1
|
Liu S, Daley EJ, My-Linh Tran L, Yu Z, Reyes M, Dean T, Khatri A, Levine PM, Balana AT, Pratt MR, Jüppner H, Gellman SH, Gardella TJ. Backbone Modification Provides a Long-Acting Inverse Agonist of Pathogenic, Constitutively Active PTH1R Variants. J Am Chem Soc 2024; 146:6522-6529. [PMID: 38417010 PMCID: PMC11162201 DOI: 10.1021/jacs.3c09694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
Parathyroid hormone 1 receptor (PTH1R) plays a key role in mediating calcium homeostasis and bone development, and aberrant PTH1R activity underlies several human diseases. Peptidic PTH1R antagonists and inverse agonists have therapeutic potential in treating these diseases, but their poor pharmacokinetics and pharmacodynamics undermine their in vivo efficacy. Herein, we report the use of a backbone-modification strategy to design a peptidic PTH1R inhibitor that displays prolonged activity as an antagonist of wild-type PTH1R and an inverse agonist of the constitutively active PTH1R-H223R mutant both in vitro and in vivo. This peptide may be of interest for the future development of therapeutic agents that ameliorate PTH1R malfunction.
Collapse
Affiliation(s)
- Shi Liu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Eileen J Daley
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Lauren My-Linh Tran
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Zhen Yu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Monica Reyes
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Thomas Dean
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Ashok Khatri
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Paul M Levine
- Departments of Chemistry and Biological Sciences, University of Southern California, Los Angeles, California 90089, USA
| | - Aaron T Balana
- Departments of Chemistry and Biological Sciences, University of Southern California, Los Angeles, California 90089, USA
| | - Matthew R Pratt
- Departments of Chemistry and Biological Sciences, University of Southern California, Los Angeles, California 90089, USA
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Samuel H. Gellman
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
2
|
Peña KA, Savransky S, Lewis B. Endosomal signaling via cAMP in parathyroid hormone (PTH) type 1 receptor biology. Mol Cell Endocrinol 2024; 581:112107. [PMID: 37981188 DOI: 10.1016/j.mce.2023.112107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023]
Abstract
Compartmentalization of GPCR signaling is an emerging topic that highlights the physiological relevance of spatial bias in signaling. The parathyroid hormone (PTH) type 1 receptor (PTH1R) was the first GPCR described to signal via heterotrimeric G-protein and cAMP from endosomes after β-arrestin mediated internalization, challenging the canonical GPCR signaling model which established that signaling is terminated by receptor internalization. More than a decade later, many other GPCRs have been shown to signal from endosomes via cAMP, and recent studies have proposed that location of cAMP generation impacts physiological outcomes of GPCR signaling. Here, we review the extensive literature regarding PTH1R endosomal signaling via cAMP, the mechanisms that regulate endosomal generation of cAMP, and the implications of spatial bias in PTH1R physiological functions.
Collapse
Affiliation(s)
- Karina A Peña
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Sofya Savransky
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Graduate Program in Molecular Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Breanna Lewis
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Perumal NL, Padidela R. Phosphate Homeostasis and Disorders of Phosphate Metabolism. Curr Pediatr Rev 2024; 20:412-425. [PMID: 36545737 DOI: 10.2174/1573396319666221221121350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022]
Abstract
Phosphate is indispensable for human life and evolutionary changes over several millions of years have established tightly regulated mechanisms to ensure phosphate homeostasis. In this process, calcium and phosphate metabolism have come to be intricately linked together. Three hormones (PTH, FGF23 and Calcitriol) maintain the fine balance of calcium and phosphate metabolism through their actions at three sites (the gut, the kidneys and the skeleton). Disorders that disrupt this balance can have serious clinical consequences. Acute changes in serum phosphate levels can result in life threatening complications like respiratory failure and cardiac arrythmias. Chronic hypophosphataemia predominantly affects the musculoskeletal system and presents as impaired linear growth, rickets, osteomalacia and dental problems. Hyperphosphataemia is very common in the setting of chronic kidney disease and can be difficult to manage. A thorough understanding of calcium and phosphate homeostasis is essential to diagnose and treat conditions associated with hypo and hyperphosphataemia. In this review, we will discuss the calcium and phosphate metabolism, aetiologies and management of hypo and hyperphosphataemia.
Collapse
Affiliation(s)
| | - Raja Padidela
- Department of Endocrinology, Royal Manchester Children's Hospital, Manchester, United Kingdom
| |
Collapse
|
4
|
Wang J, Zhao C, Zhang X, Yang L, Hu Y. Identification of a novel heterozygous PTH1R variant in a Chinese family with incomplete penetrance. Mol Genet Genomic Med 2024; 12:e2301. [PMID: 37840415 PMCID: PMC10767579 DOI: 10.1002/mgg3.2301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/21/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Mutations in PTH1R are associated with Jansen-type metaphyseal chondrodysplasia (JMC), Blomstrand osteochondrodysplasia (BOCD), Eiken syndrome, enchondroma, and primary failure of tooth eruption (PFE). Inheritance of the PTH1R gene can be either autosomal dominant or autosomal recessive, indicating the complexity of the gene. Our objective was to identify the phenotypic differences in members of a family with a novel PTH1R mutation. METHODS The proband was a 13-year, 6-month-old girl presenting with short stature, abnormal tooth eruption, skeletal dysplasia, and midface hypoplasia. The brother and father of the proband presented with short stature and abnormal tooth eruption. High-throughput sequencing was performed on the proband, and the variant was confirmed in the proband and other family members by Sanger sequencing. Amino acid sequence alignment was performed using ClustalX software. Three-dimensional structures were analyzed and displayed using the I-TASSER website and PyMOL software. RESULTS High-throughput genome sequencing and Sanger sequencing validation showed that the proband, her father, and her brother all carried the PTH1R (NM_000316) c.1393G>A (p.E465K) mutation. The c.1393G>A (p.E465K) mutation was novel, as it has not been reported in the literature database. According to the American College of Medical Genetics and Genomics (ACMG) guidelines, the p.E465K variant was considered to have uncertain significance. Biological information analysis demonstrated that this identified variant was highly conserved and highly likely pathogenic. CONCLUSIONS We identified a novel heterozygous mutation in the PTH1R gene leading to clinical manifestations with incomplete penetrance that expands the spectrum of known PTH1R mutations.
Collapse
Affiliation(s)
- Jie Wang
- Department of Pediatrics, Linyi People's HospitalPostgrad Training Base Jinzhou Med UniversityLinyiChina
- Department of PediatricsLinyi People's HospitalLinyiChina
| | - Chaoyue Zhao
- Department of Pediatrics, Linyi People's HospitalPostgrad Training Base Jinzhou Med UniversityLinyiChina
- Department of PediatricsLinyi People's HospitalLinyiChina
| | - Xin Zhang
- Department of Pediatrics, Linyi People's HospitalPostgrad Training Base Jinzhou Med UniversityLinyiChina
- Department of PediatricsLinyi People's HospitalLinyiChina
| | - Li Yang
- Department of PediatricsLinyi People's HospitalLinyiChina
| | - Yanyan Hu
- Department of PediatricsLinyi People's HospitalLinyiChina
| |
Collapse
|
5
|
Reyes M, Firat D, Hanna P, Khan M, Bruce M, Shvedova M, Kobayashi T, Schipani E, Gardella TJ, Jüppner H. Substantially Delayed Maturation of Growth Plate Chondrocytes in "Humanized" PTH1R Mice with the H223R Mutation of Jansen's Disease. JBMR Plus 2023; 7:e10802. [PMID: 37808400 PMCID: PMC10556264 DOI: 10.1002/jbm4.10802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 10/10/2023] Open
Abstract
Activating parathyroid hormone (PTH)/PTH-related Peptide (PTHrP) receptor (PTH1R) mutations causes Jansen's metaphyseal chondrodysplasia (JMC), a rare disease characterized by growth plate abnormalities, short stature, and PTH-independent hypercalcemia. Previously generated transgenic JMC mouse models, in which the human PTH1R allele with the H223R mutation (H223R-PTH1R) is expressed in osteoblasts via type Ia1 collagen or DMP1 promoters cause excess bone mass, while expression of the mutant allele via the type IIa1 collagen promoter results in only minor growth plate changes. Thus, neither transgenic JMC model adequately recapitulates the human disease. We therefore generated "humanized" JMC mice in which the H223R-PTH1R allele was expressed via the endogenous mouse Pth1r promoter and, thus, in all relevant target tissues. Founders with the H223R allele typically died within 2 months without reproducing; several mosaic male founders, however, lived longer and produced F1 H223R-PTH1R offspring, which were small and exhibited marked growth plate abnormalities. Serum calcium and phosphate levels of the mutant mice were not different from wild-type littermates, but serum PTH and P1NP were reduced significantly, while CTX-1 and CTX-2 were slightly increased. Histological and RNAscope analyses of the mutant tibial growth plates revealed markedly expanded zones of type II collagen-positive, proliferating/prehypertrophic chondrocytes, abundant apoptotic cells in the growth plate center and a progressive reduction of type X collagen-positive hypertrophic chondrocytes and primary spongiosa. The "humanized" H223R-PTH1R mice are likely to provide a more suitable model for defining the JMC phenotype and for assessing potential treatment options for this debilitating disease of skeletal development and mineral ion homeostasis. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Monica Reyes
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Damla Firat
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Patrick Hanna
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Mohd Khan
- Department of Orthopedic SurgeryUniversity of Pennsylvania, Perelman Medical SchoolPhiladelphiaPAUSA
| | - Michael Bruce
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Maria Shvedova
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Tatsuya Kobayashi
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Ernestina Schipani
- Department of Orthopedic SurgeryUniversity of Pennsylvania, Perelman Medical SchoolPhiladelphiaPAUSA
| | - Thomas J. Gardella
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Harald Jüppner
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
- Pediatric Nephrology UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
6
|
Vilardaga JP, Clark LJ, White AD, Sutkeviciute I, Lee JY, Bahar I. Molecular Mechanisms of PTH/PTHrP Class B GPCR Signaling and Pharmacological Implications. Endocr Rev 2023; 44:474-491. [PMID: 36503956 PMCID: PMC10461325 DOI: 10.1210/endrev/bnac032] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
The classical paradigm of G protein-coupled receptor (GPCR) signaling via G proteins is grounded in a view that downstream responses are relatively transient and confined to the cell surface, but this notion has been revised in recent years following the identification of several receptors that engage in sustained signaling responses from subcellular compartments following internalization of the ligand-receptor complex. This phenomenon was initially discovered for the parathyroid hormone (PTH) type 1 receptor (PTH1R), a vital GPCR for maintaining normal calcium and phosphate levels in the body with the paradoxical ability to build or break down bone in response to PTH binding. The diverse biological processes regulated by this receptor are thought to depend on its capacity to mediate diverse modes of cyclic adenosine monophosphate (cAMP) signaling. These include transient signaling at the plasma membrane and sustained signaling from internalized PTH1R within early endosomes mediated by PTH. Here we discuss recent structural, cell signaling, and in vivo studies that unveil potential pharmacological outputs of the spatial versus temporal dimension of PTH1R signaling via cAMP. Notably, the combination of molecular dynamics simulations and elastic network model-based methods revealed how precise modulation of PTH signaling responses is achieved through structure-encoded allosteric coupling within the receptor and between the peptide hormone binding site and the G protein coupling interface. The implications of recent findings are now being explored for addressing key questions on how location bias in receptor signaling contributes to pharmacological functions, and how to drug a difficult target such as the PTH1R toward discovering nonpeptidic small molecule candidates for the treatment of metabolic bone and mineral diseases.
