Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 296
Filter
1.
Bioconjug Chem ; 35(7): 981-995, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38865349

ABSTRACT

Peptide fibrillization is crucial in biological processes such as amyloid-related diseases and hormone storage, involving complex transitions between folded, unfolded, and aggregated states. We here employ light to induce reversible transitions between aggregated and nonaggregated states of a peptide, linked to the parathyroid hormone (PTH). The artificial light-switch 3-{[(4-aminomethyl)phenyl]diazenyl}benzoic acid (AMPB) is embedded into a segment of PTH, the peptide PTH25-37, to control aggregation, revealing position-dependent effects. Through in silico design, synthesis, and experimental validation of 11 novel PTH25-37-derived peptides, we predict and confirm the amyloid-forming capabilities of the AMPB-containing peptides. Quantum-chemical studies shed light on the photoswitching mechanism. Solid-state NMR studies suggest that ß-strands are aligned parallel in fibrils of PTH25-37, while in one of the AMPB-containing peptides, ß-strands are antiparallel. Simulations further highlight the significance of π-π interactions in the latter. This multifaceted approach enabled the identification of a peptide that can undergo repeated phototriggered transitions between fibrillated and defibrillated states, as demonstrated by different spectroscopic techniques. With this strategy, we unlock the potential to manipulate PTH to reversibly switch between active and inactive aggregated states, representing the first observation of a photostimulus-responsive hormone.


Subject(s)
Amyloid , Parathyroid Hormone , Parathyroid Hormone/chemistry , Amyloid/chemistry , Humans , Peptides/chemistry , Peptide Fragments/chemistry , Protein Aggregates , Light , Photochemical Processes
2.
ACS Chem Neurosci ; 15(4): 844-853, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38314550

ABSTRACT

Parathyroid hormone (PTH) type 1 receptor (PTH1R), as a typical class B1 G protein-coupled receptor (GPCR), is responsible for regulating bone turnover and maintaining calcium homeostasis, and its dysregulation has been implicated in the development of several diseases. The extracellular domain (ECD) of PTH1R is crucial for the recognition and binding of ligands, and the receptor may exhibit an autoinhibited state with the closure of the ECD in the absence of ligands. However, the correlation between ECD conformations and PTH1R activation remains unclear. Thus, this study combines enhanced sampling molecular dynamics (MD) simulations and Markov state models (MSMs) to reveal the possible relevance between the ECD conformations and the activation of PTH1R. First, 22 intermediate structures are generated from the autoinhibited state to the active state and conducted for 10 independent 200 ns simulations each. Then, the MSM is constructed based on the cumulative 44 µs simulations with six identified microstates. Finally, the potential interplay between ECD conformational changes and PTH1R activation as well as cryptic allosteric pockets in the intermediate states during receptor activation is revealed. Overall, our findings reveal that the activation of PTH1R has a specific correlation with ECD conformational changes and provide essential insights for GPCR biology and developing novel allosteric modulators targeting cryptic sites.


Subject(s)
Molecular Dynamics Simulation , Signal Transduction , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptor, Parathyroid Hormone, Type 1/metabolism , Amino Acid Sequence , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism
3.
Nature ; 626(7998): 435-442, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38109936

ABSTRACT

Many peptide hormones form an α-helix on binding their receptors1-4, and sensitive methods for their detection could contribute to better clinical management of disease5. De novo protein design can now generate binders with high affinity and specificity to structured proteins6,7. However, the design of interactions between proteins and short peptides with helical propensity is an unmet challenge. Here we describe parametric generation and deep learning-based methods for designing proteins to address this challenge. We show that by extending RFdiffusion8 to enable binder design to flexible targets, and to refining input structure models by successive noising and denoising (partial diffusion), picomolar-affinity binders can be generated to helical peptide targets by either refining designs generated with other methods, or completely de novo starting from random noise distributions without any subsequent experimental optimization. The RFdiffusion designs enable the enrichment and subsequent detection of parathyroid hormone and glucagon by mass spectrometry, and the construction of bioluminescence-based protein biosensors. The ability to design binders to conformationally variable targets, and to optimize by partial diffusion both natural and designed proteins, should be broadly useful.


