controls. Destructive bending tests indicated greater callus maximum load and stiffness in one or both abaloparatide groups at both time points versus Vehicle controls. These results provide preliminary preclinical evidence for improved fracture healing with systemically-administered abaloparatide. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.


Subject(s)
Femoral Fractures/drug therapy , Fracture Healing/drug effects , Parathyroid Hormone-Related Protein/therapeutic use , Receptor, Parathyroid Hormone, Type 1/agonists , Animals , Bony Callus/diagnostic imaging , Bony Callus/drug effects , Drug Evaluation, Preclinical , Male , Parathyroid Hormone-Related Protein/pharmacology , Rats, Sprague-Dawley , X-Ray Microtomography
9.
Proc Natl Acad Sci U S A ; 115(49): 12383-12388, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30442659

ABSTRACT

Human parathyroid hormone (PTH) and N-terminal fragments thereof activate two receptors, hPTHR1 and hPTHR2, which share ∼51% sequence similarity. A peptide comprising the first 34 residues of PTH is fully active at both receptors and is used to treat osteoporosis. We have used this system to explore the hypothesis that backbone modification of a promiscuous peptidic agonist can provide novel receptor-selective agonists. We tested this hypothesis by preparing a set of variants of PTH(1-34)-NH2 that contained a single ß-amino-acid residue replacement at each of the first eight positions. These homologs, each containing one additional backbone methylene unit relative to PTH(1-34)-NH2 itself, displayed a wide range of potencies in cell-based assays for PTHR1 or PTHR2 activation. The ß-scan series allowed us to identify two homologs, each containing two αâ†’ß replacements, that were highly selective, one for PTHR1 and the other for PTHR2. These findings suggest that backbone modification of peptides may provide a general strategy for achieving activation selectivity among polypeptide-modulated receptors, and that success requires consideration of both ß2- and ß3-residues, which differ in terms of side-chain location.


Subject(s)
Parathyroid Hormone/chemistry , Peptides/chemical synthesis , Peptides/pharmacology , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 2/agonists , Amino Acid Sequence , HEK293 Cells , Humans , Protein Binding , Protein Conformation
10.
J Med Chem ; 61(14): 5949-5962, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29932656

ABSTRACT

During the course of derivatization of HTS hit 4a, we have identified a novel small-molecule hPTHR1 agonist, 1-(3,5-dimethyl-4-(2-((2-((1 R,4 R)-4-methylcyclohexyl)-4-oxo-1,3,8-triazaspiro[4.5]dec-1-en-8-yl)sulfonyl)ethyl)phenyl)-1-methylurea (CH5447240, 14l). Compound 14l exhibited a potent in vitro hPTHR1 agonist effect with EC20 of 3.0 µM and EC50 of 12 µM and showed excellent physicochemical properties, such as high solubility in fasted state simulated intestinal fluid and good metabolic stability in human liver microsomes. Importantly, 14l showed 55% oral bioavailability and a significantly elevated serum calcium level in hypocalcemic model rats.


Subject(s)
Drug Discovery , Hypoparathyroidism/drug therapy , Methylurea Compounds/therapeutic use , Receptor, Parathyroid Hormone, Type 1/agonists , Small Molecule Libraries/pharmacology , Small Molecule Libraries/pharmacokinetics , Sulfones/therapeutic use , Urea/pharmacology , Urea/pharmacokinetics , Administration, Oral , Biological Availability , Cell Line , Humans , Methylurea Compounds/administration & dosage , Methylurea Compounds/pharmacology , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/therapeutic use , Sulfones/administration & dosage , Sulfones/pharmacology , Urea/administration & dosage , Urea/therapeutic use
11.
Osteoporos Int ; 29(3): 685-697, 2018 03.
Article in English | MEDLINE | ID: mdl-29260289