Collapse
Affiliation(s)
- Jean-Pierre Vilardaga
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Lisa J Clark
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Alex D White
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Ieva Sutkeviciute
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Ji Young Lee
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
7
|
Cary BP, Gerrard EJ, Belousoff MJ, Fletcher MM, Jiang Y, Russell IC, Piper SJ, Wootten D, Sexton PM. Molecular insights into peptide agonist engagement with the PTH receptor. Structure 2023:S0969-2126(23)00125-9. [PMID: 37148874 DOI: 10.1016/j.str.2023.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/30/2022] [Accepted: 04/03/2023] [Indexed: 05/08/2023]
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.
Collapse
Affiliation(s)
- Brian P Cary
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| | - Elliot J Gerrard
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Matthew J Belousoff
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Madeleine M Fletcher
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Yan Jiang
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Isabella C Russell
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Sarah J Piper
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Denise Wootten
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| | - Patrick M Sexton
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| |
Collapse
|
8
|
Cary BP, Zhang X, Cao J, Johnson RM, Piper SJ, Gerrard EJ, Wootten D, Sexton PM. New insights into the structure and function of class B1 GPCRs. Endocr Rev 2022; 44:492-517. [PMID: 36546772 PMCID: PMC10166269 DOI: 10.1210/endrev/bnac033] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/07/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors. Class B1 GPCRs constitute a subfamily of 15 receptors that characteristically contain large extracellular domains (ECDs) and respond to long polypeptide hormones. Class B1 GPCRs are critical regulators of homeostasis, and as such, many are important drug targets. While most transmembrane proteins, including GPCRs, are recalcitrant to crystallization, recent advances in electron cryo-microscopy (cryo-EM) have facilitated a rapid expansion of the structural understanding of membrane proteins. As a testament to this success, structures for all the class B1 receptors bound to G proteins have been determined by cryo-EM in the past five years. Further advances in cryo-EM have uncovered dynamics of these receptors, ligands, and signalling partners. Here, we examine the recent structural underpinnings of the class B1 GPCRs with an emphasis on structure-function relationships.
Collapse
Affiliation(s)
- Brian P Cary
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Xin Zhang
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Jianjun Cao
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Rachel M Johnson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Sarah J Piper
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Elliot J Gerrard
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| |
Collapse
|
9
|
Yamaguchi T, Hosomichi K, Shirota T, Miyamoto Y, Ono W, Ono N. Primary failure of tooth eruption: Etiology and management. JAPANESE DENTAL SCIENCE REVIEW 2022; 58:258-267. [PMID: 36159186 PMCID: PMC9489741 DOI: 10.1016/j.jdsr.2022.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/01/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Primary failure of eruption (PFE) is a rare disorder defined as incomplete tooth eruption despite the presence of a clear eruption pathway. PFE is known to be caused by rare variants in the parathyroid hormone 1 receptor gene (PTH1R). Although several PTH1R variants have been reported, the etiology of PFE remains unclear. However, important studies that help elucidate the pathology of PFE have recently been published. The purpose of this review is to summarize current treatment options, clinical symptoms or phenotypes for diagnosis, genetic information including solid evidence in mouse disease models and disease-specific induced pluripotent stem cells, thus approaching the etiology of PFE from the perspective of the latest research.
Collapse
Affiliation(s)
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Tatsuo Shirota
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, Tokyo, Japan
| | - Yoichi Miyamoto
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan
| | - Wanida Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| |
Collapse
|
10
|
Zhai X, Mao C, Shen Q, Zang S, Shen DD, Zhang H, Chen Z, Wang G, Zhang C, Zhang Y, Liu Z. Molecular insights into the distinct signaling duration for the peptide-induced PTH1R activation. Nat Commun 2022; 13:6276. [PMID: 36271004 PMCID: PMC9586930 DOI: 10.1038/s41467-022-34009-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/06/2022] [Indexed: 02/08/2023] Open
Abstract
The parathyroid hormone type 1 receptor (PTH1R), a class B1 G protein-coupled receptor, plays critical roles in bone turnover and Ca2+ homeostasis. Teriparatide (PTH) and Abaloparatide (ABL) are terms as long-acting and short-acting peptide, respectively, regarding their marked duration distinctions of the downstream signaling. However, the mechanistic details remain obscure. Here, we report the cryo-electron microscopy structures of PTH- and ABL-bound PTH1R-Gs complexes, adapting similar overall conformations yet with notable differences in the receptor ECD regions and the peptide C-terminal portions. 3D variability analysis and site-directed mutagenesis studies uncovered that PTH-bound PTH1R-Gs complexes display less motions and are more tolerant of mutations in affecting the receptor signaling than ABL-bound complexes. Furthermore, we combined the structural analysis and signaling assays to delineate the molecular basis of the differential signaling durations induced by these peptides. Our study deepens the mechanistic understanding of ligand-mediated prolonged or transient signaling.
Collapse
Affiliation(s)
- Xiuwen Zhai
- grid.89957.3a0000 0000 9255 8984National Clinical Research Center of Kidney Diseases, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, 211166 Jiangsu China
| | - Chunyou Mao
- grid.13402.340000 0004 1759 700XCenter for Structural Pharmacology and Therapeutics Development, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.415999.90000 0004 1798 9361Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingya Shen
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Shaokun Zang
- grid.13402.340000 0004 1759 700XDepartment of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan-Dan Shen
- grid.13402.340000 0004 1759 700XDepartment of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huibing Zhang
- grid.13402.340000 0004 1759 700XDepartment of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhaohong Chen
- grid.89957.3a0000 0000 9255 8984National Clinical Research Center of Kidney Diseases, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, 211166 Jiangsu China
| | - Gang Wang
- grid.89957.3a0000 0000 9255 8984National Clinical Research Center of Kidney Diseases, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, 211166 Jiangsu China
| | - Changming Zhang
- grid.89957.3a0000 0000 9255 8984National Clinical Research Center of Kidney Diseases, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, 211166 Jiangsu China
| | - Yan Zhang
- grid.13402.340000 0004 1759 700XCenter for Structural Pharmacology and Therapeutics Development, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China ,grid.13402.340000 0004 1759 700XDepartment of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XMOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,Zhejiang Provincial Key Laboratory of Immunity and Inflammatory diseases, Hangzhou, Zhejiang China
| | - Zhihong Liu
- grid.89957.3a0000 0000 9255 8984National Clinical Research Center of Kidney Diseases, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, 211166 Jiangsu China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| |
Collapse
|
11
|
Gorvin CM. Genetic causes of neonatal and infantile hypercalcaemia. Pediatr Nephrol 2022; 37:289-301. [PMID: 33990852 PMCID: PMC8816529 DOI: 10.1007/s00467-021-05082-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/25/2021] [Accepted: 04/06/2021] [Indexed: 12/02/2022]
Abstract
The causes of hypercalcaemia in the neonate and infant are varied, and often distinct from those in older children and adults. Hypercalcaemia presents clinically with a range of symptoms including failure to thrive, poor feeding, constipation, polyuria, irritability, lethargy, seizures and hypotonia. When hypercalcaemia is suspected, an accurate diagnosis will require an evaluation of potential causes (e.g. family history) and assessment for physical features (such as dysmorphology, or subcutaneous fat deposits), as well as biochemical measurements, including total and ionised serum calcium, serum phosphate, creatinine and albumin, intact parathyroid hormone (PTH), vitamin D metabolites and urinary calcium, phosphate and creatinine. The causes of neonatal hypercalcaemia can be classified into high or low PTH disorders. Disorders associated with high serum PTH include neonatal severe hyperparathyroidism, familial hypocalciuric hypercalcaemia and Jansen's metaphyseal chondrodysplasia. Conditions associated with low serum PTH include idiopathic infantile hypercalcaemia, Williams-Beuren syndrome and inborn errors of metabolism, including hypophosphatasia. Maternal hypocalcaemia and dietary factors and several rare endocrine disorders can also influence neonatal serum calcium levels. This review will focus on the common causes of hypercalcaemia in neonates and young infants, considering maternal, dietary, and genetic causes of calcium dysregulation. The clinical presentation and treatment of patients with these disorders will be discussed.
Collapse
Affiliation(s)
- Caroline M. Gorvin
- Institute of Metabolism and Systems Research and Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, B15 2TT UK ,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, B15 2TT UK
| |
Collapse
|
12
|
Schihada H, Shekhani R, Schulte G. Quantitative assessment of constitutive G protein-coupled receptor activity with BRET-based G protein biosensors. Sci Signal 2021; 14:eabf1653. [PMID: 34516756 DOI: 10.1126/scisignal.abf1653] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hannes Schihada
- Section for Receptor Biology and Signaling, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17165 Stockholm, Sweden
| | - Rawan Shekhani
- Section for Receptor Biology and Signaling, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17165 Stockholm, Sweden
| | - Gunnar Schulte
- Section for Receptor Biology and Signaling, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17165 Stockholm, Sweden
| |
Collapse
|
13
|
Isojima T, Sims NA. Cortical bone development, maintenance and porosity: genetic alterations in humans and mice influencing chondrocytes, osteoclasts, osteoblasts and osteocytes. Cell Mol Life Sci 2021; 78:5755-5773. [PMID: 34196732 PMCID: PMC11073036 DOI: 10.1007/s00018-021-03884-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/06/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
Cortical bone structure is a crucial determinant of bone strength, yet for many years studies of novel genes and cell signalling pathways regulating bone strength have focused on the control of trabecular bone mass. Here we focus on mechanisms responsible for cortical bone development, growth, and degeneration, and describe some recently described genetic-driven modifications in humans and mice that reveal how these processes may be controlled. We start with embryonic osteogenesis of preliminary bone structures preceding the cortex and describe how this structure consolidates then matures to a dense, vascularised cortex containing an increasing proportion of lamellar bone. These processes include modelling-induced, and load-dependent, asymmetric cortical expansion, which enables the cortex's transition from a highly porous woven structure to a consolidated and thickened highly mineralised lamellar bone structure, infiltrated by vascular channels. Sex-specific differences emerge during this process. With aging, the process of consolidation reverses: cortical pores enlarge, leading to greater cortical porosity, trabecularisation and loss of bone strength. Each process requires co-ordination between bone formation, bone mineralisation, vascularisation, and bone resorption, with a need for locational-, spatial- and cell-specific signalling pathways to mediate this co-ordination. We will discuss these processes, and a number of cell-signalling pathways identified in both murine and human genetic studies to regulate cortical bone mass, including signalling through gp130, STAT3, PTHR1, WNT16, NOTCH, NOTUM and sFRP4.
Collapse
Affiliation(s)
- Tsuyoshi Isojima
- St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC, 3122, Australia
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC, 3122, Australia.