Subject(s)
Computer-Aided Design , Deep Learning , Peptides , Proteins , Biosensing Techniques , Diffusion , Glucagon/chemistry , Glucagon/metabolism , Luminescent Measurements , Mass Spectrometry , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Structure, Secondary , Proteins/chemistry , Proteins/metabolism , Substrate Specificity , Models, Molecular
4.
Structure ; 31(6): 668-676.e5, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37148874

ABSTRACT

The parathyroid hormone (PTH) 1 receptor (PTH1R) is a G protein-coupled receptor (GPCR) that regulates skeletal development and calcium homeostasis. Here, we describe cryo-EM structures of the PTH1R in complex with fragments of the two hormones, PTH and PTH-related protein, the drug abaloparatide, as well as the engineered tool compounds, long-acting PTH (LA-PTH) and the truncated peptide, M-PTH(1-14). We found that the critical N terminus of each agonist engages the transmembrane bundle in a topologically similar fashion, reflecting similarities in measures of Gαs activation. The full-length peptides induce subtly different extracellular domain (ECD) orientations relative to the transmembrane domain. In the structure bound to M-PTH, the ECD is unresolved, demonstrating that the ECD is highly dynamic when unconstrained by a peptide. High resolutions enabled identification of water molecules near peptide and G protein binding sites. Our results illuminate the action of orthosteric agonists of the PTH1R.


Subject(s)
Parathyroid Hormone , Receptor, Parathyroid Hormone, Type 1 , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptor, Parathyroid Hormone, Type 1/metabolism , Parathyroid Hormone/pharmacology , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Peptides/pharmacology , Peptides/metabolism , Receptors, G-Protein-Coupled/metabolism , GTP-Binding Proteins/metabolism
5.
FEBS Lett ; 597(7): 995-1006, 2023 04.
Article in English | MEDLINE | ID: mdl-36700832

ABSTRACT

The parathyroid hormone (PTH) regulates the calcium and phosphate level in blood after secretion from parathyroid chief cells. The pre- and pro-sequences of precursor preproPTH get cleaved during PTH maturation. In secretory granules, PTH forms functional amyloids. Using thioflavin T fibrillation assays, circular dichroism, NMR spectroscopy, and cellular cAMP activation, we show that the pro-sequence prevents premature fibrillation by impairing primary nucleation because of Coulomb repulsion of positively charged residues. Under seeding or high salt conditions or in the presence of heparin at pH 5.5, proPTH fibril formation is delayed, but the monomer release properties are conserved. ProPTH can still activate in cellulo PTH receptor 1 but with impaired potency. These findings give some perspectives on medical applications of PTH in hormone therapy.


Subject(s)
Amyloid , Protein Precursors , Parathyroid Hormone/chemistry , Parathyroid Hormone/physiology , Parathyroid Glands , Calcium
6.
Acta Pharmacol Sin ; 44(6): 1227-1237, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36482086

ABSTRACT

Parathyroid hormone (PTH) and PTH-related peptide (PTHrP) are two endogenous hormones recognized by PTH receptor-1 (PTH1R), a member of class B G protein- coupled receptors (GPCRs). Both PTH and PTHrP analogs including teriparatide and abaloparatide are approved drugs for osteoporosis, but they exhibit distinct pharmacology. Here we report two cryo-EM structures of human PTH1R bound to PTH and PTHrP in the G protein-bound state at resolutions of 2.62 Å and 3.25 Å, respectively. Detailed analysis of these structures uncovers both common and unique features for the agonism of PTH and PTHrP. Molecular dynamics (MD) simulation together with site-directed mutagenesis studies reveal the molecular basis of endogenous hormones recognition specificity and selectivity to PTH1R. These results provide a rational template for the clinical use of PTH and PTHrP analogs as an anabolic therapy for osteoporosis and other disorders.