ABSTRACT

Abaloparatide, a novel PTH1 receptor agonist, increased bone formation in osteopenic ovariectomized cynomolgus monkeys while increasing cortical and trabecular bone mass. Abaloparatide increased bone strength and maintained or enhanced bone mass-strength relationships, indicating preserved or improved bone quality. INTRODUCTION: Abaloparatide is a selective PTH1R activator that is approved for the treatment of postmenopausal osteoporosis. The effects of 16 months of abaloparatide administration on bone formation, resorption, density, and strength were assessed in adult ovariectomized (OVX) cynomolgus monkeys (cynos). METHODS: Sixty-five 9-18-year-old female cynos underwent OVX surgery, and 15 similar cynos underwent sham surgery. After a 9-month period without treatments, OVX cynos were allocated to four groups that received 16 months of daily s.c. injections with either vehicle (n = 17) or abaloparatide (0.2, 1, or 5 µg/kg/day; n = 16/dose level), while Sham controls received s.c. vehicle (n = 15). Bone densitometry (DXA, pQCT, micro-CT), qualitative bone histology, serum calcium, bone turnover markers, bone histomorphometry, and bone strength were among the key measures assessed. RESULTS: At the end of the 9-month post-surgical bone depletion period, just prior to the treatment phase, the OVX groups exhibited increased bone turnover markers and decreased bone mass compared with sham controls. Abaloparatide administration to OVX cynos led to increased bone formation parameters, including serum P1NP and endocortical bone formation rate. Abaloparatide administration did not influence serum calcium levels, bone resorption markers, cortical porosity, or eroded surfaces. Abaloparatide increased bone mass at the whole body, lumbar spine, tibial diaphysis, femoral neck, and femoral trochanter. Abaloparatide administration was associated with greater lumbar vertebral strength, and had no adverse effects on bone mass-strength relationships for the vertebrae, femoral neck, femoral diaphysis, or humeral cortical beams. CONCLUSIONS: Abaloparatide administration was associated with increases in bone formation, bone mass and bone strength, and with maintenance of bone quality in OVX cynos, without increases in serum calcium or bone resorption parameters.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/prevention & control , Osteogenesis/drug effects , Parathyroid Hormone-Related Protein/therapeutic use , Absorptiometry, Photon/methods , Animals , Biomarkers/blood , Bone Density/drug effects , Bone Density/physiology , Bone Density Conservation Agents/pharmacology , Bone Resorption/physiopathology , Female , Lumbar Vertebrae/physiopathology , Macaca fascicularis , Osteogenesis/physiology , Ovariectomy , Parathyroid Hormone-Related Protein/pharmacology , Peptide Fragments/blood , Procollagen/blood , Receptor, Parathyroid Hormone, Type 1/agonists , X-Ray Microtomography/methods
12.
J Med Chem ; 60(21): 8816-8833, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29064243

ABSTRACT

The parathyroid hormone receptor 1 (PTHR1) is a member of the B-family of GPCRs; these receptors are activated by long polypeptide hormones and constitute targets of drug development efforts. Parathyroid hormone (PTH, 84 residues) and PTH-related protein (PTHrP, 141 residues) are natural agonists of PTHR1, and an N-terminal fragment of PTH, PTH(1-34), is used clinically to treat osteoporosis. Conventional peptides in the 20-40-mer length range are rapidly degraded by proteases, which may limit their biomedical utility. We have used the PTHR1-ligand system to explore the impact of broadly distributed replacement of α-amino acid residues with ß-amino acid residues on susceptibility to proteolysis and agonist activity. This effort led us to identify new PTHR1 agonists that contain α → ß replacements throughout their sequences, manifest potent agonist activity in cellular assays, and display remarkable resistance to proteolysis, in cases remaining active after extended exposure to simulated gastric fluid. The strategy we have employed suggests a path toward identifying protease-resistant agonists of other B-family GPCRs.


Subject(s)
Proteolysis/drug effects , Receptor, Parathyroid Hormone, Type 1/agonists , Amino Acids/chemistry , Amino Acids/pharmacology , Humans , Ligands , Parathyroid Hormone/pharmacology , Parathyroid Hormone-Related Protein/pharmacology , Protease Inhibitors/chemical synthesis
13.
J Biol Chem ; 292(24): 9865-9881, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28356352