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia.
| |
Collapse
|
14
|
Weaver SR, Taylor EL, Zars EL, Arnold KM, Bradley EW, Westendorf JJ. Pleckstrin homology (PH) domain and Leucine Rich Repeat Phosphatase 1 (Phlpp1) Suppresses Parathyroid Hormone Receptor 1 (Pth1r) Expression and Signaling During Bone Growth. J Bone Miner Res 2021; 36:986-999. [PMID: 33434347 PMCID: PMC8131217 DOI: 10.1002/jbmr.4248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/06/2020] [Accepted: 12/24/2020] [Indexed: 12/20/2022]
Abstract
Endochondral ossification is tightly controlled by a coordinated network of signaling cascades including parathyroid hormone (PTH). Pleckstrin homology (PH) domain and leucine rich repeat phosphatase 1 (Phlpp1) affects endochondral ossification by suppressing chondrocyte proliferation in the growth plate, longitudinal bone growth, and bone mineralization. As such, Phlpp1-/- mice have shorter long bones, thicker growth plates, and proportionally larger growth plate proliferative zones. The goal of this study was to determine how Phlpp1 deficiency affects PTH signaling during bone growth. Transcriptomic analysis revealed greater PTH receptor 1 (Pth1r) expression and enrichment of histone 3 lysine 27 acetylation (H3K27ac) at the Pth1r promoter in Phlpp1-deficient chondrocytes. PTH (1-34) enhanced and PTH (7-34) attenuated cell proliferation, cAMP signaling, cAMP response element-binding protein (CREB) phosphorylation, and cell metabolic activity in Phlpp1-inhibited chondrocytes. To understand the role of Pth1r action in the endochondral phenotypes of Phlpp1-deficient mice, Phlpp1-/- mice were injected with Pth1r ligand PTH (7-34) daily for the first 4 weeks of life. PTH (7-34) reversed the abnormal growth plate and long-bone growth phenotypes of Phlpp1-/- mice but did not rescue deficits in bone mineral density or trabecular number. These results show that elevated Pth1r expression and signaling contributes to increased proliferation in Phlpp1-/- chondrocytes and shorter bones in Phlpp1-deficient mice. Our data reveal a novel molecular relationship between Phlpp1 and Pth1r in chondrocytes during growth plate development and longitudinal bone growth. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
| | | | | | | | - Elizabeth W. Bradley
- Department of Orthopedic Surgery and Stem Cell Institute, University of Minnesota, Minneapolis, MN
| | - Jennifer J. Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
15
|
Josephs TM, Belousoff MJ, Liang YL, Piper SJ, Cao J, Garama DJ, Leach K, Gregory KJ, Christopoulos A, Hay DL, Danev R, Wootten D, Sexton PM. Structure and dynamics of the CGRP receptor in apo and peptide-bound forms. Science 2021; 372:science.abf7258. [DOI: 10.1126/science.abf7258] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/11/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Tracy M. Josephs
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Matthew J. Belousoff
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Yi-Lynn Liang
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Sarah J. Piper
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Jianjun Cao
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Daniel J. Garama
- Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton 3168, Victoria, Australia
| | - Katie Leach
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Karen J. Gregory
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Debbie L. Hay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9054, New Zealand
| | - Radostin Danev
- Graduate School of Medicine, University of Tokyo, N415, 7-3-1 Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Denise Wootten
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Patrick M. Sexton
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Enchondroma is a common cartilage benign tumor that develops from dysregulation of chondrocyte terminal differentiation during growth plate development. Here we provide an overview of recent progress in understanding causative mutations for enchondroma, dysregulated signaling and metabolic pathways in enchondroma, and the progression from enchondroma to malignant chondrosarcoma. RECENT FINDINGS Several signaling pathways that regulate chondrocyte differentiation are dysregulated in enchondromas. Somatic mutations in the metabolic enzymes isocitrate dehydrogenase 1 and 2 (IDH1/2) are the most common findings in enchondromas. Mechanisms including metabolic regulation, epigenetic regulation, and altered signaling pathways play a role in enchondroma formation and progression. Multiple pathways regulate growth plate development in a coordinated manner. Deregulation of the process can result in chondrocytes failing to undergo differentiation and the development of enchondroma.
Collapse
Affiliation(s)
- Hongyuan Zhang
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
17
|
Yang LK, Hou ZS, Tao YX. Biased signaling in naturally occurring mutations of G protein-coupled receptors associated with diverse human diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:165973. [PMID: 32949766 PMCID: PMC7722056 DOI: 10.1016/j.bbadis.2020.165973] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptors (GPCRs) play critical roles in transmitting a variety of extracellular signals into the cells and regulate diverse physiological functions. Naturally occurring mutations that result in dysfunctions of GPCRs have been known as the causes of numerous diseases. Significant progresses have been made in elucidating the pathophysiology of diseases caused by mutations. The multiple intracellular signaling pathways, such as G protein-dependent and β-arrestin-dependent signaling, in conjunction with recent advances on biased agonism, have broadened the view on the molecular mechanism of disease pathogenesis. This review aims to briefly discuss biased agonism of GPCRs (biased ligands and biased receptors), summarize the naturally occurring GPCR mutations that cause biased signaling, and propose the potential pathophysiological relevance of biased mutant GPCRs associated with various endocrine diseases.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Zhi-Shuai Hou
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
18
|
Noda H, Guo J, Khatri A, Dean T, Reyes M, Armanini M, Brooks DJ, Martins JS, Schipani E, Bouxsein ML, Demay MB, Potts JT, Jüppner H, Gardella TJ. An Inverse Agonist Ligand of the PTH Receptor Partially Rescues Skeletal Defects in a Mouse Model of Jansen's Metaphyseal Chondrodysplasia. J Bone Miner Res 2020; 35:540-549. [PMID: 31693237 PMCID: PMC8050614 DOI: 10.1002/jbmr.3913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 10/14/2019] [Accepted: 10/24/2019] [Indexed: 01/09/2023]
Abstract
Jansen's metaphyseal chondrodysplasia (JMC) is a rare disease of bone and mineral ion physiology that is caused by activating mutations in PTHR1. Ligand-independent signaling by the mutant receptors in cells of bone and kidney results in abnormal skeletal growth, excessive bone turnover, and chronic hypercalcemia and hyperphosphaturia. Clinical features further include short stature, limb deformities, nephrocalcinosis, and progressive losses in kidney function. There is no effective treatment option available for JMC. In previous cell-based assays, we found that certain N-terminally truncated PTH and PTHrP antagonist peptides function as inverse agonists and thus can reduce the high rates of basal cAMP signaling exhibited by the mutant PTHR1s of JMC in vitro. Here we explored whether one such inverse agonist ligand, [Leu11 ,dTrp12 ,Trp23 ,Tyr36 ]-PTHrP(7-36)NH2 (IA), can be effective in vivo and thus ameliorate the skeletal abnormalities that occur in transgenic mice expressing the PTHR1-H223R allele of JMC in osteoblastic cells via the collagen-1α1 promoter (C1HR mice). We observed that after 2 weeks of twice-daily injection and relative to vehicle controls, the IA analog resulted in significant improvements in key skeletal parameters that characterize the C1HR mice, because it reduced the excess trabecular bone mass, bone marrow fibrosis, and levels of bone turnover markers in blood and urine. The overall findings provide proof-of-concept support for the notion that inverse agonist ligands targeted to the mutant PTHR1 variants of JMC can have efficacy in vivo. Further studies of such PTHR1 ligand analogs could help open paths toward the first treatment option for this debilitating skeletal disorder. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hiroshi Noda
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Jun Guo
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Ashok Khatri
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Thomas Dean
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Monica Reyes
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Michael Armanini
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA.,Department of Orthopedic Surgery, Harvard Medical School, Boston, MA, USA.,Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Daniel J Brooks
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA.,Department of Orthopedic Surgery, Harvard Medical School, Boston, MA, USA.,Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Janaina S Martins
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | | | - Mary L Bouxsein
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA.,Department of Orthopedic Surgery, Harvard Medical School, Boston, MA, USA.,Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - John T Potts
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Nishimori S, O’Meara MJ, Castro CD, Noda H, Cetinbas M, da Silva Martins J, Ayturk U, Brooks DJ, Bruce M, Nagata M, Ono W, Janton CJ, Bouxsein ML, Foretz M, Berdeaux R, Sadreyev RI, Gardella TJ, Jüppner H, Kronenberg HM, Wein MN. Salt-inducible kinases dictate parathyroid hormone 1 receptor action in bone development and remodeling. J Clin Invest 2019; 129:5187-5203. [PMID: 31430259 PMCID: PMC6877304 DOI: 10.1172/jci130126] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/16/2019] [Indexed: 12/30/2022] Open
Abstract
The parathyroid hormone 1 receptor (PTH1R) mediates the biologic actions of parathyroid hormone (PTH) and parathyroid hormone-related protein (PTHrP). Here, we showed that salt-inducible kinases (SIKs) are key kinases that control the skeletal actions downstream of PTH1R and that this GPCR, when activated, inhibited cellular SIK activity. Sik gene deletion led to phenotypic changes that were remarkably similar to models of increased PTH1R signaling. In growth plate chondrocytes, PTHrP inhibited SIK3, and ablation of this kinase in proliferating chondrocytes rescued perinatal lethality of PTHrP-null mice. Combined deletion of Sik2 and Sik3 in osteoblasts and osteocytes led to a dramatic increase in bone mass that closely resembled the skeletal and molecular phenotypes observed when these bone cells express a constitutively active PTH1R that causes Jansen's metaphyseal chondrodysplasia. Finally, genetic evidence demonstrated that class IIa histone deacetylases were key PTH1R-regulated SIK substrates in both chondrocytes and osteocytes. Taken together, our findings establish that SIK inhibition is central to PTH1R action in bone development and remodeling. Furthermore, this work highlights the key role of cAMP-regulated SIKs downstream of GPCR action.
Collapse
Affiliation(s)
- Shigeki Nishimori
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Biochemistry, Teikyo University School of Medicine, Tokyo, Japan
| | - Maureen J. O’Meara
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian D. Castro
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hiroshi Noda
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Chugai Pharmaceutical Co., Tokyo, Japan
| | - Murat Cetinbas
- Department of Molecular Biology and Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Janaina da Silva Martins
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ugur Ayturk
- Musculoskeletal Integrity Program, Hospital for Special Surgery, New York, New York, USA
| | - Daniel J. Brooks
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Advanced Orthopedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Bruce
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mizuki Nagata
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Wanida Ono
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Christopher J. Janton
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mary L. Bouxsein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Advanced Orthopedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Marc Foretz
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology and Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas J. Gardella
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Henry M. Kronenberg
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marc N. Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Sutkeviciute I, Clark LJ, White AD, Gardella TJ, Vilardaga JP. PTH/PTHrP Receptor Signaling, Allostery, and Structures. Trends Endocrinol Metab 2019; 30:860-874. [PMID: 31699241 PMCID: PMC6857722 DOI: 10.1016/j.tem.2019.07.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/09/2019] [Accepted: 07/12/2019] [Indexed: 02/08/2023]
Abstract
The parathyroid hormone (PTH) type 1 receptor (PTHR) is the canonical G protein-coupled receptor (GPCR) for PTH and PTH-related protein (PTHrP) and the key regulator of calcium homeostasis and bone turnover. PTHR function is critical for human health to maintain homeostatic control of ionized serum Ca2+ levels and has several unusual signaling features, such as endosomal cAMP signaling, that are well-studied but not structurally understood. In this review, we discuss how recently solved high resolution near-atomic structures of hormone-bound PTHR in its inactive and active signaling states and discovery of extracellular Ca2+ allosterism shed light on the structural basis for PTHR signaling and function.