Subject(s)
Osteoporosis , Parathyroid Hormone-Related Protein , Humans , Parathyroid Hormone-Related Protein/metabolism , Parathyroid Hormone-Related Protein/pharmacology , Amino Acid Sequence , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Receptors, G-Protein-Coupled , Osteoporosis/drug therapy
7.
ACS Chem Biol ; 17(11): 3148-3158, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36282520

ABSTRACT

Polypeptides that activate the parathyroid hormone receptor-1 (PTHR1) are important in human physiology and medicine. Most previous studies of peptide binding to this receptor have involved the displacement of a radiolabeled ligand. We report a new assay format based on bioluminescence resonance energy transfer (BRET). Fusion of a NanoLuc luciferase (nLuc) unit to the N-terminus of the PTHR1 allows the direct detection of binding by an agonist peptide bearing a tetramethylrhodamine (TMR) unit. Affinity measurements from the BRET assay align well with results previously obtained via radioligand displacement. The BRET assay offers substantial operational benefits relative to affinity measurements involving radioactive compounds. The convenience of the new assay allowed us to explore several questions raised by earlier reports. For example, we show that although the first two residues of PTH(1-34) (the drug teriparatide) are critical for PTHR1 activation, these two residues contribute little or nothing to affinity. Comparisons among the well-studied agonists PTH(1-34), PTHrP(1-34), and "long-acting PTH" (LA-PTH) reveal that the high affinity of LA-PTH arises largely from a diminished rate constant for dissociation relative to the other two. A D-peptide recently reported to be comparable to PTH(1-34) as an agonist of the PTHR1 was found not to bind detectably to the receptor and to be a very weak agonist.


Subject(s)
Parathyroid Hormone , Receptor, Parathyroid Hormone, Type 1 , Humans , Receptor, Parathyroid Hormone, Type 1/metabolism , Parathyroid Hormone/chemistry , Luciferases , Thermodynamics , Peptide Fragments/metabolism
8.
J Bone Miner Res ; 37(12): 2586-2601, 2022 12.
Article in English | MEDLINE | ID: mdl-36153665

ABSTRACT

The approach utilized a systematic review of the medical literature executed with specifically designed criteria that focused on the etiologies and pathogenesis of hypoparathyroidism. Enhanced attention by endocrine surgeons to new knowledge about parathyroid gland viability are reviewed along with the role of intraoperative parathyroid hormone (ioPTH) monitoring during and after neck surgery. Nonsurgical etiologies account for a significant proportion of cases of hypoparathyroidism (~25%), and among them, genetic etiologies are key. Given the pervasive nature of PTH deficiency across multiple organ systems, a detailed review of the skeletal, renal, neuromuscular, and ocular complications is provided. The burden of illness on affected patients and their caregivers contributes to reduced quality of life and social costs for this chronic endocrinopathy. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Hypoparathyroidism , Humans , Hypoparathyroidism/etiology , Hypoparathyroidism/physiopathology , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Quality of Life , Parathyroid Glands/pathology , Parathyroid Glands/surgery
9.
Vitam Horm ; 120: 345-370, 2022.
Article in English | MEDLINE | ID: mdl-35953116

ABSTRACT

Primary cilia are subcellular structures specialized in sensing different stimuli in a diversity of cell types. In bone, the primary cilium is involved in mechanical sensing and transduction of signals that regulate the behavior of mesenchymal osteoprogenitors, osteoblasts and osteocytes. To perform its functions, the primary cilium modulates a plethora of molecules including those stimulated by the parathyroid hormone (PTH) receptor type I (PTH1R), a master regulator of osteogenesis. Binding of the agonists PTH or PTH-related protein (PTHrP) to the PTH1R or direct agonist-independent stimulation of the receptor activate PTH1R signaling pathways. In turn, activation of PTH1R leads to regulation of bone formation and remodeling. Herein, we describe the structure, function and molecular partners of primary cilia in the context of bone, playing special attention to those signaling pathways that are mediated directly or indirectly by PTH1R in association with primary cilia during the process of osteogenesis.