ABSTRACT

The glucagon receptor (GCGR) belongs to the secretin-like (class B) family of G protein-coupled receptors (GPCRs) and is activated by the peptide hormone glucagon. The structures of an activated class B GPCR have remained unsolved, preventing a mechanistic understanding of how these receptors are activated. Using a combination of structural modeling and mutagenesis studies, we present here two modes of ligand-independent activation of GCGR. First, we identified a GCGR-specific hydrophobic lock comprising Met-338 and Phe-345 within the IC3 loop and transmembrane helix 6 (TM6) and found that this lock stabilizes the TM6 helix in the inactive conformation. Disruption of this hydrophobic lock led to constitutive G protein and arrestin signaling. Second, we discovered a polar core comprising conserved residues in TM2, TM3, TM6, and TM7, and mutations that disrupt this polar core led to constitutive GCGR activity. On the basis of these results, we propose a mechanistic model of GCGR activation in which TM6 is held in an inactive conformation by the conserved polar core and the hydrophobic lock. Mutations that disrupt these inhibitory elements allow TM6 to swing outward to adopt an active TM6 conformation similar to that of the canonical ß2-adrenergic receptor complexed with G protein and to that of rhodopsin complexed with arrestin. Importantly, mutations in the corresponding polar core of several other members of class B GPCRs, including PTH1R, PAC1R, VIP1R, and CRFR1, also induce constitutive G protein signaling, suggesting that the rearrangement of the polar core is a conserved mechanism for class B GPCR activation.


Subject(s)
Models, Molecular , Receptor, Parathyroid Hormone, Type 1/agonists , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Glucagon/agonists , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/agonists , Receptors, Vasoactive Intestinal Polypeptide, Type I/agonists , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Cell Line , Conserved Sequence , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Stability , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Glucagon/chemistry , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/chemistry , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Second Messenger Systems , Structural Homology, Protein
14.
Nat Commun ; 7: 13384, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27857062

ABSTRACT

Parathyroid hormone (PTH) is essential for calcium homeostasis and its action is mediated by the PTH type 1 receptor (PTHR1), a class B G-protein-coupled receptor. Hypoparathyroidism and osteoporosis can be treated with PTH injections; however, no orally effective PTH analogue is available. Here we show that PCO371 is a novel, orally active small molecule that acts as a full agonist of PTHR1. PCO371 does not affect the PTH type 2 receptor (PTHR2), and analysis using PTHR1-PTHR2 chimeric receptors indicated that Proline 415 of PTHR1 is critical for PCO371-mediated PTHR1 activation. Oral administration of PCO371 to osteopenic rats provokes a significant increase in bone turnover with limited increase in bone mass. In hypocalcemic rats, PCO371 restores serum calcium levels without increasing urinary calcium, and with stronger and longer-lasting effects than PTH injections. These results strongly suggest that PCO371 can provide a new treatment option for PTH-related disorders, including hypoparathyroidism.


Subject(s)
Hypoparathyroidism/drug therapy , Imidazolidines/chemical synthesis , Receptor, Parathyroid Hormone, Type 1/agonists , Spiro Compounds/chemical synthesis , Animals , Dogs , Female , Gene Expression Regulation/drug effects , Humans , Imidazolidines/pharmacology , Male , Molecular Structure , Mutation , Parathyroid Glands/drug effects , Parathyroid Glands/surgery , Rats , Spiro Compounds/pharmacology
15.
Methods ; 92: 51-63, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-25986936

ABSTRACT

Ligands possessing different physico-chemical structures productively interact with G protein-coupled receptors generating distinct downstream signaling events due to their abilities to activate/select idiosyncratic receptor entities ('receptorsomes') from the full spectrum of potential receptor partners. We have employed multiple novel informatic approaches to identify and characterize the in vivo transcriptomic signature of an arrestin-signaling biased ligand, [D-Trp(12),Tyr(34)]-bPTH(7-34), acting at the parathyroid hormone type 1 receptor (PTH1R), across six different murine tissues after chronic drug exposure. We are able to demonstrate that [D-Trp(12),Tyr(34)]-bPTH(7-34) elicits a distinctive arrestin-signaling focused transcriptomic response that is more coherently regulated, in an arrestin signaling-dependent manner, across more tissues than that of the pluripotent endogenous PTH1R ligand, hPTH(1-34). This arrestin-focused response signature is strongly linked with the transcriptional regulation of cell growth and development. Our informatic deconvolution of a conserved arrestin-dependent transcriptomic signature from wild type mice demonstrates a conceptual framework within which the in vivo outcomes of biased receptor signaling may be further investigated or predicted.