Collapse
Affiliation(s)
- Ieva Sutkeviciute
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Lisa J Clark
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Graduate Program in Molecular Biophysics and Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Alex D White
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Graduate Program in Molecular Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jean-Pierre Vilardaga
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
21
|
Abstract
Fibroblast growth factor 23 (FGF23), one of the endocrine fibroblast growth factors, is a principal regulator in the maintenance of serum phosphorus concentration. Binding to its cofactor αKlotho and a fibroblast growth factor receptor is essential for its activity. Its regulation and interaction with other factors in the bone-parathyroid-kidney axis is complex. FGF23 reduces serum phosphorus concentration through decreased reabsorption of phosphorus in the kidney and by decreasing 1,25 dihydroxyvitamin D (1,25(OH)2D) concentrations. Various FGF23-mediated disorders of renal phosphate wasting share similar clinical and biochemical features. The most common of these is X-linked hypophosphatemia (XLH). Additional disorders of FGF23 excess include autosomal dominant hypophosphatemic rickets, autosomal recessive hypophosphatemic rickets, fibrous dysplasia, and tumor-induced osteomalacia. Treatment is challenging, requiring careful monitoring and titration of dosages to optimize effectiveness and to balance side effects. Conventional therapy for XLH and other disorders of FGF23-mediated hypophosphatemia involves multiple daily doses of oral phosphate salts and active vitamin D analogs, such as calcitriol or alfacalcidol. Additional treatments may be used to help address side effects of conventional therapy such as thiazides to address hypercalciuria or nephrocalcinosis, and calcimimetics to manage hyperparathyroidism. The recent development and approval of an anti-FGF23 antibody, burosumab, for use in XLH provides a novel treatment option.
Collapse
Affiliation(s)
- Anisha Gohil
- Indiana University School of Medicine, Riley Hospital for Children, Fellow, Endocrinology and Diabetes, 705 Riley Hospital Drive, Room 5960, Indianapolis, IN 46202, USA, E-mail:
| | - Erik A Imel
- Indiana University School of Medicine, Riley Hospital for Children, Associate Professor of Medicine and Pediatrics, 1120 West Michigan Street, CL 459, Indianapolis, IN 46202, USA
| |
Collapse
|
22
|
Yip RK, Chan D, Cheah KS. Mechanistic insights into skeletal development gained from genetic disorders. Curr Top Dev Biol 2019; 133:343-385. [DOI: 10.1016/bs.ctdb.2019.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Moirangthem A, Narayanan DL, Jacob P, Nishimura G, Mortier G, Girisha KM. Report of second case and clinical and molecular characterization of Eiken syndrome. Clin Genet 2018; 94:457-460. [PMID: 29987841 DOI: 10.1111/cge.13413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 11/29/2022]
Abstract
We report a boy with Eiken syndrome caused by a homozygous missense variant in Parathyroid hormone 1 receptor (PTH1R) c.103G > A [p.(Glu35Lys)]. Eiken syndrome is a very rare skeletal dysplasia due to bi-allelic variants in PTH1R. Only one affected family has been known to-date. The hallmarks include delayed ossification of bone including the epiphyses, pubic symphysis, and primary ossification centers of the short tubular bones, coarse bone trabeculae, and modeling abnormalities. The phenotype being described here recapitulates the delayed ossification and modeling abnormalities of Eiken syndrome. In addition, supernumerary epiphyses of the tubular bones of the hands and primary failure of eruption of teeth were observed in our proband. This report characterizes Eiken syndrome and confirms that bi-allelic hypomorphic variants in PTH1R are probably to cause this condition.
Collapse
Affiliation(s)
- A Moirangthem
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - D L Narayanan
- Department of Medical Genetics, Nizam's Institute of Medical Sciences, Hyderabad, India
| | - P Jacob
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - G Nishimura
- Center for Intractable Diseases, Saitama Medical University Hospital, Saitama, Japan
| | - G Mortier
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - K M Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
24
|
Saito H, Noda H, Gatault P, Bockenhauer D, Loke KY, Hiort O, Silve C, Sharwood E, Martin RM, Dillon MJ, Gillis D, Harris M, Rao SD, Pauli RM, Gardella TJ, Jüppner H. Progression of Mineral Ion Abnormalities in Patients With Jansen Metaphyseal Chondrodysplasia. J Clin Endocrinol Metab 2018; 103:2660-2669. [PMID: 29788189 PMCID: PMC6486824 DOI: 10.1210/jc.2018-00332] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/09/2018] [Indexed: 12/21/2022]
Abstract
CONTEXT Five different activating PTH/PTH-related peptide (PTHrP) receptor (PTHR1) mutations have been reported as causes of Jansen metaphyseal chondrodysplasia (JMC), a rare disorder characterized by severe growth plate abnormalities and PTH-independent hypercalcemia. OBJECTIVES Assess the natural history of clinical and laboratory findings in 24 patients with JMC and characterize the disease-causing mutant receptors in vitro. PATIENTS AND METHODS The H223R mutation occurred in 18 patients. T410P, I458R and I458K each occurred in single cases; T410R was present in a father and his two sons. Laboratory records were analyzed individually and in aggregate. RESULTS Postnatal calcium levels were normal in most patients, but elevated between 0.15 and 10 years (11.8 ± 1.37 mg/dL) and tended to normalize in adults (10.0 ± 1.03 mg/dL). Mean phosphate levels were at the lower end of the age-specific normal ranges. Urinary calcium/creatinine (mg/mg) were consistently elevated (children, 0.80 ± 0.40; adults, 0.28 ± 0.19). Adult heights were well below the 3rd percentile for all patients, except for those with the T410R mutation. Most patients with JMC had undergone orthopedic surgical procedures, most had nephrocalcinosis, and two had advanced chronic kidney disease. The five PTHR1 mutants showed varying degrees of constitutive and PTH-stimulated cAMP signaling activity when expressed in HEK293 reporter cells. The inverse agonist [L11,dW12,W23,Y36]PTHrP(7-36) reduced basal cAMP signaling for each PTHR1 mutant. CONCLUSIONS Except for T410R, the other PTHR1 mutations were associated with indistinguishable mineral ion abnormalities and cause similarly severe growth impairment. Hypercalciuria persisted into adulthood. An inverse agonist ligand effectively reduced in vitro PTH-independent cAMP formation at all five PTHR1 mutants, suggesting a potential path toward therapy.
Collapse
Affiliation(s)
- Hiroshi Saito
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hiroshi Noda
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Philippe Gatault
- Service de Néphrologie-Hypertensiologie, Transplantation - Dialyses, University Hospital, Tours, France
| | - Detlef Bockenhauer
- University College London Centre for Nephrology, Royal Free Hospital, London United Kingdom
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, London United Kingdom
| | - Kah Yin Loke
- Department of Pediatrics, National University Hospital, Singapore Singapore
| | - Olaf Hiort
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Caroline Silve
- Service de Biochimie et Génétique Moléculaires, Hôpital Cochin, AP-HP, Paris, France
- Centre de Référence des Maladies rares du Calcium et du Phosphore and Filière de Santé Maladies Rares OSCAR, AP-HP, Paris, France
| | - Erin Sharwood
- Endocrinology Department, Lady Cilento Children's Hospital, Brisbane, Queensland, Australia
| | - Regina Matsunaga Martin
- Osteometabolic Disorders Unit, Division of Endocrinology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
- Laboratory of Hormones and Molecular Genetics/LIM42, Division of Endocrinology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Michael J Dillon
- University College London Centre for Nephrology, Royal Free Hospital, London United Kingdom
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, London United Kingdom
| | - David Gillis
- Department of Pediatrics, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Mark Harris
- Endocrinology Department, Lady Cilento Children's Hospital, Brisbane, Queensland, Australia
- Mater Research Institute–University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Sudhaker D Rao
- Bone and Mineral Research Laboratory and Division of Endocrinology, Diabetes, and Bone & Mineral Disorders, Department of Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Richard M Pauli
- Division of Genetics and Metabolism, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Correspondence and Reprint Requests: Harald Jüppner, MD, Endocrine Unit, Massachusetts General Hospital, 50 Blossom Street, Boston, Massachusetts 02114. E-mail:
| |
Collapse
|
25
|
Brommage R, Ohlsson C. Translational studies provide insights for the etiology and treatment of cortical bone osteoporosis. Best Pract Res Clin Endocrinol Metab 2018; 32:329-340. [PMID: 29779585 DOI: 10.1016/j.beem.2018.02.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increasing attention is being focused on the important contributions of cortical bone to bone strength, fractures and osteoporosis therapies. Recent progress in human genome wide association studies in combination with high-throughput mouse gene knockout phenotyping efforts of multiple genes and advanced conditional gene inactivation in mouse models have successfully identified genes with crucial roles in cortical bone homeostasis. Particular attention in this review is given to genes, such as WNT16, POSTN and SFRP4, that differentially affect cortical and trabecular bone architecture. We propose that animal models of cortical bone metabolism will substantially contribute to developing anabolic osteoporosis therapies that improve cortical bone mass and reduce non-vertebral fracture risk.
Collapse
Affiliation(s)
- Robert Brommage
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
26
|
Hu L, Chang L, Zhang Y, Zhai L, Zhang S, Qi Z, Yan H, Yan Y, Luo X, Zhang S, Wang Y, Kunapuli SP, Ye H, Ding Z. Platelets Express Activated P2Y 12 Receptor in Patients With Diabetes Mellitus. Circulation 2017. [PMID: 28637879 DOI: 10.1161/circulationaha.116.026995] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Platelets from patients with diabetes mellitus are hyperactive. Hyperactivated platelets may contribute to cardiovascular complications and inadequate responses to antiplatelet agents in the setting of diabetes mellitus. However, the underlying mechanism of hyperactivated platelets is not completely understood. METHODS We measured P2Y12 expression on platelets from patients with type 2 diabetes mellitus and on platelets from rats with diabetes mellitus. We also assayed platelet P2Y12 activation by measuring cAMP and VASP phosphorylation. The antiplatelet and antithrombotic effects of AR-C78511 and cangrelor were compared in rats. Finally, we explored the role of the nuclear factor-κB pathway in regulating P2Y12 receptor expression in megakaryocytes. RESULTS Platelet P2Y12 levels are 4-fold higher in patients with type 2 diabetes mellitus compared with healthy subjects. P2Y12 expression correlates with ADP-induced platelet aggregation (r=0.89, P<0.01). P2Y12 in platelets from patients with diabetes mellitus is constitutively activated. Although both AR-C78511, a potent P2Y12 inverse agonist, and cangrelor have similar antiplatelet efficacy on platelets from healthy subjects, AR-C78511 exhibits more powerful antiplatelet effects on diabetic platelets than cangrelor (aggregation ratio 36±3% versus 49±5%, respectively, P<0.05). Using a FeCl3-injury mesenteric arteriole thrombosis model in rats and an arteriovenous shunt thrombosis model in rats, we found that the inverse agonist AR-C78511 has greater antithrombotic effects on GK rats with diabetes mellitus than cangrelor (thrombus weight 4.9±0.3 mg versus 8.3±0.4 mg, respectively, P<0.01). We also found that a pathway involving high glucose-reactive oxygen species-nuclear factor-κB increases platelet P2Y12 receptor expression in diabetes mellitus. CONCLUSIONS Platelet P2Y12 receptor expression is significantly increased and the receptor is constitutively activated in patients with type 2 diabetes mellitus, which contributes to platelet hyperactivity and limits antiplatelet drug efficacy in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Liang Hu
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin Chang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Zhang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Zhai
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shenghui Zhang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiyong Qi
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongmei Yan
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Yan
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinping Luo
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Si Zhang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiping Wang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Satya P Kunapuli
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongying Ye
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongren Ding
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
27
|
Tamura T, Noda H, Joyashiki E, Hoshino M, Watanabe T, Kinosaki M, Nishimura Y, Esaki T, Ogawa K, Miyake T, Arai S, Shimizu M, Kitamura H, Sato H, Kawabe Y. Identification of an orally active small-molecule PTHR1 agonist for the treatment of hypoparathyroidism. Nat Commun 2016; 7:13384. [PMID: 27857062 PMCID: PMC5120204 DOI: 10.1038/ncomms13384] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 09/28/2016] [Indexed: 01/09/2023] Open
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. Hypoparathyroidism and osteoporosis can be treated with parathyroid hormone, but frequent injections are required. Here the authors develop a small-molecule agonist for the parathyroid hormone type I receptor that can be administered orally, and demonstrate its efficacy in rats.