Subject(s)
Osteogenesis , Parathyroid Hormone , Cilia/metabolism , Humans , Osteoblasts , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism
10.
Mol Cell ; 82(18): 3468-3483.e5, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35932760

ABSTRACT

Endogenous parathyroid hormone (PTH) and PTH-related peptide (PTHrP) bind to the parathyroid hormone receptor 1 (PTH1R) and activate the stimulatory G-protein (Gs) signaling pathway. Intriguingly, the two ligands have distinct signaling and physiological properties: PTH evokes prolonged Gs activation, whereas PTHrP evokes transient Gs activation with reduced bone-resorption effects. The distinct molecular actions are ascribed to the differences in ligand recognition and dissociation kinetics. Here, we report cryoelectron microscopic structures of six forms of the human PTH1R-Gs complex in the presence of PTH or PTHrP at resolutions of 2.8 -4.1 Å. A comparison of the PTH-bound and PTHrP-bound structures reveals distinct ligand-receptor interactions underlying the ligand affinity and selectivity. Furthermore, five distinct PTH-bound structures, combined with computational analyses, provide insights into the unique and complex process of ligand dissociation from the receptor and shed light on the distinct durations of signaling induced by PTH and PTHrP.


Subject(s)
Parathyroid Hormone-Related Protein , Receptor, Parathyroid Hormone, Type 1 , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Ligands , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Parathyroid Hormone-Related Protein/chemistry , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism
11.
FEBS Lett ; 596(22): 2928-2939, 2022 11.
Article in English | MEDLINE | ID: mdl-35903816

ABSTRACT

In acidic secretory granules of mammalian cells, peptide hormones including the parathyroid hormone are presumably stored in the form of functional amyloid fibrils. Mature PTH, however, is considerably positively charged in acidic environments, a condition known to impede unassisted self-aggregation into fibrils. Here, we studied the role of the polyanion heparin on promoting fibril formation of PTH. Employing ITC, CD spectroscopy, NMR, SAXS, and fluorescence-based assays, we could demonstrate that heparin binds PTH with submicromolar affinity and facilitates its conversion into fibrillar seeds, enabling rapid formation of amyloid fibrils under acidic conditions. In the absence of heparin, PTH remained in a soluble monomeric state. We suspect that heparin-like surfaces are required in vivo to convert PTH efficiently into fibrillar deposits.


Subject(s)
Amyloid , Heparin , Animals , Heparin/metabolism , Amyloid/metabolism , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Scattering, Small Angle , X-Ray Diffraction , Hydrogen-Ion Concentration , Mammals
12.
Biochemistry ; 61(11): 981-991, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35533300

ABSTRACT

Human parathyroid hormone (PTH) is an 84-amino acid peptide that contains two methionine (Met) residues located at positions 8 and 18. It has long been recognized that Met residues in PTH are subject to oxidation to become Met sulfoxide, resulting in a decreased biological function of the peptide. However, the mechanism of the lost biological function of PTH oxidation remains elusive. To characterize whether the shift from the hydrophobic nature of the native Met residue to the hydrophilic nature of Met sulfoxide plays a role in the reduction of biological activity upon PTH oxidation, we conducted in silico and in vitro site-directed mutagenesis of Met-8 and Met-18 to the hydrophilic residue asparagine (Asn) or to the hydrophobic residue leucine (Leu) and compared the behavior of these mutated peptides with that of PTH oxidized at Met-8 and/or Met-18. Our results showed that the biological activity of the Asn-8 and Asn-8/Asn-18 mutants was significantly reduced, similar to Met-8 sulfoxide and Met-8/Met-18 sulfoxide analogues, while the functions of Asn-18, Leu-8, Leu-8/Leu-18 mutants, or Met-18 sulfoxide analogues were similar to wild-type PTH. This is rationalized from molecular modeling and immunoprecipitation assay, demonstrating disruption of hydrophobic interactions between Met-8 and Met-18 of PTH and type-1 PTH receptor (PTHR1) upon mutation or oxidation. Thus, these novel findings support the notion that the loss of biological function of PTH upon oxidation of Met-8 is due, at least in part, to the conversion from a hydrophobic to a hydrophilic residue that disrupts direct hydrophobic interaction between PTH and PTHR1.