Subject(s)
Gene Regulatory Networks/physiology , Informatics/methods , Parathyroid Hormone/pharmacology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Animals , Cattle , Gene Regulatory Networks/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/physiology , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects
16.
Mol Pharmacol ; 87(4): 706-17, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637603

ABSTRACT

Biased G protein-coupled receptor agonists engender a restricted repertoire of downstream events from their cognate receptors, permitting them to produce mixed agonist-antagonist effects in vivo. While this opens the possibility of novel therapeutics, it complicates rational drug design, since the in vivo response to a biased agonist cannot be reliably predicted from its in cellula efficacy. We have employed novel informatic approaches to characterize the in vivo transcriptomic signature of the arrestin pathway-selective parathyroid hormone analog [d-Trp(12), Tyr(34)]bovine PTH(7-34) in six different murine tissues after chronic drug exposure. We find that [d-Trp(12), Tyr(34)]bovine PTH(7-34) elicits a distinctive arrestin-signaling focused transcriptomic response that is more coherently regulated across tissues than that of the pluripotent agonist, human PTH(1-34). This arrestin-focused network is closely associated with transcriptional control of cell growth and development. Our demonstration of a conserved arrestin-dependent transcriptomic signature suggests a framework within which the in vivo outcomes of arrestin-biased signaling may be generalized.


Subject(s)
Arrestins/metabolism , Parathyroid Hormone/pharmacology , Peptide Fragments/pharmacology , Transcriptome , Animals , Arrestins/genetics , Cattle , Computational Biology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction , Species Specificity , Transcription, Genetic
17.
J Pept Sci ; 19(6): 337-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23564659

ABSTRACT

With an increasing number of new chemical entities entering clinical studies, and an increasing share of the market, peptides and peptidomimetics constitute one of the most promising classes of therapeutics. The success of synthetic peptides as therapeutics relies on the lead optimization step in which the lead candidates are modified to improve drug-like properties of peptides related to potency, pharmacokinetics, solubility, and stability, among others. Peptidomimetics based on the N-terminal stretch of the first 11 amino acids of the PTH have been investigated as potential lead compounds for the treatment of osteoporosis. On the basis of a peptide reported in the literature, referred to here as the Parent Peptide (H-Aib-Val-Aib-Glu-Ile-Gln-Leu-Nle-His-Gln-Har-NH2), we conducted systematic SAR analyses to investigate the effects of altering peptide hydrophobicity on PTH receptor functional potency as measured by the cAMP (cyclic adenosine monophosphate) accumulation and ß-arrestin recruitment assays. Among hydrophobic residues, we found that the Val2 position shows the least flexibility in terms of the SAR studies, whereas the Leu7 position appeared to be most flexible. Through circular dichroism and nuclear magnetic resonance spectroscopy studies, we were able to establish that changes in hydrophobic residues significantly change the extent of peptide helicity and that the helical character correlates well with receptor agonist activity. Here, we report several novel PTH 1-11 peptidomimetics that show comparable or enhanced potency to stimulate Gs-signaling over ß-arrestin recruitment as compared with such properties of PTH 1-34 and the Parent Peptide.


Subject(s)
Molecular Probes/pharmacology , Oligopeptides/pharmacology , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptor, Parathyroid Hormone, Type 1/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Molecular , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Receptor, Parathyroid Hormone, Type 1/agonists , Structure-Activity Relationship
18.
Cell Signal ; 25(2): 527-38, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23159578

ABSTRACT

Parathyroid hormone (PTH) is an anabolic agent that mediates bone formation through activation of the Gα(s)-, Gα(q)- and ß-arrestin-coupled parathyroid hormone receptor type 1 (PTH1R). Pharmacological evidence based on the effect of PTH(7-34), a PTH derivative that is said to preferentially activate ß-arrestin signaling through PTH1R, suggests that PTH1R-activated ß-arrestin signaling mediates anabolic effects on bone. Here, we performed a thorough evaluation of PTH(7-34) signaling behaviour using quantitative assays for ß-arrestin recruitment, Gα(s)- and Gα(q)-signaling. We found that PTH(7-34) inhibited PTH-induced cAMP accumulation, but was unable to induce ß-arrestin recruitment, PTH1R internalization and ERK1/2 phosphorylation in HEK293, CHO and U2OS cells. Thus, the ß-arrestin bias of PTH(7-34) is not apparent in every cell type examined, suggesting that correlating in vivo effects of PTH(7-34) to in vitro pharmacology should be done with caution.