Collapse
Affiliation(s)
- Tatsuya Tamura
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Hiroshi Noda
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Eri Joyashiki
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Maiko Hoshino
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Tomoyuki Watanabe
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Masahiko Kinosaki
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Yoshikazu Nishimura
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Tohru Esaki
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Kotaro Ogawa
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Taiji Miyake
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Shinichi Arai
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Masaru Shimizu
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Hidetomo Kitamura
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Haruhiko Sato
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Yoshiki Kawabe
- Research Division, Chugai Pharmaceutical Co., Ltd., 1-135, Komakado, Gotemba, Shizuoka 412-8513, Japan
| |
Collapse
|
28
|
Nampoothiri S, Fernández-Rebollo E, Yesodharan D, Gardella TJ, Rush ET, Langman CB, Jüppner H. Jansen Metaphyseal Chondrodysplasia due to Heterozygous H223R-PTH1R Mutations With or Without Overt Hypercalcemia. J Clin Endocrinol Metab 2016; 101:4283-4289. [PMID: 27410178 PMCID: PMC5095231 DOI: 10.1210/jc.2016-2054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CONTEXT Jansen's metaphyseal chondrodysplasia (JMC) is a rare skeletal dysplasia characterized by abnormal endochondral bone formation and typically severe hypercalcemia despite normal/low levels of PTH. Five different heterozygous activating PTH/PTHrP receptor (PTH1R) mutations that change one of three different amino acid residues are known to cause JMC. OBJECTIVES Establishing the diagnosis of JMC during infancy or early childhood can be challenging, especially in the absence of family history and/or overt hypercalcemia. We therefore sought to provide radiographic findings supporting this diagnosis early in life. PATIENTS AND METHODS Three patients, a mother and her two sons, had radiographic evidence for JMC. However, obvious hypercalcemia and suppressed PTH levels were encountered only in both affected children. Sanger sequencing and endonuclease (SphI) digestion of PCR-amplified genomic DNA were performed to search for the H223R-PTH1R mutation. RESULTS The heterozygous H223R mutation was identified in all three affected individuals. Surprisingly, however, the now 38-year-old mother was never overtly hypercalcemic and was therefore not diagnosed until her sons were found to be affected by JMC at the ages of 28 months and 40 days, respectively. The presented radiographic findings at different ages will help diagnose other infants/toddlers suspected of having JMC. CONCLUSION The H223R mutation is typically associated with profound hypercalcemia despite low/normal PTH levels. However, the findings presented herein show that overt hypercalcemia is not always encountered in JMC, even if caused by this relatively frequent mutation, which is similar to observations with other PTH1R mutations that show less constitutive activity.
Collapse
Affiliation(s)
- Sheela Nampoothiri
- Department of Pediatric Genetics (S.N., D.Y.), Amrita Institute of Medical Sciences and Research Center, Aims Ponekkara PO, Cochin 682041, Kerala, India; Munroe-Meyer Institute for Genetics and Rehabilitation (E.T.R.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Kidney Diseases (C.B.L.), Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; and Endocrine Unit (E.F.-R., T.J.G., H.J.) and Pediatric Nephrology Unit (H.J.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Eduardo Fernández-Rebollo
- Department of Pediatric Genetics (S.N., D.Y.), Amrita Institute of Medical Sciences and Research Center, Aims Ponekkara PO, Cochin 682041, Kerala, India; Munroe-Meyer Institute for Genetics and Rehabilitation (E.T.R.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Kidney Diseases (C.B.L.), Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; and Endocrine Unit (E.F.-R., T.J.G., H.J.) and Pediatric Nephrology Unit (H.J.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Dhanya Yesodharan
- Department of Pediatric Genetics (S.N., D.Y.), Amrita Institute of Medical Sciences and Research Center, Aims Ponekkara PO, Cochin 682041, Kerala, India; Munroe-Meyer Institute for Genetics and Rehabilitation (E.T.R.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Kidney Diseases (C.B.L.), Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; and Endocrine Unit (E.F.-R., T.J.G., H.J.) and Pediatric Nephrology Unit (H.J.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Thomas J Gardella
- Department of Pediatric Genetics (S.N., D.Y.), Amrita Institute of Medical Sciences and Research Center, Aims Ponekkara PO, Cochin 682041, Kerala, India; Munroe-Meyer Institute for Genetics and Rehabilitation (E.T.R.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Kidney Diseases (C.B.L.), Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; and Endocrine Unit (E.F.-R., T.J.G., H.J.) and Pediatric Nephrology Unit (H.J.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Eric T Rush
- Department of Pediatric Genetics (S.N., D.Y.), Amrita Institute of Medical Sciences and Research Center, Aims Ponekkara PO, Cochin 682041, Kerala, India; Munroe-Meyer Institute for Genetics and Rehabilitation (E.T.R.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Kidney Diseases (C.B.L.), Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; and Endocrine Unit (E.F.-R., T.J.G., H.J.) and Pediatric Nephrology Unit (H.J.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Craig B Langman
- Department of Pediatric Genetics (S.N., D.Y.), Amrita Institute of Medical Sciences and Research Center, Aims Ponekkara PO, Cochin 682041, Kerala, India; Munroe-Meyer Institute for Genetics and Rehabilitation (E.T.R.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Kidney Diseases (C.B.L.), Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; and Endocrine Unit (E.F.-R., T.J.G., H.J.) and Pediatric Nephrology Unit (H.J.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Harald Jüppner
- Department of Pediatric Genetics (S.N., D.Y.), Amrita Institute of Medical Sciences and Research Center, Aims Ponekkara PO, Cochin 682041, Kerala, India; Munroe-Meyer Institute for Genetics and Rehabilitation (E.T.R.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Kidney Diseases (C.B.L.), Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; and Endocrine Unit (E.F.-R., T.J.G., H.J.) and Pediatric Nephrology Unit (H.J.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
29
|
Cheloha RW, Watanabe T, Dean T, Gellman SH, Gardella TJ. Backbone Modification of a Parathyroid Hormone Receptor-1 Antagonist/Inverse Agonist. ACS Chem Biol 2016; 11:2752-2762. [PMID: 27533344 DOI: 10.1021/acschembio.6b00404] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A backbone-modified peptide derived from parathyroid hormone (PTH) is shown to function as an inhibitor and inverse agonist of parathyroid hormone receptor-1 (PTHR1) signaling. This receptor acts to regulate calcium and phosphate homeostasis, as well as bone turnover and development. PTH is a natural agonist of PTHR1, and PTH(1-34) displays full activity relative to the natural 84-residue hormone. PTH(1-34) is used clinically to treat osteoporosis. N-terminally truncated derivatives of PTH(1-34), such as PTH(7-34), are known to bind to PTHR1 without initiating intracellular signaling and can thus act as competitive antagonists of PTH-induced signaling at PTHR1. In some cases, N-terminally truncated PTH derivatives also act as inverse agonists of PTHR1 variants that display pathologically high levels of signaling in the absence of PTH. Many analogues of PTH, however, are rapidly degraded by proteases, which may limit biomedical application. We show that backbone modification via periodic replacement of α-amino acid residues with homologous β-amino acid residues leads to an α/β-PTH(7-34) peptide that retains the antagonist and inverse agonist activities of the prototype α-peptide while exhibiting enhanced stability in the presence of aggressive proteases. These findings highlight the value of backbone-modified peptides derived from PTH as tools for investigating determinants of PTH metabolism and provide guidance for designing therapeutic agents for diseases arising from excessive ligand-dependent or ligand-independent PTHR1 activity.
Collapse
Affiliation(s)
- Ross W. Cheloha
- Department
of Chemistry, University of Wisconsin—Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Tomoyuki Watanabe
- Endocrine
Unit, Massachusetts General Hospital, 50 Blossom Street, Boston, Massachusetts 02114, United States
| | - Thomas Dean
- Endocrine
Unit, Massachusetts General Hospital, 50 Blossom Street, Boston, Massachusetts 02114, United States
| | - Samuel H. Gellman
- Department
of Chemistry, University of Wisconsin—Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Thomas J. Gardella
- Endocrine
Unit, Massachusetts General Hospital, 50 Blossom Street, Boston, Massachusetts 02114, United States
| |
Collapse
|
30
|
Martin TJ. Parathyroid Hormone-Related Protein, Its Regulation of Cartilage and Bone Development, and Role in Treating Bone Diseases. Physiol Rev 2016; 96:831-71. [DOI: 10.1152/physrev.00031.2015] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although parathyroid hormone-related protein (PTHrP) was discovered as a cancer-derived hormone, it has been revealed as an important paracrine/autocrine regulator in many tissues, where its effects are context dependent. Thus its location and action in the vasculature explained decades-long observations that injection of PTH into animals rapidly lowered blood pressure by producing vasodilatation. Its roles have been specified in development and maturity in cartilage and bone as a crucial regulator of endochondral bone formation and bone remodeling, respectively. Although it shares actions with parathyroid hormone (PTH) through the use of their common receptor, PTHR1, PTHrP has other actions mediated by regions within the molecule beyond the amino-terminal sequence that resembles PTH, including the ability to promote placental transfer of calcium from mother to fetus. A striking feature of the physiology of PTHrP is that it possesses structural features that equip it to be transported in and out of the nucleus, and makes use of a specific nuclear import mechanism to do so. Evidence from mouse genetic experiments shows that PTHrP generated locally in bone is essential for normal bone remodeling. Whereas the main physiological function of PTH is the hormonal regulation of calcium metabolism, locally generated PTHrP is the important physiological mediator of bone remodeling postnatally. Thus the use of intermittent injection of PTH as an anabolic therapy for bone appears to be a pharmacological application of the physiological function of PTHrP. There is much current interest in the possibility of developing PTHrP analogs that might enhance the therapeutic anabolic effects.
Collapse
Affiliation(s)
- T. John Martin
- St Vincent's Institute of Medical Research, Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Australia
| |
Collapse
|
31
|
Biosse Duplan M, Komla-Ebri D, Heuzé Y, Estibals V, Gaudas E, Kaci N, Benoist-Lasselin C, Zerah M, Kramer I, Kneissel M, Porta DG, Di Rocco F, Legeai-Mallet L. Meckel's and condylar cartilages anomalies in achondroplasia result in defective development and growth of the mandible. Hum Mol Genet 2016; 25:2997-3010. [PMID: 27260401 PMCID: PMC5181594 DOI: 10.1093/hmg/ddw153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 02/07/2023] Open
Abstract
Activating FGFR3 mutations in human result in achondroplasia (ACH), the most frequent form of dwarfism, where cartilages are severely disturbed causing long bones, cranial base and vertebrae defects. Because mandibular development and growth rely on cartilages that guide or directly participate to the ossification process, we investigated the impact of FGFR3 mutations on mandibular shape, size and position. By using CT scan imaging of ACH children and by analyzing Fgfr3Y367C/+ mice, a model of ACH, we show that FGFR3 gain-of-function mutations lead to structural anomalies of primary (Meckel’s) and secondary (condylar) cartilages of the mandible, resulting in mandibular hypoplasia and dysmorphogenesis. These defects are likely related to a defective chondrocyte proliferation and differentiation and pan-FGFR tyrosine kinase inhibitor NVP-BGJ398 corrects Meckel’s and condylar cartilages defects ex vivo. Moreover, we show that low dose of NVP-BGJ398 improves in vivo condyle growth and corrects dysmorphologies in Fgfr3Y367C/+ mice, suggesting that postnatal treatment with NVP-BGJ398 mice might offer a new therapeutic strategy to improve mandible anomalies in ACH and others FGFR3-related disorders.