Subject(s)
Asparagine , Methionine , Humans , Leucine/genetics , Leucine/chemistry , Asparagine/genetics , Methionine/genetics , Methionine/chemistry , Parathyroid Hormone/genetics , Parathyroid Hormone/chemistry , Peptides/chemistry , Racemethionine , Mutation , Sulfoxides
13.
ACS Chem Neurosci ; 13(11): 1666-1674, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35543321

ABSTRACT

The parathyroid hormone type 1 receptor (PTH1R), a canonical class B GPCR, is regulated by a positive allosteric modulator, extracellular Ca2+. Calcium ions prolong the residence time of PTH on the PTH1R, leading to increased receptor activation and duration of cAMP signaling. But the essential mechanism of the allosteric behavior of PTH1R is not fully understood. Here, extensive molecular dynamics (MD) simulations are performed for the PTH1R-G-protein combinations with and without Ca2+ to describe how calcium ions allosterically engage receptor-G-protein coupling. We find that the binding of Ca2+ stabilizes the conformation of the PTH1R-PTH-spep (the α5 helix of Gs protein) complex, especially the extracellular loop 1 (ECL1). Moreover, the MM-GBSA result indicates that Ca2+ allosterically promotes the interaction between PTH1R and spep, consistent with the observation of steered molecular dynamics (SMD) simulations. We further illuminate the possible allosteric signaling pathway from the stable Ca2+-coupling site to the intracellular G-protein binding site. These results unveil structural determinants for Ca2+ allosterism in the PTH1R-PTH-spep complex and give insights into pluridimensional GPCR signaling regulated by calcium ions.


Subject(s)
Calcium , Parathyroid Hormone , Calcium/metabolism , GTP-Binding Proteins/metabolism , Molecular Dynamics Simulation , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptor, Parathyroid Hormone, Type 1/metabolism
14.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34940833

ABSTRACT

CONTEXT: Parathyroid hormone (PTH) replacement is a promising approach in the management of hypoparathyroidism but long-acting analogues need to be developed. To date, animal models for testing PTH required parathyroidectomy by surgery. We have developed a nonsurgical rodent hypoparathyroid model and tested a delayed-clearance PTH molecule (DC-PTH). OBJECTIVE: The aim of this study was to use cinacalcet to suppress calcium levels in normal rats and to reverse these effects with the administration of PTH or PTH analogues. METHODS: Male Wistar rats were gavaged with either 30 mg/kg cinacalcet-HCl (cinacalcet) or vehicle only. Animals were then dosed with either single or repeated subcutaneous doses of PTH 1-34 or a DC-PTH at 20 nmol/kg. Control animals received vehicle only. Serum samples were analyzed for ionized calcium (iCa), phosphate, PTH, and DC-PTH. A pharmacokinetic-pharmacodynamic (PK-PD) model was built for cinacalcet, PTH 1-34, and DC-PTH using Phoenix64. RESULTS: Cinacalcet reduced iCa levels between 2 and 24 hours, returning to baseline by 72 hours post dose with nadir at 8 hours (analysis of variance P < .001), associated with a fall in rat PTH. For phosphate there was a variable biphasic response. Single-dose PTH abrogated the cinacalcet-induced fall in iCa for up to 2 hours. DC-PTH prevented the fall in iCa from 4 hours post dose and gave a prolonged response, with iCa levels quicker to return to baseline than controls. DC-PTH has a half-life of 11.5 hours, approximately 44 times longer than human PTH 1-34. The PK-PD models defined the reproducible effect of cinacalcet on iCa and that DC-PTH had prolonged biological activity. CONCLUSION: The administration of cinacalcet provides a robust and reproducible nonsurgical animal model of hypoparathyroidism. DC-PTH holds promise for the treatment of hypoparathyroidism in the future.