Subject(s)
Arrestins/metabolism , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction/drug effects , Animals , CHO Cells , Calcium/metabolism , Cell Line , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Parathyroid Hormone/analogs & derivatives , Phosphorylation , Receptor, Parathyroid Hormone, Type 1/agonists , beta-Arrestins
19.
ACS Chem Biol ; 8(3): 617-25, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23237450

ABSTRACT

GPCRs mediate intracellular signaling upon external stimuli, making them ideal drug targets. However, little is known about their activation mechanisms due to the difficulty in purification. Here, we introduce a method to purify GPCRs in nanodiscs, which incorporates GPCRs into lipid bilayers immediately after membrane solubilization, followed by single-step purification. Using this approach, we purified a family B GPCR, parathyroid hormone 1 receptor (PTH1R), which regulates calcium and phosphate homeostasis and is a drug target for osteoporosis. We demonstrated that the purified PTH1R in nanodiscs can bind to PTH(1-34) and activate G protein. We also observed that Ca(2+) is a weak agonist of PTH1R, and Ca(2+) in millimolar concentration can switch PTH(1-34) from an inverse agonist to an agonist. Hence, our results show that nanodiscs are a viable vehicle for GPCR purification, enabling studies of GPCRs under precise experimental conditions without interference from other cellular or membrane components.


Subject(s)
Calcium/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Nanostructures/chemistry , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction , Binding Sites , Calcium/pharmacology , HEK293 Cells , Humans , Ligands , Models, Molecular , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/isolation & purification , Structure-Activity Relationship
20.
Mini Rev Med Chem ; 12(9): 856-65, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22681253

ABSTRACT

'Biased agonism' refers to the ability of a ligand to selectively recruit different intracellular signaling proteins to elicit distinct phenotypic effects in cells. While conventional G protein-coupled receptor (GPCR) agonism and antagonism can be regarded as modulating the quantity of efficacy, functionally selective or 'biased' ligands qualitatively change the trafficking of information flowing across the plasma membrane. The concept of ligand directed signaling fundamentally raises the potential of pharmacologic agents with novel therapeutic profiles possessing improved therapeutic efficacy or reduced side effects. Currently, there is little experimental evidence that biased ligands offer advantages over conventional agonists/antagonists in vivo. Recent work examining biased agonism at the type I parathyroid hormone receptor (PTH1R) demonstrates that selective activation of G protein-independent arrestin-mediated signaling pathways elicits a physiologic response in bone distinct from that induced by the conventional PTH1R agonist PTH(1-34). While intermittent (daily) administration of PTH(1-34) (teriparitide) is effective in increasing bone formation, PTH(1-34) administration is also associated with increases in bone resorption and a propensity to promote hypercalcemia/hypercalcuria. In contrast, D-Trp12,Tyr34-bPTH(7-34) (PTH-ßarr), an arrestin pathway-selective agonist for the PTH1R, induces anabolic bone formation independent of classic G protein-coupled signaling mechanisms. Unlike PTH(1-34), PTH-ßarr appears to 'uncouple' the anabolic effects of PTH1R activation from its catabolic and calcitropic effects. Such findings offer evidence that arrestin pathway-selective GPCR agonists can elicit potentially beneficial effects in vivo that cannot be achieved using conventional agonist or antagonist ligands.


Subject(s)
Arrestins/agonists , Bone and Bones/drug effects , Osteogenesis/drug effects , Parathyroid Hormone-Related Protein/administration & dosage , Receptor, Parathyroid Hormone, Type 1/agonists , Animals , Arrestins/metabolism , Bone Resorption/prevention & control , Bone and Bones/physiology , Humans , Hypercalcemia/chemically induced , Hypercalcemia/prevention & control , Hypercalciuria/chemically induced , Hypercalciuria/prevention & control , Ligands , Mice , Osteogenesis/physiology , Parathyroid Hormone/administration & dosage , Parathyroid Hormone/metabolism , Parathyroid Hormone-Related Protein/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...