Collapse
Affiliation(s)
- Martin Biosse Duplan
- INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France.,Service d'Odontologie, Hôpital Bretonneau, HUPNVS, AP-HP, Paris, France
| | - Davide Komla-Ebri
- INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Yann Heuzé
- UMR5199 PACEA, Université de Bordeaux, Bordeaux Archaeological Sciences Cluster Of Excellence, Université de Bordeaux, Bordeaux, France
| | - Valentin Estibals
- INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Emilie Gaudas
- INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Nabil Kaci
- INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
| | | | - Michel Zerah
- Neurochirurgie Pédiatrique, Unité de Chirurgie Craniofaciale, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Ina Kramer
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | - Federico Di Rocco
- INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France.,Neurochirurgie Pédiatrique, Unité de Chirurgie Craniofaciale, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Laurence Legeai-Mallet
- INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France .,Service de Génétique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| |
Collapse
|
32
|
Structure-Based Sequence Alignment of the Transmembrane Domains of All Human GPCRs: Phylogenetic, Structural and Functional Implications. PLoS Comput Biol 2016; 12:e1004805. [PMID: 27028541 PMCID: PMC4814114 DOI: 10.1371/journal.pcbi.1004805] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 02/11/2016] [Indexed: 11/23/2022] Open
Abstract
The understanding of G-protein coupled receptors (GPCRs) is undergoing a revolution due to increased information about their signaling and the experimental determination of structures for more than 25 receptors. The availability of at least one receptor structure for each of the GPCR classes, well separated in sequence space, enables an integrated superfamily-wide analysis to identify signatures involving the role of conserved residues, conserved contacts, and downstream signaling in the context of receptor structures. In this study, we align the transmembrane (TM) domains of all experimental GPCR structures to maximize the conserved inter-helical contacts. The resulting superfamily-wide GpcR Sequence-Structure (GRoSS) alignment of the TM domains for all human GPCR sequences is sufficient to generate a phylogenetic tree that correctly distinguishes all different GPCR classes, suggesting that the class-level differences in the GPCR superfamily are encoded at least partly in the TM domains. The inter-helical contacts conserved across all GPCR classes describe the evolutionarily conserved GPCR structural fold. The corresponding structural alignment of the inactive and active conformations, available for a few GPCRs, identifies activation hot-spot residues in the TM domains that get rewired upon activation. Many GPCR mutations, known to alter receptor signaling and cause disease, are located at these conserved contact and activation hot-spot residue positions. The GRoSS alignment places the chemosensory receptor subfamilies for bitter taste (TAS2R) and pheromones (Vomeronasal, VN1R) in the rhodopsin family, known to contain the chemosensory olfactory receptor subfamily. The GRoSS alignment also enables the quantification of the structural variability in the TM regions of experimental structures, useful for homology modeling and structure prediction of receptors. Furthermore, this alignment identifies structurally and functionally important residues in all human GPCRs. These residues can be used to make testable hypotheses about the structural basis of receptor function and about the molecular basis of disease-associated single nucleotide polymorphisms. G-protein coupled receptors (GPCRs) are a large superfamily of integral membrane proteins that share a characteristic 7 transmembrane helix fold. They detect various molecules outside of the cell and signal their presence to the inside of the cell. At least half of the 800 human GPCRs are potential drug targets, so understanding their structure and function is critical. Experimental structures are now available for at least one receptor from each GPCR class. The structure of the 7 helix fold is highly conserved even for receptors with very low sequence similarity. We analyze the available experimental structures and compare the common inter-helical contacts. Our analysis leads to a unified sequence-structure alignment of the GPCR superfamily that can then be used as the starting point for structure prediction of all other GPCRs. A key result of our analysis is a list of conserved contact residues and activation “hot-spots” residues that are critical for GPCR folding and function. We propose that mutations and natural variants of amino acids at these locations in the GPCRs can dramatically influence their activation state and alter intracellular signaling. This provides hypotheses for the molecular mechanisms underlying disease causing mutants for any GPCR.
Collapse
|
33
|
Jelani M, Kang C, Mohamoud HSA, Al-Rehaili R, Almramhi MM, Serafi R, Yang H, Al-Aama JY, Naeem M, Alkhiary YM. A novel homozygous PTH1R variant identified through whole-exome sequencing further expands the clinical spectrum of primary failure of tooth eruption in a consanguineous Saudi family. Arch Oral Biol 2016; 67:28-33. [PMID: 27019138 DOI: 10.1016/j.archoralbio.2016.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 01/28/2016] [Accepted: 03/22/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVES The present study aimed to identify the genetic cause of non-syndromic primary failure of tooth eruption in a five-generation consanguineous Saudi family using whole-exome sequencing (WES) analysis. DESIGN The family pedigree and phenotype were obtained from patient medical records. WES of all four affected family members was performed using the 51 Mb SureSelect V4 library kit and then sequenced using the Illumina HiSeq2000 sequencing system. Sequence alignment, variant calling, and the annotation of single nucleotide polymorphisms and indels were performed using standard bioinformatics pipelines. The genotype of candidate variants was confirmed in all available family members by Sanger sequencing. RESULTS Pedigree analysis suggested that the inheritance was autosomal recessive. WES of all affected individuals identified a novel homozygous variant in exon 8 of the parathyroid hormone 1 receptor gene (PTH1R) (NM_000316: c.611T>A: p.Val204Glu). CONCLUSION To the best of our knowledge, this is the first report of primary failure of eruption caused by a homozygous mutation in PTH1R. Our findings prove the application of WES as an efficient molecular diagnostics tool for this rare phenotype and further broaden the clinical spectrum of PTH1R pathogenicity.
Collapse
Affiliation(s)
- Musharraf Jelani
- Princess Al-Jawhara Albrahim Centre of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia; Medical Genetics and Molecular Biology Unit, Biochemistry Department, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan.
| | - Changsoo Kang
- Department of Biology and Institute of Basic Sciences, Sungshin Women's University, Seoul, Republic of Korea, Republic of Korea
| | - Hussein Sheikh Ali Mohamoud
- Princess Al-Jawhara Albrahim Centre of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia; Human Genetics Research Centre, Division of Biomedical Sciences (BMS), St. George's University of London (SGUL), London SW17 0RE, United Kingdom, UK
| | - Rayan Al-Rehaili
- Oral and Maxillofacial Prosthodontics Department, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mona Mohammad Almramhi
- Princess Al-Jawhara Albrahim Centre of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rehab Serafi
- Department of Dermatology, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Huanming Yang
- Princess Al-Jawhara Albrahim Centre of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia; BGI-Shenzhen, Shenzhen, China
| | - Jumana Yousuf Al-Aama
- Princess Al-Jawhara Albrahim Centre of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Naeem
- Medical Genetics Research Laboratory, Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Yaser Mohammad Alkhiary
- Oral and Maxillofacial Prosthodontics Department, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
34
|
Chan ASM, Clairfeuille T, Landao-Bassonga E, Kinna G, Ng PY, Loo LS, Cheng TS, Zheng M, Hong W, Teasdale RD, Collins BM, Pavlos NJ. Sorting nexin 27 couples PTHR trafficking to retromer for signal regulation in osteoblasts during bone growth. Mol Biol Cell 2016; 27:1367-82. [PMID: 26912788 PMCID: PMC4831889 DOI: 10.1091/mbc.e15-12-0851] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/10/2016] [Indexed: 12/26/2022] Open
Abstract
The parathyroid hormone 1 receptor (PTHR) is central to the process of bone formation and remodeling. PTHR signaling requires receptor internalization into endosomes, which is then terminated by recycling or degradation. Here we show that sorting nexin 27 (SNX27) functions as an adaptor that couples PTHR to the retromer trafficking complex. SNX27 binds directly to the C-terminal PDZ-binding motif of PTHR, wiring it to retromer for endosomal sorting. The structure of SNX27 bound to the PTHR motif reveals a high-affinity interface involving conserved electrostatic interactions. Mechanistically, depletion of SNX27 or retromer augments intracellular PTHR signaling in endosomes. Osteoblasts genetically lacking SNX27 show similar disruptions in PTHR signaling and greatly reduced capacity for bone mineralization, contributing to profound skeletal deficits in SNX27-knockout mice. Taken together, our data support a critical role for SNX27-retromer mediated transport of PTHR in normal bone development.
Collapse
Affiliation(s)
- Audrey S M Chan
- Cellular Orthopaedic Laboratory, School of Surgery, University of Western Australia, Nedlands 6009, Australia
| | - Thomas Clairfeuille
- Institute for Molecular Bioscience, University of Queensland, St. Lucia 4072, Australia
| | - Euphemie Landao-Bassonga
- Cellular Orthopaedic Laboratory, School of Surgery, University of Western Australia, Nedlands 6009, Australia
| | - Genevieve Kinna
- Institute for Molecular Bioscience, University of Queensland, St. Lucia 4072, Australia
| | - Pei Ying Ng
- Cellular Orthopaedic Laboratory, School of Surgery, University of Western Australia, Nedlands 6009, Australia
| | - Li Shen Loo
- Institute of Molecular and Cell Biology, A*STAR, Singapore 138673
| | - Tak Sum Cheng
- Cellular Orthopaedic Laboratory, School of Surgery, University of Western Australia, Nedlands 6009, Australia
| | - Minghao Zheng
- Cellular Orthopaedic Laboratory, School of Surgery, University of Western Australia, Nedlands 6009, Australia
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, A*STAR, Singapore 138673
| | - Rohan D Teasdale
- Institute for Molecular Bioscience, University of Queensland, St. Lucia 4072, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, University of Queensland, St. Lucia 4072, Australia
| | - Nathan J Pavlos
- Cellular Orthopaedic Laboratory, School of Surgery, University of Western Australia, Nedlands 6009, Australia
| |
Collapse
|
35
|
Cheloha RW, Gellman SH, Vilardaga JP, Gardella TJ. PTH receptor-1 signalling-mechanistic insights and therapeutic prospects. Nat Rev Endocrinol 2015; 11:712-24. [PMID: 26303600 PMCID: PMC4651712 DOI: 10.1038/nrendo.2015.139] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Parathyroid hormone/parathyroid hormone-related protein receptor (PTH/PTHrP type 1 receptor; commonly known as PTHR1) is a family B G-protein-coupled receptor (GPCR) that regulates skeletal development, bone turnover and mineral ion homeostasis. PTHR1 transduces stimuli from PTH and PTHrP into the interior of target cells to promote diverse biochemical responses. Evaluation of the signalling properties of structurally modified PTHR1 ligands has helped to elucidate determinants of receptor function and mechanisms of downstream cellular and physiological responses. Analysis of PTHR1 responses induced by structurally modified ligands suggests that PTHR1 can continue to signal through a G-protein-mediated pathway within endosomes. Such findings challenge the longstanding paradigm in GPCR biology that the receptor is transiently activated at the cell membrane, followed by rapid deactivation and receptor internalization. Evaluation of structurally modified PTHR1 ligands has further led to the identification of ligand analogues that differ from PTH or PTHrP in the type, strength and duration of responses induced at the receptor, cellular and organism levels. These modified ligands, and the biochemical principles revealed through their use, might facilitate an improved understanding of PTHR1 function in vivo and enable the treatment of disorders resulting from defects in PTHR1 signalling. This Review discusses current understanding of PTHR1 modes of action and how these findings might be applied in future therapeutic agents.