Subject(s)
Cinacalcet/pharmacology , Hypoparathyroidism/physiopathology , Parathyroid Hormone/blood , Animals , CHO Cells , Calcium/chemistry , Calcium/metabolism , Cricetulus , Disease Models, Animal , Male , Parathyroid Glands/physiopathology , Parathyroid Hormone/chemistry , Parathyroidectomy , Phosphates/chemistry , Rats , Rats, Wistar , Treatment Outcome
15.
J Chem Inf Model ; 62(21): 5110-5119, 2022 Nov 14.
Article in English | MEDLINE | ID: mdl-34464108

ABSTRACT

Parathyroid hormone (PTH) is an endogenous ligand that activates the PTH type 1 receptor (PTH1R) signaling. Ca2+, a common second messenger, acts as an allosteric regulator for prolonging the activation of PTH1R. However, a clear picture of the underlying allosteric mechanism is still missing. Herein, extensive molecular dynamics (MD) simulations are performed for PTH1R-PTH complexes with and without Ca2+ ions, allowing us to delineate the molecular details of calcium-induced allostery. Our results indicate that acidic residues in the extracellular loop 1 (ECL1) (D251, E252, E254, and E258-E260) and PTH (E19 and E22) serve as key determinants for local Ca2+-coupling structures and rigidity of ECL1. Moreover, the binding of Ca2+ induces conformational changes of transmembrane domain 6/7 (TM6/7) that are related to PTH1R activation and strengthens the residue-residue communication within PTH and TMD allosterically. Moreover, our results demonstrate that the presence of Ca2+ ions potentiates the interaction between PTH and PTH1R via steered molecular dynamics (SMD) simulations, while the point mutation in the PTH (PTHR25C) weakens the binding of PTH and PTH1R. These results support that Ca2+ ions might further prolong the residence time of PTH on PTH1R and facilitate the positive allostery of PTH1R. Together, the present work provides new insights into the allosteric regulation mechanism of GPCRs induced by ions and related drug design targeting the PTH1R allosteric pathway.


Subject(s)
Parathyroid Hormone , Receptor, Parathyroid Hormone, Type 1 , Humans , Allosteric Regulation , Calcium/metabolism , Parathyroid Hormone/chemistry , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction
16.
Biochem Biophys Res Commun ; 572: 15-19, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34332324

ABSTRACT

BACKGROUND: Novel human parathyroid hormone (hPTH) peptides of unknown biological activity have recently been identified in the serum of subjects with normal renal function, chronic renal failure, and end-stage renal disease through the application of liquid chromatography-high resolution mass spectrometry. PURPOSE: of experiments: To determine the bioactivity of these peptides, we synthesized hPTH28-84, hPTH38-84, and hPTH45-84 peptides by solid phase peptide synthesis and tested their bioactivity in MC3T3-E1 mouse osteoblasts, either individually or together with the native hormone, hPTH1-84, by assessing the accumulation of 3´,5´-cyclic adenosine monophosphate (cAMP) and the induction of alkaline phosphatase activity. RESULTS: Increasing doses of hPTH1-84 (1-100 nM) increased the accumulation of cAMP and alkaline phosphatase activity in osteoblasts. hPTH28-84, hPTH38-84, and hPTH45-84 in concentrations of 1-100 nM were biologically inert. Surprisingly, 100 nM hPTH38-84 and hPTH45-84 increased the accumulation of cAMP in osteoblasts treated with increasing amounts of hPTH1-84. Human PTH28-84 had no effects on cAMP activity alone or in combination with hPTH1-84. Conversely, 100 nM hPTH38-84, hPTH45-84, and hPTH28-84 blocked the activation of alkaline phosphatase activity by hPTH1-84. CONCLUSIONS: The data show that the short carboxyl-terminal hPTH peptides, hPTH38-84 and hPTH45-84, increase the amount of cellular cAMP generated in cultured osteoblasts in response to treatment with full-length hPTH1-84 when compared to full-length hPTH1-84 alone. Human PTH28-84 had no effect on cAMP activity alone or in combination with hPTH1-84. Human PTH28-84, hPTH38-84 and hPTH45-84 reduced the effects of hPTH1-84 in osteoblasts with respect to the induction of alkaline phosphatase activity compared to hPTH1-84 alone. Short carboxyl peptides of human PTH are biologically inert but when administered together with full-length hPTH1-84 modulate the bioactivity of hPTH1-84 in osteoblasts.


Subject(s)
Osteoblasts/metabolism , Parathyroid Hormone/metabolism , 3T3 Cells , Animals , Cells, Cultured , Mice , Parathyroid Hormone/chemical synthesis , Parathyroid Hormone/chemistry , Signal Transduction
17.
Int J Mol Sci ; 22(3)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499384

ABSTRACT

Na+/H+ exchange factor-1 (NHERF1), a multidomain PDZ scaffolding phosphoprotein, is required for the type II sodium-dependent phosphate cotransporter (NPT2A)-mediated renal phosphate absorption. Both PDZ1 and PDZ2 domains are involved in NPT2A-dependent phosphate uptake. Though harboring identical core-binding motifs, PDZ1 and PDZ2 play entirely different roles in hormone-regulated phosphate transport. PDZ1 is required for the interaction with the C-terminal PDZ-binding sequence of NPT2A (-TRL). Remarkably, phosphocycling at Ser290 distant from PDZ1, the penultimate step for both parathyroid hormone (PTH) and fibroblast growth factor-23 (FGF23) regulation, controls the association between NHERF1 and NPT2A. PDZ2 interacts with the C-terminal PDZ-recognition motif (-TRL) of G Protein-coupled Receptor Kinase 6A (GRK6A), and that promotes phosphorylation of Ser290. The compelling biological puzzle is how PDZ1 and PDZ2 with identical GYGF core-binding motifs specifically recognize distinct binding partners. Binding determinants distinct from the canonical PDZ-ligand interactions and located "outside the box" explain PDZ domain specificity. Phosphorylation of NHERF1 by diverse kinases and associated conformational changes in NHERF1 add more complexity to PDZ-binding diversity.


Subject(s)
Hormones/chemistry , Phosphoproteins/chemistry , Sodium-Hydrogen Exchangers/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIa/chemistry , Amino Acid Motifs , Fibroblast Growth Factor-23 , G-Protein-Coupled Receptor Kinases/chemistry , Humans , Ion Transport , Ligands , Mutation , Parathyroid Hormone/chemistry , Phosphates/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Serine/chemistry
18.
FASEB J ; 34(11): 15269-15281, 2020 11.
Article in English | MEDLINE | ID: mdl-32964520