Collapse
Affiliation(s)
- Ross W Cheloha
- Department of Chemistry, 1101 University Avenue, University of Wisconsin, Madison, WI 53706, USA
| | - Samuel H Gellman
- Department of Chemistry, 1101 University Avenue, University of Wisconsin, Madison, WI 53706, USA
| | - Jean-Pierre Vilardaga
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital, 50 Blossom Street, Boston, MA 02114, USA
| |
Collapse
|
36
|
Gardella TJ, Vilardaga JP. International Union of Basic and Clinical Pharmacology. XCIII. The parathyroid hormone receptors--family B G protein-coupled receptors. Pharmacol Rev 2015; 67:310-37. [PMID: 25713287 DOI: 10.1124/pr.114.009464] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The type-1 parathyroid hormone receptor (PTHR1) is a family B G protein-coupled receptor (GPCR) that mediates the actions of two polypeptide ligands; parathyroid hormone (PTH), an endocrine hormone that regulates the levels of calcium and inorganic phosphate in the blood by acting on bone and kidney, and PTH-related protein (PTHrP), a paracrine-factor that regulates cell differentiation and proliferation programs in developing bone and other tissues. The type-2 parathyroid hormone receptor (PTHR2) binds a peptide ligand, called tuberoinfundibular peptide-39 (TIP39), and while the biologic role of the PTHR2/TIP39 system is not as defined as that of the PTHR1, it likely plays a role in the central nervous system as well as in spermatogenesis. Mechanisms of action at these receptors have been explored through a variety of pharmacological and biochemical approaches, and the data obtained support a basic "two-site" mode of ligand binding now thought to be used by each of the family B peptide hormone GPCRs. Recent crystallographic studies on the family B GPCRs are providing new insights that help to further refine the specifics of the overall receptor architecture and modes of ligand docking. One intriguing pharmacological finding for the PTHR1 is that it can form surprisingly stable complexes with certain PTH/PTHrP ligand analogs and thereby mediate markedly prolonged cell signaling responses that persist even when the bulk of the complexes are found in internalized vesicles. The PTHR1 thus appears to be able to activate the Gα(s)/cAMP pathway not only from the plasma membrane but also from the endosomal domain. The cumulative findings could have an impact on efforts to develop new drug therapies for the PTH receptors.
Collapse
Affiliation(s)
- Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts (T.J.G.); and Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (J.-P.V.)
| | - Jean-Pierre Vilardaga
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts (T.J.G.); and Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (J.-P.V.)
| |
Collapse
|
37
|
Qiu T, Xian L, Crane J, Wen C, Hilton M, Lu W, Newman P, Cao X. PTH receptor signaling in osteoblasts regulates endochondral vascularization in maintenance of postnatal growth plate. J Bone Miner Res 2015; 30:309-17. [PMID: 25196529 PMCID: PMC4730385 DOI: 10.1002/jbmr.2327] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 07/22/2014] [Accepted: 07/25/2014] [Indexed: 11/09/2022]
Abstract
Longitudinal growth of postnatal bone requires precise control of growth plate cartilage chondrocytes and subsequent osteogenesis and bone formation. Little is known about the role of angiogenesis and bone remodeling in maintenance of cartilaginous growth plate. Parathyroid hormone (PTH) stimulates bone remodeling by activating PTH receptor (PTH1R). Mice with conditional deletion of PTH1R in osteoblasts showed disrupted trabecular bone formation. The mice also exhibited postnatal growth retardation with profound defects in growth plate cartilage, ascribable predominantly to a decrease in number of hypertrophic chondrocytes, resulting in premature fusion of the growth plate and shortened long bones. Further characterization of hypertrophic zone and primary spongiosa revealed that endochondral angiogenesis and vascular invasion of the cartilage were impaired, which was associated with aberrant chondrocyte maturation and cartilage development. These studies reveal that PTH1R signaling in osteoblasts regulates cartilaginous growth plate for postnatal growth of bone.
Collapse
Affiliation(s)
- Tao Qiu
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lingling Xian
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janet Crane
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chunyi Wen
- Department of Orthopaedics and Traumatology, University of Hong Kong, Hong Kong, China
| | - Matthew Hilton
- Department of Orthopaedics and Rehabilitation, University of Rochester School of Medicine, Rochester, NY, USA
| | - William Lu
- Department of Orthopaedics and Traumatology, University of Hong Kong, Hong Kong, China
| | - Peter Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, USA
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Singh R, Ahalawat N, Murarka RK. Activation of corticotropin-releasing factor 1 receptor: insights from molecular dynamics simulations. J Phys Chem B 2015; 119:2806-17. [PMID: 25607803 DOI: 10.1021/jp509814n] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
G-protein-coupled receptors (GPCRs) constitute the largest family of membrane-bound proteins involved in translation of extracellular signals into intracellular responses. They regulate diverse physiological and pathophysiological processes, and hence, they are prime drug targets for therapeutic intervention. In spite of the recent advancements in membrane protein crystallography, limited information is available on the molecular signatures of activation of GPCRs. Although few studies have been reported for class A GPCRs, the activation mechanism of class B GPCRs remains unexplored. Corticotropin-releasing factor 1 receptor (CRF1R), a class B GPCR, is associated with various disease conditions including stress, anxiety, and irritable bowel syndrome. Here, we report the activation of CRF1R using accelerated molecular dynamics simulations of the apo receptor. The breakage of His155(2.50)-Glu209(3.50) and Glu209(3.50)-Thr316(6.42) interactions is found to be crucial in transition of the receptor to its active conformation. Compared to the inactive crystal structure, major structural rearrangements occurred in the intracellular region of the transmembrane (TM) domain upon activation: TM3 twisted away from TM2, and an opening of the G-protein binding site occurred as a result of the outward movements of TM5 and TM6 from the helical bundle. Further, an inward tilt of TM7 toward the helical core is observed at the extracellular side, in agreement with recent findings (Coin et al. Cell 2013, 155, 1258-1269), where it is proposed that this movement helps in establishing favorable interactions with peptide agonist. Moreover, different allosteric pathways in the inactive and active states are identified using the correlations in torsion angle space. The inactive state is found to be less dynamic as compared to the putative active state of the receptor. Results from the current study could present a model for class B GPCRs activation and aid in the design of CRF1R modulators against brain and metabolic disorders.
Collapse
Affiliation(s)
- Rajesh Singh
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal , Indore By-pass Road, Bhauri, Bhopal 462066, MP, India
| | | | | |
Collapse
|
39
|
Carter PH, Dean T, Bhayana B, Khatri A, Rajur R, Gardella TJ. Actions of the small molecule ligands SW106 and AH-3960 on the type-1 parathyroid hormone receptor. Mol Endocrinol 2015; 29:307-21. [PMID: 25584411 DOI: 10.1210/me.2014-1129] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The parathyroid hormone receptor-1 (PTHR1) plays critical roles in regulating blood calcium levels and bone metabolism and is thus of interest for small-molecule ligand development. Of the few small-molecule ligands reported for the PTHR1, most are of low affinity, and none has a well-defined mechanism of action. Here, we show that SW106 and AH-3960, compounds previously identified to act as an antagonist and agonist, respectively, on the PTHR1, each bind to PTHR1-delNT, a PTHR1 construct that lacks the large amino-terminal extracellular domain used for binding endogenous PTH peptide ligands, with the same micromolar affinity with which it binds to the intact PTHR1. SW106 antagonized PTHR1-mediated cAMP signaling induced by the peptide analog, M-PTH(1-11), as well as by the native PTH(1-9) sequence, as tethered to the extracellular end of transmembrane domain (TMD) helix-1 of the receptor. SW106, however, did not function as an inverse agonist on either PTHR1-H223R or PTHR1-T410P, which have activating mutations at the cytoplasmic ends of TMD helices 2 and 6, respectively. The overall data indicate that SW106 and AH-3960 each bind to the PTHR1 TMD region and likely to within an extracellularly exposed area that is occupied by the N-terminal residues of PTH peptides. Additionally, they suggest that the inhibitory effects of SW106 are limited to the extracellular portions of the TMD region that mediate interactions with agonist ligands but do not extend to receptor-activation determinants situated more deeply in the helical bundle. The study helps to elucidate potential mechanisms of small-molecule binding at the PTHR1.
Collapse
Affiliation(s)
- Percy H Carter
- Endocrine Unit (T.D., A.K., T.J.G.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02492; Department of Photomedicine (B.B.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02492; CreaGen Biosciences, Inc (R.R.), Woburn, Massachusetts 01801; and Bristol-Myers Squibb Co (P.H.C.), Princeton, New Jersey 08543
| | | | | | | | | | | |
Collapse
|
40
|
Meng QH, Wagar EA. Laboratory approaches for the diagnosis and assessment of hypercalcemia. Crit Rev Clin Lab Sci 2014; 52:107-19. [DOI: 10.3109/10408363.2014.970266] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Li D, Opas EE, Tuluc F, Metzger DL, Hou C, Hakonarson H, Levine MA. Autosomal dominant hypoparathyroidism caused by germline mutation in GNA11: phenotypic and molecular characterization. J Clin Endocrinol Metab 2014; 99:E1774-83. [PMID: 24823460 PMCID: PMC4154081 DOI: 10.1210/jc.2014-1029] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CONTEXT Most cases of autosomal dominant hypoparathyroidism (ADH) are caused by gain-of-function mutations in CASR or dominant inhibitor mutations in GCM2 or PTH. OBJECTIVE Our objectives were to identify the genetic basis for ADH in a multigenerational family and define the underlying disease mechanism. SUBJECTS Here we evaluated a multigenerational family with ADH in which affected subjects had normal sequences in these genes and were shorter than unaffected family members. METHODS We collected clinical and biochemical data from 6 of 11 affected subjects and performed whole-exome sequence analysis on DNA from two affected sisters and their affected father. Functional studies were performed after expression of wild-type and mutant Gα11 proteins in human embryonic kidney-293-CaR cells that stably express calcium-sensing receptors. RESULTS Whole-exome-sequencing followed by Sanger sequencing revealed a heterozygous mutation, c.179G>T; p.R60L, in GNA11, which encodes the α-subunit of G11, the principal heterotrimeric G protein that couples calcium-sensing receptors to signal activation in parathyroid cells. Functional studies of Gα11 R60L showed increased accumulation of intracellular concentration of free calcium in response to extracellular concentration of free calcium with a significantly decreased EC50 compared with wild-type Gα11. By contrast, R60L was significantly less effective than the oncogenic Q209L form of Gα11 as an activator of the MAPK pathway. Compared to subjects with CASR mutations, patients with GNA11 mutations lacked hypercalciuria and had normal serum magnesium levels. CONCLUSIONS Our findings indicate that the germline gain-of-function mutation of GNA11 is a cause of ADH and implicate a novel role for GNA11 in skeletal growth.