ABSTRACT

Fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH) are regulators of renal phosphate excretion and vitamin D metabolism. In chronic kidney disease (CKD), circulating FGF23 and PTH concentrations progressively increase as renal function declines. Oxidation of PTH at two methionine residues (positions 8 and 18) causes a loss of function. The impact of n-oxPTH and oxPTH on FGF23 synthesis, however, and how n-oxPTH and oxPTH concentrations are affected by CKD, is yet unknown. The effects of oxidized and non-oxidized PTH 1-34 on Fgf23 gene expression were analyzed in UMR106 osteoblast-like cells. Furthermore, we investigated the relationship between n-oxPTH and oxPTH, respectively, with FGF23 in two independent patients' cohorts (620 children with CKD and 600 kidney transplant recipients). While n-oxPTH stimulated Fgf23 mRNA synthesis in vitro, oxidation of PTH in particular at Met8 led to a markedly weaker stimulation of Fgf23. The effect was even stronger when both Met8 and Met18 were oxidized. In both clinical cohorts, n-oxPTH-but not oxPTH-was significantly associated with FGF23 concentrations, independent of known confounding factors. Moreover, with progressive deterioration of kidney function, intact PTH (iPTH) and oxPTH increased substantially, whereas n-oxPTH increased only moderately. In conclusion, n-oxPTH, but not oxPTH, stimulates Fgf23 gene expression. The increase in PTH with decreasing GFR is mainly due to an increase in oxPTH in more advanced stages of CKD.


Subject(s)
Fibroblast Growth Factors/metabolism , Glomerular Filtration Rate , Osteoblasts/pathology , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Renal Insufficiency, Chronic/pathology , Adolescent , Animals , Child , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Humans , Male , Osteoblasts/metabolism , Oxidation-Reduction , Prospective Studies , Rats , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism
19.
Nat Chem Biol ; 16(10): 1096-1104, 2020 10.
Article in English | MEDLINE | ID: mdl-32632293

ABSTRACT

Peptide ligands of class B G-protein-coupled receptors act via a two-step binding process, but the essential mechanisms that link their extracellular binding to intracellular receptor-arrestin interactions are not fully understood. Using NMR, crosslinking coupled to mass spectrometry, signaling experiments and computational approaches on the parathyroid hormone (PTH) type 1 receptor (PTHR), we show that initial binding of the PTH C-terminal part constrains the conformation of the flexible PTH N-terminal signaling epitope before a second binding event occurs. A 'hot-spot' PTH residue, His9, that inserts into the PTHR transmembrane domain at this second step allosterically engages receptor-arrestin coupling. A conformational change in PTHR intracellular loop 3 permits favorable interactions with ß-arrestin's finger loop. These results unveil structural determinants for PTHR-arrestin complex formation and reveal that the two-step binding mechanism proceeds via cooperative fluctuations between ligand and receptor, which extend to other class B G-protein-coupled receptors.


Subject(s)
Arrestin/metabolism , Parathyroid Hormone/metabolism , Arrestin/chemistry , Calcium Phosphates , Cryoelectron Microscopy , Cyclic AMP , Escherichia coli , HEK293 Cells , Humans , Molecular Dynamics Simulation , Parathyroid Hormone/chemistry , Receptors, G-Protein-Coupled
20.
J Fluoresc ; 30(2): 329-334, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32020436

ABSTRACT

Excessive secretion of PTH leads to disturbance of calcium and phosphorus metabolism in the body, which promotes bone, kidney, digestive system and nervous system diseases. Due to the short half-life of PTH, it becomes a difficult issue for PTH detection in the clinical diagnosis field. We explored a competitive immunofluorescent sensing mode based on FRET of two-color CdTe QDs for ratiometric PTH 1-84 antigen detection. The FRET effect and ratiometric fluorescence between the two-color CdTe QDs motivated accurate quantification of PTH 1-84 antigen concentration from 0.01 ng mL-1 to 0.08 ng mL-1 with a limit of detection of 3 pg mL-1. More importantly, under UV irradiation, samples with different concentrations of PTH 1-84 antigen achieved fluorescence visualization, which provides huge possibility for the practical application of PTH 1-84 antigen point-of-care detection.


Subject(s)
Antigens/analysis , Biosensing Techniques , Fluorescence Resonance Energy Transfer , Parathyroid Hormone/chemistry , Point-of-Care Systems , Antigen-Antibody Reactions , Humans
SELECTION OF CITATIONS
SEARCH DETAIL