Collapse
Affiliation(s)
- Dong Li
- Center for Applied Genomics (D.L., C.H., H.H.), Division of Endocrinology and Diabetes (E.E.O., M.A.L.), Division of Allergy and Immunology (F.T.), Division of Pulmonary Medicine (H.H.), and Center for Bone Health (M.A.L.), The Children's Hospital of Philadelphia; and Department of Pediatrics (H.H., M.A.L.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104; and Endocrinology and Diabetes Unit (D.L.M.), British Columbia Children's Hospital, and Department of Pediatrics (D.L.M.), University of British Columbia, Vancouver, British Columbia, Canada V5Z 4H4
| | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Gattineni J, Alphonse P, Zhang Q, Mathews N, Bates CM, Baum M. Regulation of renal phosphate transport by FGF23 is mediated by FGFR1 and FGFR4. Am J Physiol Renal Physiol 2014; 306:F351-8. [PMID: 24259513 PMCID: PMC3920047 DOI: 10.1152/ajprenal.00232.2013] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 11/19/2013] [Indexed: 12/30/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) is a bone-derived hormone that acts on the proximal tubule to decrease phosphate reabsorption and serum levels of 1,25-dihydroxyvitamin D₃ [1,25(OH)₂ Vitamin D₃]. Abnormal FGF23 metabolism has been implicated in several debilitating hypophosphatemic and hyperphosphatemic disorders. The renal receptors responsible for the phosphaturic actions of FGF23 have not been elucidated. There are four fibroblast growth factor receptors (FGFR); 1-4 with "b" and "c" isoforms for receptors 1, 2, and 3. FGFR1, 3, and 4 are expressed in the mouse proximal tubule, and deletion of any one receptor did not affect serum phosphate levels, suggesting that more than one receptor is involved in mediating the phosphaturic actions of FGF23. To determine the receptors responsible for the phosphaturic actions of FGF23, we studied Fgfr1 (kidney conditional) and Fgfr4 (global) double mutant mice (Fgfr1⁻/⁻/Fgfr4⁻/⁻). Fgfr1⁻/⁻/Fgfr4⁻/⁻ mice have higher FGF23 levels than their wild-type counterparts (108.1 ± 7.3 vs. 4,953.6 ± 675.0 pg/ml; P < 0.001). Despite the elevated FGF23 levels, Fgfr1⁻/⁻/Fgfr4⁻/⁻ mice have elevated serum phosphorus levels, increased brush-border membrane vesicle (BBMV) phosphate transport, and increased Na-P(i) cotransporter 2c (NaPi-2c) protein expression compared with wild-type mice. These data are consistent with FGFR1 and FGFR4 being the critical receptors for the phosphaturic actions of FGF23.
Collapse
Affiliation(s)
- Jyothsna Gattineni
- Dept. of Pediatrics, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX.
| | | | | | | | | | | |
Collapse
|
44
|
Frazier-Bowers SA, Hendricks HM, Wright JT, Lee J, Long K, Dibble CF, Bencharit S. Novel mutations in PTH1R associated with primary failure of eruption and osteoarthritis. J Dent Res 2013; 93:134-9. [PMID: 24300310 DOI: 10.1177/0022034513513588] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Autosomal dominant mutations in PTH1R segregate with primary failure of eruption (PFE), marked by clinical eruption failure of adult teeth without mechanical obstruction. While the diagnosis of PFE conveys a poor dental prognosis, there are no reports of PFE patients who carry PTH1R mutations and exhibit any other skeletal problems. We performed polymerase chain reaction-based mutational analysis of the PTH1R gene to determine the genetic contribution of PTH1R in 10 families with PFE. Sequence analysis of the coding regions and intron-exon boundaries of the PTH1R gene in 10 families (n = 54) and 7 isolated individuals revealed 2 novel autosomal dominant mutations in PTH1R (c.996_997insC and C.572delA) that occur in the coding region and result in a truncated protein. One family showed incomplete penetrance. Of 10 families diagnosed with PFE, 8 did not reveal functional (nonsynonymous) mutations in PTH1R; furthermore, 4 families and 1 sporadic case carried synonymous single-nucleotide polymorphisms. Five PFE patients in 2 families carried PTH1R mutations and presented with osteoarthritis. We propose that the autosomal dominant mutations of PTH1R that cause PFE may also be associated with osteoarthritis; a dose-dependent model may explain isolated PFE and osteoarthritis in the absence of other known symptoms in the skeletal system.
Collapse
Affiliation(s)
- S A Frazier-Bowers
- Department of Orthodontics, School of Dentistry, University of North Carolina at Chapel Hill
| | | | | | | | | | | | | |
Collapse
|
45
|
Savoldi G, Izzi C, Signorelli M, Bondioni MP, Romani C, Lanzi G, Moratto D, Verdoni L, Pinotti M, Prefumo F, Superti-Furga A, Pilotta A. Prenatal presentation and postnatal evolution of a patient with Jansen metaphyseal dysplasia with a novel missense mutation in PTH1R. Am J Med Genet A 2013; 161A:2614-9. [PMID: 23950054 DOI: 10.1002/ajmg.a.36115] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 05/30/2013] [Indexed: 11/10/2022]
Abstract
Wave-shaped ribs were detected at prenatal ultrasound in a 20(+1) week female fetus. At birth, skeletal radiographs showed marked hypomineralization and suggested hypophosphatasia. However, elevated blood calcium and alkaline phosphatase excluded hypophosphatasia and raised the possibility of Jansen metaphyseal dysplasia. Molecular analysis of the PTH/PTHrP receptor gene (PTH1R) showed heterozygosity for a previously undescribed transversion variant (c.1373T>A), which predicts p.Ile458Lys. In vitro evaluation of wild type and mutant PTH/PTHrP receptors supported the pathogenic role of the p.Ile458Lys substitution, and confirmed the diagnosis of Jansen metaphyseal dysplasia. This disorder may present prenatally with wavy ribs and in the newborn with hypomineralization, and may therefore be confused with hypophosphatasia. The mottled metaphyseal lesions typically associated with this disease appear only in childhood.
Collapse
Affiliation(s)
- Gianfranco Savoldi
- Laboratory of Genetic Disorders of Childhood, A. Nocivelli Institute for Molecular Medicine, Department of Pathology, Spedali Civili, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Koole C, Savage EE, Christopoulos A, Miller LJ, Sexton PM, Wootten D. Minireview: Signal bias, allosterism, and polymorphic variation at the GLP-1R: implications for drug discovery. Mol Endocrinol 2013; 27:1234-44. [PMID: 23864649 DOI: 10.1210/me.2013-1116] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) controls the physiological responses to the incretin hormone glucagon-like peptide-1 and is a major therapeutic target for the treatment of type 2 diabetes, owing to the broad range of effects that are mediated upon its activation. These include the promotion of glucose-dependent insulin secretion, increased insulin biosynthesis, preservation of β-cell mass, improved peripheral insulin action, and promotion of weight loss. Regulation of GLP-1R function is complex, with multiple endogenous and exogenous peptides that interact with the receptor that result in the activation of numerous downstream signaling cascades. The current understanding of GLP-1R signaling and regulation is limited, with the desired spectrum of signaling required for the ideal therapeutic outcome still to be determined. In addition, there are several single-nucleotide polymorphisms (used in this review as defining a natural change of single nucleotide in the receptor sequence; clinically, this is viewed as a single-nucleotide polymorphism only if the frequency of the mutation occurs in 1% or more of the population) distributed within the coding sequence of the receptor protein that have the potential to produce differential responses for distinct ligands. In this review, we discuss the current understanding of GLP-1R function, in particular highlighting recent advances in the field on ligand-directed signal bias, allosteric modulation, and probe dependence and the implications of these behaviors for drug discovery and development.
Collapse
Affiliation(s)
- Cassandra Koole
- Department of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
47
|
Polar transmembrane interactions drive formation of ligand-specific and signal pathway-biased family B G protein-coupled receptor conformations. Proc Natl Acad Sci U S A 2013; 110:5211-6. [PMID: 23479653 DOI: 10.1073/pnas.1221585110] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Recently, the concept of ligand-directed signaling--the ability of different ligands of an individual receptor to promote distinct patterns of cellular response--has gained much traction in the field of drug discovery, with the potential to sculpt biological response to favor therapeutically beneficial signaling pathways over those leading to harmful effects. However, there is limited understanding of the mechanistic basis underlying biased signaling. The glucagon-like peptide-1 receptor is a major target for treatment of type-2 diabetes and is subject to ligand-directed signaling. Here, we demonstrate the importance of polar transmembrane residues conserved within family B G protein-coupled receptors, not only for protein folding and expression, but also in controlling activation transition, ligand-biased, and pathway-biased signaling. Distinct clusters of polar residues were important for receptor activation and signal preference, globally changing the profile of receptor response to distinct peptide ligands, including endogenous ligands glucagon-like peptide-1, oxyntomodulin, and the clinically used mimetic exendin-4.
Collapse
|
48
|
Abstract
Much of the mammalian skeleton is composed of bones that originate from cartilage templates through endochondral ossification. Elucidating the mechanisms that control endochondral bone development is critical for understanding human skeletal diseases, injury response, and aging. Mouse genetic studies in the past 15 years have provided unprecedented insights about molecules regulating chondrocyte formation, chondrocyte maturation, and osteoblast differentiation, all key processes of endochondral bone development. These include the roles of the secreted proteins IHH, PTHrP, BMPs, WNTs, and FGFs, their receptors, and transcription factors such as SOX9, RUNX2, and OSX, in regulating chondrocyte and osteoblast biology. This review aims to integrate the known functions of extracellular signals and transcription factors that regulate development of the endochondral skeleton.
Collapse
Affiliation(s)
- Fanxin Long
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | |
Collapse
|
49
|
Abstract
This is the continuation of a two-part review of rickets. This part emphasizes the specific pathophysiology, clinical features, pathoanatomy and radiographic findings of vitamin D deficiency rickets. Other forms of rickets, differential diagnostic considerations and the potential relationship between low levels of vitamin D metabolites and unexplained fractures in infants are also discussed.
Collapse
|
50
|
Vohra S, Taddese B, Conner AC, Poyner DR, Hay DL, Barwell J, Reeves PJ, Upton GJG, Reynolds CA. Similarity between class A and class B G-protein-coupled receptors exemplified through calcitonin gene-related peptide receptor modelling and mutagenesis studies. J R Soc Interface 2012; 10:20120846. [PMID: 23235263 PMCID: PMC3565703 DOI: 10.1098/rsif.2012.0846] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Modelling class B G-protein-coupled receptors (GPCRs) using class A GPCR structural templates is difficult due to lack of homology. The plant GPCR, GCR1, has homology to both class A and class B GPCRs. We have used this to generate a class A–class B alignment, and by incorporating maximum lagged correlation of entropy and hydrophobicity into a consensus score, we have been able to align receptor transmembrane regions. We have applied this analysis to generate active and inactive homology models of the class B calcitonin gene-related peptide (CGRP) receptor, and have supported it with site-directed mutagenesis data using 122 CGRP receptor residues and 144 published mutagenesis results on other class B GPCRs. The variation of sequence variability with structure, the analysis of polarity violations, the alignment of group-conserved residues and the mutagenesis results at 27 key positions were particularly informative in distinguishing between the proposed and plausible alternative alignments. Furthermore, we have been able to associate the key molecular features of the class B GPCR signalling machinery with their class A counterparts for the first time. These include the [K/R]KLH motif in intracellular loop 1, [I/L]xxxL and KxxK at the intracellular end of TM5 and TM6, the NPXXY/VAVLY motif on TM7 and small group-conserved residues in TM1, TM2, TM3 and TM7. The equivalent of the class A DRY motif is proposed to involve Arg2.39, His2.43 and Glu3.46, which makes a polar lock with T6.37. These alignments and models provide useful tools for understanding class B GPCR function.
Collapse
Affiliation(s)
- Shabana Vohra
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, UK
| | | | | | | | | | | | | | | | | |
Collapse
|